WO2019186344A1 - Anti-cancer and disintegrin scorpion venoms - Google Patents

Anti-cancer and disintegrin scorpion venoms Download PDF

Info

Publication number
WO2019186344A1
WO2019186344A1 PCT/IB2019/052358 IB2019052358W WO2019186344A1 WO 2019186344 A1 WO2019186344 A1 WO 2019186344A1 IB 2019052358 W IB2019052358 W IB 2019052358W WO 2019186344 A1 WO2019186344 A1 WO 2019186344A1
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
cells
cancer
cell
composition
Prior art date
Application number
PCT/IB2019/052358
Other languages
French (fr)
Inventor
Riadh KHARRAT
Oussema KHAMESSI
Original Assignee
Institut Pasteur De Tunis
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institut Pasteur De Tunis filed Critical Institut Pasteur De Tunis
Publication of WO2019186344A1 publication Critical patent/WO2019186344A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids

Definitions

  • the invention is related to novel scorpion venoms and methods of using the same to inhibit cell migration, cell proliferation, and angiogenesis, as well as to treat cancer, pathological angiogenesis, and integrin-mediated diseases.
  • Cancer cells share characteristics that make it possible to identify them, despite the great diversity of cancers. They are a genetically unstable type cell, able to explore functions of the entire genome and to use any migration or proliferative advantage to select and transmit it to its offspring.
  • the tumor masses oscillate between proliferation and apoptosis.
  • the development of new blood vessels in the tumor also facilitates metastasis formation, Cell migration and proliferation in response to pro-angiogenic factors.
  • Cell migration, proliferation and angiogenesis are key phenomena in tumor growth. They give rise to numerous cell interactions with each other and with their environment, leading to non-trivial collective effects.
  • Angiogenesis is required to maintain the continuous growth of malignant cells by providing with nutrients and oxygen and allow them to discard the wastes.
  • the ability of tumor cells to induce the neovascularization is a significant step in tumorigenesis.
  • the vast majority of tumor cells must initiate tumor angiogenesis by producing proangiogenic diffusion-promoting tumor substances to form detectable tumors. For that, blocking the action of proangiogenic substances can stop tumor growth.
  • Cubans have been treating cancer patients with blue scorpion venom. The results are not always so great, but thousands of patients say their pain has been relieved, their muscle strength is increased, and their energy is boosted by taking this medication. In fact, many of the scorpion venom peptides are active on different tumoral metastasis stages.
  • Charybdotoxin (ChTx) and Iberiotoxin are capable to inhibit the proliferation and migration of glioma and melanoma cells, respectively [1 , 2]
  • the few peptides from scorpion venom that have an anti-tumor effect are generally cytotoxic.
  • Integrins are transmembrane heterodimeric proteins of non-covalently associated a and b subunits, are implicated in all the processes of carcinogenesis [3] The association between their a and b subunits forms adhesion receptors which binds to the extracellular matrix and provide critical adhesive and signalling functions [4] Integrins are capable to affect cellular functions [5] such as cytoskeleton organization, transduction of intracellular signals [6], cell differentiation, growth, and apoptosis [7-8] Different subtypes of integrins have been characterized on the basis of their interactions with different motifs of the extracellular matrix implicated in cell adhesion [9] These motifs coordinate the biological responses between endothelial cells, tumor cells and the extracellular matrix [10] In fact, these integrins were first identified on the basis of their ability to recognize the RGD amino acid sequence [11-12], and are implicated in cell proliferation, invasion and viability.
  • Integrins bind to collagen by using their al Domain, a1 b1 and a2b1 represent the most known collagen receptors which are members of integrin family and are structurally very similar [14] Their ligand binds to a Mg2+ ion in the Metal Ion-Dependent Adhesion Site (MIDAS) [15] a1 b1 integrin is the principal collagen IV receptor [16] but its contribution in tumor formation and progression is poorly defined compared to anb3 and a5b1. The critical role of a1 b1 integrin in tumorigenicity was demonstrated with works elaborated in colon cancer cells that associate with talin and paxillin, resulting in promotion of cancer cell invasion [17]
  • MIDAS Metal Ion-Dependent Adhesion Site
  • disintegrins The ability of multiple polypeptides to inhibit the integrin activity was previously discovered for different molecules purified mainly from snake venoms called disintegrins. Disintegrins were proven to have antitumour effects involving angiogenesis and cancer metastatic dissemination. The functional classification of disintegrins depends on their ability to interact with specific integrins [18], which is determined by the presence of a particular integrin-binding motif localized in the hairpin loop, unless they are present in the same fold. Functionally, disintegrins can be divided into three classes containing RGD, MLD, and R/KTS motifs have been identified [19] RGD-disintegrins block anb3, anb1 , a5b1 and a8b1 integrins.
  • R/KTS-disintegrins are a potent and selective inhibitors of a1 b1 integrin [21]
  • Scorpion peptides are well known for their pharmaceutical potential on different targets. These targets are mainly ion channels, which were found to be highly expressed in many diseases including cancer, auto-immune pathologies and Alzheimer. However, to date, disintegrin activity has only been found in snake venom molecules, not scorpion. Scorpion toxins have been the subject of many studies which explore their pharmacological potential toward diverse molecular targets, known to monitor key mechanisms in cancer such as proliferation, migration and angiogenesis. The few peptides from scorpion venom that have an anti-tumor effect are generally cytotoxic.
  • Scorpion peptides are disulfide bond-rich molecules presenting a sequence length ranging between 23 and about 80 amino acids. The majority of scorpion peptides have the sequence signature of the cystine stabilized a/b (CSa/b) motif [25] Some scorpion CSa/b peptides display a remarkable specificity for certain subtypes of ion channels [26] Scorpion venom contains also a significant number of peptides, without disulfide bridge, that exhibit antimicrobial, immunomodulatory, Bradykinin-potentiating and/or hemolytic activities [27]
  • the inventors present the first description of a first short 14 amino acid peptide (called RK1), purified from the venom of Buthus occitanus tunetanus, with the particular capabilities, among different other scorpion peptides, to inhibit cell proliferation, migration and angiogenesis of U87 (Glioblastoma) and IGR39 (Melanoma).
  • RK1 is a first peptide derived from scorpion venom exhibiting a potential anti-tumoral activity with no manifest toxicity.
  • Our results suggest that, in terms of its primary structure, RK1 is unique compared to a variety of known peptides purified from scorpion venoms.
  • RK1 is the first natural peptide able to abolish completely the proliferation of cancer cells.
  • the chicken chorioallantoic membrane model revealed that RK1 strongly inhibits ex-vivo vascular growth.
  • RK1 is a small peptide of 14 amino acids containing a single disulfide bond.
  • RK1 may represent the first member of a new group of scorpion peptides and can be classified as one of the first anti-cancer scorpion venom peptides that do not have cytotoxic activity.
  • RK1 could open new perspectives for the pharmaceutical application of short scorpion venom peptides in anticancer activity and may represent the first member of a new group of scorpion peptides.
  • RK Rostanus peptide
  • the inventors isolated and characterized RK, from the Buthus occitanus tunetanus scorpion venom targeting the cell adhesion activity by acting on integrins. To our knowledge, this is the first disintegrin-like peptide purified from a scorpion venom.
  • RK peptide is unique among the variety of known peptides purified from scorpion venoms in terms of its structure. It is a small peptide of 17 amino acids (IDCGTVMIPSECDPKSS; SEQ ID NO:1) containing a single disulfide bond.
  • RK may represent the first member of a new group of scorpion peptides.
  • the molecular docking study shows that RK involves mainly two segments in its structure to interact with the aibi integrin. M 7 of RK peptide seems to play an important role due to its ability to occupy a small hydrophobic pocket on the surface of the integrin. h and D 2 are capable to interact with the C-loop of the receptor, while K15 establishes a salt bridge interaction with its E 28 s.
  • the molecular modeling study suggests the key contribution of the E H C I 2 D I 3 segment in the interaction with a n b3 integrin. Our results highlight a new class of disintegrin molecules which opens a interesting perspectives for the pharmaceutical application of short scorpion venom peptides in cancer treatment.
  • the present invention is directed to novel scorpion peptides with various biological activities and their uses in the treatment of various disorders.
  • An isolated peptide selected from the group consisting of peptides whose amino acid sequence comprises or consists of:
  • IDCSKVNLTAECSS (peptide RK1 ; SEQ ID NO:2);
  • IDCGTVMIPSECDPKS (peptide RK; SEQ ID NO:3);
  • a sequence has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 91 %, at least about 92%, at least about 93%, at least about 94%, at last about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity to RK1 ;
  • a sequence has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 91 %, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity to RK.
  • the peptide according to embodiment 1 comprising at least one substitution modification, insertion, or deletion modification relative to IDCSKVNLTAECSS (SEQ ID NO:2) and IDCGTVMIPSECDPKSS (SEQ ID NO: 1).
  • DCSKXXXXXECDS (SEQ ID NO:5) of the RK1 peptide.
  • polyalkylethers e.g. polyethylene glycol and polypropylene glycol
  • polylactic acid polyglycolic acid
  • polyoxyalkenes polyvinylalcohol
  • polyvinylpyrrolidone cellulose and cellulose derivatives
  • dextran and dextran derivatives
  • polyvinyl pyrrolidones glycopeptides, and polyamino acids.
  • peptide according to any one of embodiments 1-19, wherein the peptide is modified by the addition of cysteine or biotin.
  • peptide according to any one of embodiments 1-20, wherein the peptide is modified by a moiety to facilitate crosslinking.
  • peptide according to any one of embodiments 1-21 , wherein the moiety is chosen from benzophenone, maleimide, and activated esters.
  • the peptide according to any one of embodiments 1-22 wherein the peptide is not able to alter cell adhesion through a2b1 , anbq, anb5, a5b1 and a6b4 integrins, but significantly reduces the adhesive activity of a1 b1 and anb3 integrins to extracellular matrix proteins.
  • the extracellular matrix protein comprises vitronectin, fibronectin, fibrinogen, osteopoietin, and/or bone sialoprotein.
  • peptide according to any one of embodiments 1-25, wherein the peptide competes with a synthetic RGD peptide for binding to aibi and/or a n b3 integrins
  • composition comprising the peptide according to any one of embodiments 1-26 and a pharmaceutically acceptable carrier, diluent, or excipient.
  • composition of embodiment 27, wherein the composition is a sustained release formulation or is in a sustained release carrier.
  • composition according to any one of embodiments 27-28, wherein the composition is formulated to be administered in microspheres, liposomes, or one of other microparticulate delivery systems.
  • composition according to any one of embodiment 27-30 wherein the semipermeable polymer matrix comprises one or more of polylactides copolymers of L-glutamic acid and gamma ethyl-L-glutamate, poly (2-hydroxyethyl-methacrylate), or ethylene vinyl acetate.
  • a method of treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31.
  • the method according to any one of embodiments 32-25 or 45-60, wherein the dosage of the peptide to be achieved in vivo may be equivalent to an in vitro level of 1 mM, 2 pM, 3 pM, 4 pM, 5 pM, 6 pM, 7 pM, 1 pM, 8 pM, 9 pM, 10 pM, 20 pM, 30 pM, 40 pM, 50 pM, 70 pM, 80 pM, 90 pM, or 100 pM.
  • the dosage of the peptide to be achieved in vivo may be a blood/plasma level of at least 1 pM, 2 pM, 3 pM, 4 pM, 5 pM, 6 pM, 7 pM, 1 pM, 8 pM, 9 pM, 10 pM, 20 pM, 30 pM, 40 pM, 50 pM, 70 pM, 80 pM, 90 pM, or 100 pM.
  • composition is administered orally, parenterally, topically, by inhalation, intranasally, or recta I ly.
  • composition is administered intravenously, oral, sublingual, intranasal, intraocular, rectal, transdermal, mucosal, topical or parenteral.
  • composition comprises at least one additive chosen from a pharmaceutically acceptable excipients, carriers, preservatives, buffers, stabilizers, antioxidants, or other additives.
  • composition is in a form chosen from a tablet, capsule, powder, or liquid.
  • the liquid comprises at least one additive chosen from liquid carriers, petroleum, animal oils, vegetable oils, mineral oils, synthetic oils, physiological saline solutions, saccharide solutions, and glycols.
  • the cancer is selected from glioblastoma, melanoma, leukemia, and hepatocellular cancers, sarcoma, vascular endothelial cancers, breast cancers, central nervous system cancers (e.g.
  • astrocytoma astrocytoma, gliosarcoma, neuroblastoma, oligodendroglioma and glioblastoma
  • prostate cancers lung and bronchus cancers, larynx cancers, esophagus cancers, colon cancers, colorectal cancers, gastro-intestinal cancers, melanomas, ovarian and endometrial cancer, renal and bladder cancer, liver cancer, endocrine cancer (e.g. thyroid), and pancreatic cancer.
  • a method of inhibiting angiogenesis in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31.
  • a method for treatment of an integrin-associated disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31.
  • the integrin-associated disease is osteoporosis, bone tumor or cancer growth, angiogenesis-related tumor growth and metastasis, tumor metastasis in bone, malignancy-induced hypercalcemia, angiogenesis-related eye diseases, Paget's disease, rheumatic arthritis, ovariectomy-induced physiological change, inflammation, coagulation diseases, or osteoarthritis.
  • said therapeutic agent is selected from the group consisting of an antiangiogenic agent, a cytotoxic agent, a cytostatic agent, and an immunomodulatory agent.
  • the at least one an antiangiogenic agent, a cytotoxic agent, a cytostatic agent comprises at least one agent chosen from endostatin, angiostatin, VEGF inhibitors, cytotoxic agents, alkaloids, antimetabolites, cancer growth inhibitors, gene therapy therapeutics, cancer vaccines, interferons, monoclonal antibodies, radiotherapy, hormonal therapy, and other supportive therapy.
  • the therapeutic agent is selected from Alkylating Agents, DNA Alkylating-like Agents, Alkylating Antineoplastic Agents, DNA replication and repair inhibitors, Cell Cycle Modulators, Apoptosis Regulators, Angiogenesis Inhibitors, Proteasome Inhibitors, Kinase Inhibitors, Protein Synthesis Inhibitors, Histone deacetylase inhibitors, Topoisomerase I Inhibitors, Topoisomerase II Inihibitors, DNA Intercalating Agents, RNA/DNA Antimetabolites, DNA Antimetabolites, Mitochondria Inhibitors, Antimitotic Agents, Nuclear Export Inhibitors, and Hormonal Therapies.
  • therapeutically effective means that the administration of the composition to the subject results in any demonstrated clinical benefit compared with no therapy (when appropriate) or to a known standard of care.
  • a method of decreasing binding of aibi and/or a n b3 integrins expressing cells to extracellular molecules comprising contacting the cells with according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31.
  • a method of screening for inhibition of tumor cell growth comprising administering a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31 to a subject, and determining whether the peptide reduces tumor cell growth.
  • a method of inhibiting tumor cell proliferation comprising contacting the tumor cell with an effective amount of a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31.
  • a method of inhibiting tumor cell migration comprising contacting the tumor cell with an effective amount of a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31.
  • tumor cells are glioblastoma cells or melanoma cells.
  • a method of designing or screening for a disintegrin peptide or other integrin inhibitor comprises using the model described in the figures as a basis for the design or screening.
  • a peptide according to any one of embodiments 1-26 as a medicament is a medicament.
  • a peptide according to any one of embodiments 1-26 or a composition according to anyone of embodiments 27-31 for the preparation of a drug for the treatment of cancer, inhibiting tumor cell migration, inhibiting tumor cell proliferation, decreasing binding of aibi and/or a n b3 integrins expressing cells to extracellular molecules, treatment of an integrin-associated disease, and/or disorders mediated by/associated with angiogenesis.
  • a peptide according to any one of embodiments 1-26 or a composition according to anyone of embodiments 27-31 for the preparation of a drug for the treatment of epilepsy, memory, learning, neuropsychiatric, neurological, neuromuscular, and immunological disorders, schizophrenia, bipolar disorder, sleep apnea, neurodegeneration, smooth muscle disorders, bacterial diseases, fungal diseases, malaria, viral diseases, Immuno-modulator-responsive disorders or pain.
  • kits comprising a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31.
  • FIG. 1A and B Purification of RK1 from the venom Buthus occitanus tunetanus.
  • Fig. 2A and B Effect of RK1 on tumour cell viability and proliferation
  • FIG. 4A and B Chicken chorioallantoic membrane assay CAM
  • FIG. 5A-D Purification of RK from the venom Buthus occitanus tunetanus.
  • A Purification of BotG50 on semi preparative C8 reverse phase HPLC column. Fractions 19-22 min were collected.
  • B Separation of fraction 19-22 min on C18-RP-HPLC, fraction eluted at 33.5 min was collected.
  • C Final purification step to ensure the purity of peak eluting at 33.5 min (RK).
  • D Amino acid sequence of RK (SEQ ID NO:1).
  • Fig. 7A-C RK inhibits cell adhesion Glioblastoma cells U87 (A), Melanoma cells IGR39 (B) and Rat pheochromocytoma cells PC12 (C)
  • ECM proteins type IV collagen, fibrinogen (Fg), fibronectin (Fn), laminin I (Lnl)
  • Fg fibrinogen
  • Fn fibronectin
  • Lnl laminin I
  • FIG. 9A and B Effect of RK on various integrins in cell adhesion assays
  • Adhesion assays were performed with various cell/ECM protein pairs involving unique integrins: aibi (PC12/type I collagen), a 2 bi (HT1080/type I collagen), a 6 b 4 (HT29-D4/laminin), a n b 6 (HT29- D4/fibronectin), a n b3 (HT29 D4/fibrinogen), a n b d (HT29-D4/vitronectin) and a 5 bi (K562/fibronectin). Cells were preincubated without or with 15mM of RK for 30 min at room temperature.
  • FIG. 10A-C Molecular modeling and molecular dynamics of RK peptide
  • A We present the predicted model returned by the PEP-FOLD server. The Ca atoms of each of the residues are indicated in spheres. The disulfide bond between the 3 rd and the 12 th amino acid is colored in red.
  • B RMSD time evolution (upper panel) and RMSF profile (lower panel) of RK peptide calculating for a molecular dynamics trajectory of 300 ns.
  • C Potential of mean forces of the RK peptide calculated from the molecular dynamics simulation. The reaction coordinates consist of the radius of gyration (Rg) and the RMSD values relative to an average structure computed from the 300 ns simulation time.
  • Rg radius of gyration
  • Fig. 1 1A-C Peptide-Protein docking of RK with a1 b1
  • A RMSD time evolution of the best retained complex between RK and a1 b1 simulated for 10 ns.
  • B Interaction of RK peptide (red) with the collagen IV binding site on the a1 b1.
  • C The key interaction with the integrin are expressed in more details.
  • the M7 residue is capable to interact with a small hydrophobic pocket on the surface of the integrin formed by the residues in yellow.
  • Fig. 12A-C Analysis of the interaction of RK with a n b3
  • A We fitted the atoms of the ECD segment from each snapshot the molecular dynamics trajectory of RK to those of the RGD coordinates from the 1 L5G PDB structure. The ROSETTA clash score was then calculated. The reaction coordinates consists of the distance between E11 CA and D13 CG, and the angle between En CA, C12 CA and D13 CG.
  • B The conformation of RK with the least RMSD value is presented along with RGD peptide (yellow sticks).
  • C The complex of RK with a n b 3 was constructed by fitting the conformation corresponding to the lowest clash score of the ECD segment with the RGD coordinates of the cilengetide from the PDB structure 1 LG5.
  • the term "about” is understood as within a range of normal tolerance in the art, for example within 2 standard deviations of the mean. About can be understood as within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1 %, 0.5%, 0.1 %, 0.05%, or 0.01 % of the stated value. Unless otherwise clear from context, all numerical values provided herein are modified by the term about.
  • compositions or methods provided herein can be combined with one or more of any of the other compositions and methods provided herein.
  • the terms “determining”, “assessing”, “assaying”, “measuring” and “detecting” refer to both quantitative and qualitative determinations, and as such, the term “determining” is used interchangeably herein with “assaying,” “measuring,” and the like. Where a quantitative determination is intended, the phrases “determining an amount” of an analyte and the like can be used. Where a qualitative and/or quantitative determination is intended, the phrase “determining a level" of an analyte or “detecting” an analyte is used.
  • control reference method is a reference method in which all of the parameters are identical to those of the method being compared with exception of the variable being tested.
  • subject is meant a mammal, including, but not limited to, a human or non human mammal, such as a bovine, equine, canine, ovine, or feline.
  • Ranges provided herein are understood to be shorthand for all of the values within the range.
  • a range of 1 to 50 is understood to include any number, combination of numbers, or sub-range from the group consisting 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 ,
  • Binding affinity generally refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g., of an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein, "binding affinity”, “bind to”, “binds to” or “binding to” refers to intrinsic binding affinity that reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody Fab fragment and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD).
  • Standard assays for quantifying binding and determining binding affinity include, e.g., equilibrium dialysis, equilibrium binding, gel filtration, surface plasmon resonance, the use of a labeled binding partners, ELISAs and indirect binding assays (e.g., competitive inhibition assays).
  • the dissociation constant of a protein can be determined by contacting the protein with a binding partner and measuring the concentration of bound and free protein as a function of its concentration.
  • the term “linked” identifies a covalent linkage between two different groups (e.g., nucleic acid sequences, polypeptides, polypeptide domains) that may have an intervening atom or atoms between the two groups that are being linked.
  • “directly linked” identifies a covalent linkage between two different groups (e.g., nucleic acid sequences, polypeptides, polypeptide domains) that does not have any intervening atoms between the two groups that are being linked.
  • disintegrin refers to a class of cysteine-rich proteins that are potent soluble ligands of integrins and which are involved in regulating many processes such as cell-cell and cell-extracellular matrix adhesion, migration and invasion, cell cycle progression, differentiation and cell type speciation during development of many metazoan organisms, cell death and apoptosis.
  • the tri-peptide motif RGD (Arg-Gly-Asp) is conserved in most monomeric disintegrins and is located at the tip of a flexible loop, the integrin-binding loop, which is stabilized by disulfide bonds and protruding from the main body of the polypeptide chain.
  • disintegrins purified from snake venoms bind to the fibrinogen receptor, integrin ⁇ 3 ⁇ 4,b3 the binding of which results in the inhibition of fibrinogen-dependent platelet aggregation. Most disintegrins also bind to a n b3 (a vitronectin receptor) and anbi (a fibronectin receptor) in an RGD-dependent manner.
  • Disease refers to any condition, infection, disorder, or syndrome that requires medical intervention or for which medical intervention is desirable. Such medical intervention can include treatment, diagnosis, and/or prevention.
  • Treatment covers any administration or application of remedies for disease in a mammal, including a human, and includes inhibiting the disease, arresting its development, or relieving the disease, for example, by causing regression, or restoring or repairing a lost, missing, or defective function; or stimulating an inefficient process.
  • the term includes obtaining a desired pharmacologic and/or physiologic effect, covering any treatment of a pathological condition or disorder in a mammal, including a human.
  • the effect may be prophylactic in terms of completely or partially preventing a disorder or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disorder and/or adverse affect attributable to the disorder.
  • the invention provides both treatment and prophylaxis.
  • It includes (1) preventing the disorder from occurring or recurring in a subject who may be predisposed to the disorder but is not yet symptomatic, (2) inhibiting the disorder, such as arresting its development, (3) stopping or terminating the disorder or at least its associated symptoms, so that the host no longer suffers from the disorder or its symptoms, such as causing regression of the disorder or its symptoms, for example, by restoring or repairing a lost, missing or defective function, or stimulating an inefficient process, or (4) relieving, alleviating, or ameliorating the disorder, or symptoms associated therewith, where ameliorating is used in a broad sense to refer to at least a reduction in the magnitude of a parameter, such as inflammation, pain, and/or tumor size.
  • a parameter such as inflammation, pain, and/or tumor size.
  • an“effective amount” means the amount of an agent that is effective for producing a desired effect in a subject.
  • the term“therapeutically effective amount” denotes that amount of a drug or pharmaceutical agent that will elicit therapeutic response of an animal or human that is being sought.
  • the actual dose which comprises the effective amount may depend upon the route of administration, the size and health of the subject, the disorder being treated, and the like.
  • Therapeutically effective can mean to result in at least one anti-tumor effect chosen from reduction of tumor cell growth and reduction of tumor cell proliferation in the subject.
  • therapeutically effective in treating cancer is meant that the use of the peptide of the invention to treat cancer in a patient results in any demonstrated clinical benefit compared with no therapy (when appropriate) or to a known standard of care.
  • Clinical benefit can be assessed by any method known to one of ordinary skill in the art.
  • clinical benefit is assessed based on objective response rate (ORR) (determined using RECIST version 1.1), duration of response (DOR), progression-free survival (PFS), and/or overall survival (OS).
  • ORR objective response rate
  • DOR duration of response
  • PFS progression-free survival
  • OS overall survival
  • a complete response indicates therapeutic benefit.
  • a partial response indicates therapeutic benefit.
  • stable disease indicates therapeutic benefit.
  • an increase in overall survival indicates therapeutic benefit.
  • therapeutic benefit may constitute an improvement in time to disease progression and/or an improvement in symptoms or quality of life. In other embodiments, therapeutic benefit may not translate to an increased period of disease control, but rather a markedly reduced symptom burden resulting in improved quality of life.
  • a therapeutic benefit may be observed using the peptide of the invention alone (monotherapy) or adjunctive to, or with, other anti-cancer therapies and/or targeted or non- targeted anti-cancer agents.
  • Various methods for assessing therapeutic benefit are well known in the art.
  • therapeutic effect for determination of a particular dosage regimen is assessed using standard clinical tests designed to measure the response to a new treatment for cancer.
  • standard clinical tests designed to measure the response to a new treatment for cancer.
  • RECIST Response Evaluation Criteria In Solid Tumors
  • ECOG Eastern Cooperative Oncology Group
  • irRC immune-related response criteria
  • disease evaluable by assessment of tumor antigens (5) validated patient reported outcome scales, and/or (6) Kaplan-Meier estimates for overall survival and progression free survival.
  • pharmaceutically acceptable carrier may refer to compounds and compositions that are suitable for use in human or animal subjects, as for example, for therapeutic compositions administered for the treatment of a disorder or disease of interest.
  • composition that may be administered to a mammalian host, e.g., orally, parenterally, topically, by inhalation spray, intranasally, or rectally, in unit dosage formulations containing conventional non-toxic carriers, diluents, adjuvants, vehicles and the like.
  • parenteral includes subcutaneous injections, intravenous, intramuscular, intracisternal injection, or infusion techniques.
  • Inhibit as used herein may mean prevent, suppress, repress, reduce or eliminate.
  • the invention provides peptide RK1 (IDCSKVNLTAECSS; SEQ ID NO:2, a 14 mer), which is the first very short peptide from Buthus occitanus tunetanus that inhibits tumor cell migration, proliferation and angiogenesis.
  • the invention provides peptide RK (IDCGTVMIPSECDPKSS; SEQ ID NO: 1 , a 17 mer), which is the first scorpion peptide with dual disintegrin activity on a1 b1 and anb3 integrins.
  • the peptide is isolated from Buthus occitanus tunetanus scorpion venom. Methods for isolation are exemplified in the EXAMPLES.
  • the peptide is produced synthetically.
  • the peptides of the invention include modifications of the RK1 and RK peptides according to which the resulting peptides are not natural. In some embodiments, these are modifications to the actual sequence or to the chemistry of the amino acids (e.g., non-natural amino acids), as exemplified below. In some embodiments, these modifications are additions or conjugations of the peptides with other molecules, as exemplified below. In some embodiments, the modifications are the result of the formulation of the peptides with compounds that affect the properties of the peptides, be it solubility, half-life, antigenicity, biological activity, stability, or any other physical and/or chemical change to the native peptide.
  • the invention provides a peptide whose amino acid sequence has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to IDCSKVNLTAECSS or IDCGTVMIPSECDPKSS (RK1 ;SEQ ID NO:2, or RK; SEQ ID NO: 1).
  • Peptides of the invention may be able to inhibit tumor cell migration, proliferation, and/or angiogenesis.
  • Peptides of the invention may also have dual disintegrin activity on a1 b1 and anb3 integrins.
  • the amino acid sequence of the peptide is IDCSKVNLTAECSS (RK1 ; SEQ ID NO:2). In another embodiment, the amino acid sequence of the peptide is IDCGTVMIPSECDPKSS (RK; SEQ ID NO: 1).
  • the sequence comprises a disulfide bond.
  • the sequence comprises at least one substitution modification, insertion, or deletion modification relative to IDCSKVNLTAECSS (SEQ ID NO:2) and IDCGTVMIPSECDPKSS (SEQ ID NO: 1).
  • the peptide is not able to alter cell adhesion through a2b1 , anb6, anb5, a5b1 and a6b4 integrins, but significantly reduces the adhesive activity of a1 b1 and anb3 integrins to extracellular matrix proteins.
  • the extracellular matrix protein comprises collagen.
  • the extracellular matrix protein comprises vitronectin, fibronectin, fibrinogen, osteopoietin, and/or bone sialoprotein.
  • the peptide competes with a synthetic RGD peptide for binding to aibi and/or a n b3 integrins. Competition assays are well known in the art.
  • the sequence comprises the segment V6, M7, and I8 of the RK peptide and (and still binds a1 b1 integrin).
  • the sequence comprises residues SnECDPKSi 6 (aa 11-16 of SEQ ID NO:3), Ei 2 CDPKi 5 (aa 12-15 of SEQ ID NO:3), or Di 4 PKi 6 (aa 14-16 of SEQ ID NO:3), of the RK peptide, or DCSK (aa 2-5 of SEQ ID NO:2), DCSKXXXXECS (SEQ ID NO:4) or DCSKXXXXXECDS (SEQ ID NO:5) of the RK1 peptide.
  • Peptide K can be divided into three subsegments using the computation study and molecular model described in the EXAMPLES.
  • the peptide comprises at least one of the RK segments. In one embodiment, the peptide comprises two of these RK segments. In one embodiment, the peptide comprises all segments. In some embodiments, the sequence of one or more of the segments has been modified.
  • sequence of the peptide is designed while considering the molecular docking studies and results described in this disclosure.
  • the invention comprises the application of the information gathered using any one or more of the computational studies, molecular dynamic studies, a molecular docking model described in this disclosure, particularly in designing disintegrins and peptides of the invention.
  • the terms "at least 70% identical” or “at least 70% identity” means that a polypeptide or peptide sequence or a polynucleotide sequence shares 70%-100% sequence identity with a reference sequence.
  • This range of identity is inclusive of all whole numbers (e.g., 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) or partial numbers (e.g., 72.15, 87.27%, 92.83%, 98.11 %-to two significant Fig.s) embraced within the recited range numbers, therefore forming a part of this description. For example, an amino acid sequence with 200 residues that share 85% identity with a reference sequence would have 170 identical residues and 30 non-identical residues.
  • an amino acid sequence with 235 residues may have 200 residues that are identical to a reference sequence, thus the amino acid sequence will be 85.1 1 % identical to the reference sequence.
  • the terms “at least 80%, “ “at least 90%, “ “at least 95%” and “at least 99%” and the like are inclusive of all whole or partial numbers within the recited range.
  • the terms “greater than 95% identical” or “greater than 95% identity” means that a sequence shares 95.01 %-100% sequence identity with a reference sequence. This range is all inclusive. Differences in identity can be due to additions, deletions or substitutions of residues in a first sequences compared to a second sequence.
  • the percent identity can be measured using sequence comparison software or algorithms or by visual inspection.
  • Various algorithms and software are known in the art that can be used to obtain alignments of amino acid or nucleotide sequences (see e.g., Karlin et al., 1990, Proc. Natl. Acad. Sci. USA, 87:2264-2268, as modified in Karlin et al., 1993, Proc. Natl. Acad. Sci. USA, 90:5873-5877, and incorporated into the NBLAST and XBLAST programs (Altschul et al., 1991 , Nucleic Acids Res., 25:3389-3402).
  • Gapped BLAST can be used as described in Altschul et al., 1997, Nucleic Acids Res. 25:3389-3402.
  • BLAST-2, WU- BLAST-2 Altschul et al., 1996, Methods in Enzymology, 266:460-480
  • ALIGN ALIGN-2 (Genentech, South San Francisco, Calif.) or Megalign (DNASTAR).
  • the peptide is 20 amino acids long or less. In some embodiments, the peptide is between 20 and 15 amino acids long. In some embodiments, the peptide is 14 amino acids long or less. In some embodiments, the peptide is 17 amino acids long or less.
  • the peptide comprises a modification relative to IDCSKVNLTAECSS (SEQ ID NO:2) or IDCGTVMIPSECDPKSS (SEQ ID NO: 1).
  • Substitutes for an amino acid within the peptide sequence may be selected from other members of the class to which the amino acid belongs.
  • the nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine.
  • Amino acids containing aromatic ring structures include phenylalanine, tryptophan, and tyrosine.
  • the polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine.
  • the positively charged (basic) amino acids include arginine and lysine.
  • the negatively charged (acidic) amino acids include aspartic acid and glutamic acid. For example, one or more amino acid residues within the sequence can be substituted by another amino acid of a similar polarity, which acts as a functional equivalent, resulting in a silent alteration.
  • the peptide of the present disclosure comprises one or more of the following conservative amino acid substitutions: replacement of an aliphatic amino acid, such as alanine, valine, leucine, and isoleucine, with another aliphatic amino acid; replacement of a serine with a threonine; replacement of a threonine with a serine; replacement of an acidic residue, such as aspartic acid and glutamic acid, with another acidic residue; replacement of a residue bearing an amide group, such as asparagine and glutamine, with another residue bearing an amide group; exchange of a basic residue, such as lysine and arginine, with another basic residue; and replacement of an aromatic residue, such as phenylalanine and tyrosine, with another aromatic residue.
  • conservative amino acid substitutions replacement of an aliphatic amino acid, such as alanine, valine, leucine, and isoleucine, with another aliphatic amino acid
  • Particularly preferred amino acid substitutions include: a) Ala for Glu or vice versa, such that a negative charge may be reduced; b) Lys for Arg or vice versa, such that a positive charge may be maintained; c) Ala for Arg or vice versa, such that a positive charge may be reduced; d) Glu for Asp or vice versa, such that a negative charge may be maintained; e) Ser for Thr or vice versa, such that a free --OH can be maintained; f) Gin for Asn or vice versa, such that a free NH2 can be maintained; g) lie for Leu or for Val or vice versa, as roughly equivalent hydrophobic amino acids; h) Phe for Tyr or vice versa, as roughly equivalent aromatic amino acids; and i) Ala for Cys or vice versa, such that disulphide bonding is affected.
  • nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the peptide sequence.
  • Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids, 2,4- diaminobutyric acid, a-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, g- Abu, e-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, b-alanine, fluoro-amino acids, designer amino acids such
  • the peptide comprises an N-terminal and/or C-terminal modification.
  • the modifications can comprise the addition or replacement of the termini with amino acid analogs.
  • Peptidomimetics can be incorporated into a peptide to induce or favor specific secondary structures.
  • a desamino or descarboxy residue can be incorporated at the terminal ends of the peptide, so that there is no terminal amino or carboxyl group, to decrease susceptibility to proteases or to restrict conformation.
  • C-terminal functional groups include amide, amide lower alkyl, amide di (lower alkyl), lower alkoxy, hydroxy, and carboxy, and the lower ester derivatives thereof, and the pharmaceutically acceptable salts thereof.
  • the peptide might also comprise other modifications.
  • the peptide may be glycosylated, phosphorylated, sulfated, glycosylated, animated, carboxylated, acetylated.
  • the C-terminal may be modified with addition of peptide alcohols and aldehydes, addition of esters, addition of p-nitorailine and thioesteres and multipelantigens peptides.
  • the N-terminal and side chains may be modified by PEGylation, acetylation, formylation, addition of a fatty acid, addition of benzoyl, addition of bromoacetyl, addition of pyroglutamyl, succinylation, addition of tetrabutyoxycarbonyl and addition of 3-mercaptopropyl, acylations (e.g. lipopeptides), biotinylation, phosphorylation, sulfation, glycosylation, introduction of maleimido group, chelating moieties, chromophores and flurophores.
  • PEGylation e.g. lipopeptides
  • the peptide may be conjugated to a fatty acid, e.g. the peptide is myristoylated.
  • a fatty acid may be conjugated to the N-terminus of the peptide, such fatty acids include caprylic acid (C8), capric acid (C10), lauric acid (C12), myristic acid (C14), palmitic acid (C16) or stearic acid (C18) etc.
  • cysteines in peptides can be palmitoylated.
  • the peptide may be conjugated or linked to another peptide, such as a carrier peptide.
  • the carrier peptide may facilitate cell-penetration, such as antennapedia peptide, penetratin peptide, TAT, tranportan or polyarginine.
  • the peptides may be cyclic.
  • the peptide disclosed herein may be cyclized by adding a single or multiple disulfide bridges, adding a single or multiple amide bonds between the N- and C-terminus, heat to tail cyclization, side chain cyclization (e.g. lactam bridge, thioester), hydrocarbon-stabled peptides.
  • the peptide may be labeled with heavy isotope labeling, e.g. 15 N, 13 C, FITC, conjugation to a carrier protein, conjugation to imaging agent, FRET substrates with a fluorophore/quencher pair, peptide-DNA conjugation, peptide-RNA conjugation and peptide- enzyme labeling.
  • the peptide may be within a fusion protein such as fused to a polypeptide or peptide which promotes oligomerization, such as a leucine zipper domain; a polypeptide or peptide which increases stability or to increase half-life, such as an immunoglobulin constant region; and a polypeptide which has a therapeutic activity different from peptide or the invention, a chemotherapeutic agent, an antibody or protein for tissue specific targeting.
  • Peptides of the invention include fusion peptides.
  • fusion peptides may comprise peptides of the invention linked, for example, to antibodies that target the peptides to diseased tissue, for example, tumor tissue or the retina.
  • the peptides may be fused with the constant domain of immunoglobulins (IgA, IgE, IgG, IgM), or portions thereof (CH1 , CH2, CH3, or any combination thereof), resulting in chimeric polypeptides.
  • immunoglobulins IgA, IgE, IgG, IgM
  • CH1 , CH2, CH3, or any combination thereof immunoglobulins
  • fusion polypeptides or proteins can facilitate purification and may show an increased half-life in vivo.
  • Such fusion proteins may be more efficient in binding and neutralizing other molecules than monomeric polypeptides or fragments thereof alone.
  • Fusion proteins of the invention also include the peptides fused with albumin, for example recombinant human serum albumin or fragments or variants thereof (see, e.g., US Patent No. 5876969, EP Patent 0413622 and US Patent No. 5766883).
  • Fusions can be made either at the amino terminus or at the carboxy terminus of the polypeptide.
  • the fusion proteins may be direct with no linker or adapter molecule or indirect using a linker or adapter molecule.
  • a linker or adapter molecule may be one or more amino acid residues, typically up to about 20 to about 50 amino acid residues.
  • a linker or adapter molecule may also be designed with a cleavage site for a protease to allow for the separation of the fused moieties.
  • the peptide may be fused to one or more domains of an Fc region of human IgG to increase the half-life of the peptide or the addition of a Fab variable domain to shorten the half-life of the peptide.
  • a peptide of the invention can be modified with or covalently coupled to one or more of a variety of hydrophilic polymers to increase solubility and circulation half-life of the peptide.
  • Suitable nonproteinaceous hydrophilic polymers for coupling to a peptide include, but are not limited to, polyalkylethers as exemplified by polyethylene glycol and polypropylene glycol, polylactic acid, polyglycolic acid, polyoxyalkenes, polyvinylalcohol, polyvinylpyrrolidone, cellulose and cellulose derivatives, dextran, and dextran derivatives, polyvinyl pyrrolidones, glycopeptides, and polyamino acids.
  • hydrophilic polymers have an average molecular weight ranging from about 500 to about 100,000 daltons, from about 2,000 to about 40,000 daltons, or from about 5,000 to about 20,000 daltons.
  • the peptide can be derivatized with or coupled to such polymers using any of the methods known in the art.
  • the peptide is linked to a coupling partner.
  • the coupling partner is chosen from an effector molecule, a label, a drug, a toxin, a carrier, and a transport molecule.
  • polypeptide is modified by the addition of cysteine or biotin.
  • the peptide is modified by a moiety to facilitate crosslinking.
  • the moiety is chosen from benzophenone, maleimide, and activated esters.
  • the peptides may be in the form of multimers.
  • multimers of 2, 3 or more individual RK1 , RK, or other peptide of the invention monomeric units, or two or more other peptides of the invention, or their combination, are within the scope of the invention.
  • such multimers may be used to prepare a monomeric peptide by preparing a multimeric peptide that includes the monomeric unit, and a cleavable site (i.e., an enzymatically cleavable site), and then cleaving the multimer to yield a desired monomer.
  • the multimers can be homomers or heteromers.
  • the use of multimers can increase the binding affinity for an integrin.
  • the integrin is a1 b1 and/or anb3 integrin.
  • Homodimeric and monomeric peptides described herein may be used for any purposes for which native homodimeric disintegrins may be employed.
  • the peptides described herein are intended, at least in some embodiments, to be administered to a human or other mammal to treat or prevent a disorder associated with tumor cell migration, tumor cell proliferation, cell adhesion, and angiogenesis.
  • Peptides are typically administered parenterally, e.g., by intravenous, subcutaneous, or intramuscular injection, or via the intranasal cavity, and may be readily metabolized by plasma proteases.
  • the peptide may be delivered in microcapsules of poly(DL-lactide-co-glycolide) - controlled release over 30 days.
  • a prodrug is produced by modifying a pharmaceutically active compound such that the active compound will be regenerated upon in vivo administration.
  • the prodrug can be designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug.
  • Peptides or polypeptides can be conjugated to various moieties, such as polymeric moieties, to modify the physiochemical properties of the peptide drugs, for example, to increase resistance to acidic and enzymatic degradation and to enhance penetration of such drugs across mucosal membranes.
  • the peptides can be provided in a suitable capsule or tablet with an enteric coating, so that the peptide is not released in the stomach.
  • the peptide can be provided as a prodrug, such as the prodrugs described above.
  • the peptides are present in drug delivery devices as prodrugs.
  • the activity of the peptides of the invention can be measured by methods that are routine in that art. In some embodiments, the activity is measured according to the methods described in the EXAMPLES.
  • cell detachment and adhesion may be detected by any suitable method, which may include, but are not limited to, cell cytometry (e.g. trypan blue), fluorescent based cell detection assays (e.g. Calcein AM (InVitrogen, Inc.), and Mitotracker Red (InVitrogen, Inc.), luminescent based detection assays (e.g. Cell-Titer glo (Promega, Inc.) and spectrophotometry based detection assays (e.g. crystal violet, MTS/MTT assays such as Promega CellTiter 96.RTM. AQueous Non-Radioactive Cell Proliferation Assay, Promega, Inc. and Chemicon Cell Adhesion Assays).
  • cell cytometry e.g. trypan blue
  • fluorescent based cell detection assays e.g. Calcein AM (InVitrogen, Inc.), and Mitotracker Red (InVitrogen, Inc.
  • cell migration or invasion may be detected by any suitable method, which may include, but are not limited to, the scratch wound assay, cell invasion assays using fluorescent detection of cell invasion (e.g. activin, serum) (BD BioCoat matrigel invasion chambers, Fisher Scientific; EMD, Calbiochem; Chemicon International) with or without the use of a chemotaxic agent.
  • suitable method may include, but are not limited to, the scratch wound assay, cell invasion assays using fluorescent detection of cell invasion (e.g. activin, serum) (BD BioCoat matrigel invasion chambers, Fisher Scientific; EMD, Calbiochem; Chemicon International) with or without the use of a chemotaxic agent.
  • Cell viability may be detected by any suitable method, which may include, but are not limited to, cell cytometry (e.g. trypan blue), fluorescent based cell detection assays (e.g. calcein AM (InVitrogen, Inc.), and Mitotracker Red (InVitrogen, Inc.)), luminescent based detection assays (e.g. Cell-Titer glo (Promega, Inc.)) and spectrophotometry based detection assays (e.g. crystal violet, MTS/MTT assays such as Promega CellTiter 96.RTM. AQueous Non- Radioactive Cell Proliferation Assay, Promega, Inc.).
  • cell cytometry e.g. trypan blue
  • fluorescent based cell detection assays e.g. calcein AM (InVitrogen, Inc.), and Mitotracker Red (InVitrogen, Inc.)
  • luminescent based detection assays e.g. Cell-Titer
  • the effects of the peptides of the invention on cell proliferation may be assessed by any suitable method known in the art, including the method exemplified in the EXAMPLES section. Numerous methods of assessing cell proliferation are known in the art, including, for example, DNA synthesis cell proliferation assays (BrdU, IdU and CldU, EdU), metabolic proliferation assays (MTT, XTT, WST-1), detection of proliferation markers (PCNA, Ki67, MCM- 2), measuring ATP.
  • the detection methods for these assays can be ICC, IHC, FACS, ELISA, WB, microscopy, among others.
  • mice Animal models to determine antitumor efficacy of a compound are generally carried out in mice. Either murine tumor cells are inoculated subcutaneously into the hind flank of mice from the same species (syngeneic models) or human tumor cells are inoculated subcutaneously into the hind flank of severe combined immune deficient (SCID) mice or other immune deficient mouse (nude mice) (xenograft models). Advances in mouse genetics have generated a number of mouse models for the study of various human diseases including cancer.
  • SCID severe combined immune deficient mice
  • xenograft models Advances in mouse genetics have generated a number of mouse models for the study of various human diseases including cancer.
  • the MMHCC (Mouse models of Human Cancer Consortium) web page (emice.nci.nih.gov), sponsored by the National Cancer Institute, provides disease-site-specific compendium of known cancer models, and has links to the searchable Cancer Models Database (cancermodels.nci.nih.gov), as well as the NCI-MMHCC mouse repository.
  • Mouse repositories can also be found at: The Jackson Laboratory, Charles River Laboratories, Taconic, Harlan, Mutant Mouse Regional Resource Centers (MMRRC) National Network and at the European Mouse Mutant Archive.
  • Such models may be used for in vivo testing of the peptides of the invention, as well as for determining a therapeutically effective dose.
  • RK and RK1 bear many excellent properties that can serve as an alternative protein scaffold for biomedical application.
  • RK and RK1 are bioactive mini-proteins from scorpion venoms that have the unique topological feature of cyclic backbone . Because of this structure, they are ultra-stable, present low immunogenicity (important to reduce unexpected side effects and damage of healthy tissue) and have advantages in passing tissue barrier. Thus, they attract interest as a peptide- based templates for drug design applications or diagnostic agents. This allows them to be used as pharmaceutical templates onto which bioactive peptide sequences can be grafted. Production of native or modified peptide
  • RK and RK1 present many other benefits, such as cost reduction during development/production (chemical and genetical engineering) due to their limited amino-acid composition.
  • the peptides of the present invention can be synthesized de novo using conventional solid phase synthesis methods.
  • the peptide chain is prepared by a series of coupling reactions in which the constituent amino acids are added to the growing peptide chain in the desired sequence.
  • N-protecting groups e.g., the carbobenzyloxy group or the t-butyloxycarbonyl group
  • various coupling reagents e.g., dicyclohexylcarbodiimide or carbonyldimidazole
  • various active esters e.g., esters of N- hydroxyphthalimide or N-hydroxy-succinimide
  • various cleavage reagents e.g., trifluoroactetic acid (TFA), HCI in dioxane, boron tris-(trifluoracetate) and cyanogen bromide
  • reaction in solution with isolation and purification of intermediates are within the skill in the art.
  • a preferred peptide synthesis method follows conventional Merrifield solid phase procedures well known to those skilled in the art.
  • compositions comprising one or more of the peptides of the invention.
  • the compositions comprise a pharmaceutically acceptable carrier, diluent, or excipient.
  • pharmaceutically acceptable carrier refers to a carrier for administration of a therapeutic agent.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., intravenous, oral topical, aerosol, suppository, parenteral or spinal injection.
  • Such carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.
  • pharmaceutically acceptable carriers include, but are not limited to pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives.
  • Suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents. Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.
  • Excipients or other additives included in a composition have different purposes depending, for example on the nature of the drug, and the mode of administration.
  • examples of generally used excipients include, without limitation: stabilizing agents, solubilizing agents and surfactants, buffers, antioxidants and preservatives, tonicity agents, bulking agents, lubricating agents, emulsifiers, suspending or viscosity agents, inert diluents, fillers, disintegrating agents, binding agents, wetting agents, lubricating agents, antibacterials, chelating agents, sweetners, perfuming agents, flavouring agents, coloring agents, administration aids, and combinations thereof.
  • the peptides of the invention can be formulated for sustained release or comprise a sustained release carrier.
  • the sustained release carrier comprises a semipermeable polymer matrix.
  • the semipermeable polymer matrix comprises one or more of polylactides copolymers of L-glutamic acid and gamma ethyl-L- glutamate, poly (2-hydroxyethyl-methacrylate), or ethylene vinyl acetate.
  • the peptides of the invention can be formulated for oral, sublingual, intranasal, intraocular, rectal, transdermal, mucosal, topical or parenteral administration for the therapeutic or prophylactic treatment of a variety of disorders, as described below.
  • Parenteral modes of administration include without limitation, intradermal, subcutaneous (s.c., s.q., sub-Q, Hypo), intramuscular (i.m.), intravenous (i.v.), intraperitoneal (i.p.), intra-arterial, intramedulary, intracardiac, intra-articular (joint), intrasynovial (joint fluid area), intracerabral or intracranial, intraspinal, intracisternal, and intrathecal (spinal fluids). Any known device useful for parenteral injection or infusion of drug formulations can be used to effect such administration.
  • compounds of the present invention can be mixed with conventional pharmaceutical carriers and excipients and used in the form of solutions, emulsions, tablets, capsules, soft gels, elixirs, suspensions, syrups, wafers and the like.
  • parenteral administration in particular intravenous injection, is most commonly used for administering peptides, and the composition may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water.
  • a suitable vehicle e.g., sterile pyrogen-free water.
  • a sterile composition of the peptide and optionally one or more additives, including solubilizers or surfactants, can be dissolved or suspended in any of the commonly used intravenous fluids and administered by infusion.
  • Intravenous fluids include, without limitation, physiological saline, phosphate buffered saline, 5% glucose or Ringer's solution.
  • a sterile formulation of the peptide of the present invention or suitable soluble salts or prodrugs forming the peptide can be dissolved and administered in a pharmaceutical diluent such as Water-for-lnjection (WFI), physiological saline or 5% glucose.
  • WFI Water-for-lnjection
  • a suitable insoluble form of the compound may be prepared and administered as a suspension in an aqueous base or a pharmaceutically acceptable oil base, e.g. an ester of a long chain fatty acid such as ethyl oleate.
  • Sterile injectable solutions are prepared by incorporating the peptides of the invention in the required amount in the appropriate solvent with various of the other ingredients enumerated below, as required. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and/or the other ingredients. In the case of sterile powders for the preparation of sterile injectable solutions, suspensions or emulsion, the preferred methods of preparation are vacuum-drying or freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered liquid medium thereof.
  • the liquid medium should be suitably buffered if necessary and the liquid diluent first rendered isotonic prior to injection with sufficient saline or glucose.
  • the composition must be stable under the conditions of manufacture and storage, and preserved against the contaminating action of microorganisms, such as bacteria and fungi. It will be appreciated that endotoxin contamination should be kept minimally at a safe level, for example, less that 0.5 ng/mg protein.
  • Suitable pharmaceutically acceptable carriers for parenteral composition include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine;
  • Aqueous injection suspensions may contain compounds which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, dextran, or the like.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • suspensions of the active compounds may be prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl cleats or triglycerides, or liposomes.
  • solid formulations such as tablets and capsules are particularly useful. Sustained released or enterically coated preparations may also be devised. For pediatric and geriatric applications, suspension, syrups and chewable tablets are especially suitable.
  • the pharmaceutical compositions are in the form of, for example, tablets, capsules, suspensions or liquid syrups or elixirs, wafers and the like.
  • excipient or additives include, but are not limited to inert diluents, fillers, disintegrating agents, binding agents, wetting agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives.
  • the oral pharmaceutical composition is preferably made in the form of a unit dosage containing a therapeutically-effective amount of the active ingredient.
  • dosage units are tablets and capsules.
  • the tablets and capsules which can contain, in addition to the active ingredient, conventional carriers such as: inert diluents (e.g., sodium and calcium carbonate, sodium and calcium phosphate, and lactose), binding agents (e.g., acacia gum, starch, gelatin, sucrose, polyvinylpyrrolidone (Providone), sorbitol, or tragacanth methylcellulose, sodium carboxymethylcellulose, hydroxypropyl methylcellulose, and ethylcellulose), fillers (e.g., calcium phosphate, glycine, lactose, maize-starch, sorbitol, or sucrose), lubricants or lubricating agents (e.g., magnesium stearate or other metallic stearates, stearic acid, poly
  • Oral liquid preparations generally in the form of aqueous or oily solutions, suspensions, emulsions, syrups or elixirs, may contain conventional additives such as suspending agents, emulsifying agents, non-aqueous agents, preservatives, coloring agents and flavoring agents.
  • additives for liquid preparations include acacia, almond oil, ethyl alcohol, fractionated coconut oil, gelatin, glucose syrup, glycerin, hydrogenated edible fats, lecithin, methyl cellulose, methyl or propyl para-hydroxybenzoate, propylene glycol, sorbitol, or sorbic acid.
  • flavoring agents such as peppermint, oil of wintergreen, cherry, grape, fruit flavoring or the like can also be used. It may also be desirable to add a coloring agent to make the dosage form more aesthetic in appearance or to help identify the product.
  • the compounds of present invention can also be prepared in suitable forms to be applied to the skin, or mucus membranes of the nose and throat, and can take the form of creams, ointments, liquid sprays or inhalants, lozenges, or throat paints.
  • Such topical formulations further can include chemical compounds such as dimethylsulfoxide (DMSO) to facilitate surface penetration of the active ingredient.
  • DMSO dimethylsulfoxide
  • the compounds of the present invention can be presented in liquid or semi-liquid form formulated in hydrophobic or hydrophilic bases as ointments, creams, lotions, paints or powders.
  • the compounds of the present invention can be administered in the form of suppositories admixed with conventional carriers such as cocoa butter, wax or other glyceride.
  • the peptides of the invention may be formulated into a composition in a free acid or base, neutral or salt form.
  • Pharmaceutically acceptable salts are salts that substantially retain the biological activity of the free acid or base. These include the acid addition salts, e.g., those formed with the free amino groups of a proteinaceous composition, or which are formed with inorganic acids such as for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric or mandelic acid.
  • Salts formed with the free carboxyl groups can also be derived from inorganic bases such as for example, sodium, potassium, ammonium, calcium or ferric hydroxides; or such organic bases as isopropylamine, trimethylamine, histidine or procaine. Pharmaceutical salts tend to be more soluble in aqueous and other protic solvents than are the corresponding free base forms.
  • compositions comprising a peptide of the present invention can contain from about 0.1 % to about 99.9%, about 1 % to about 98%, about 5% to about 95%, about 10% to about 80% or about 15% to about 60% by weight of the active peptide.
  • the invention provides peptide RK1 (a 14 mer), which is the first very short peptide from Buthus occitanus tunetanus that inhibits tumor cell migration, proliferation, and angiogenesis.
  • the peptides may be used to treat diseases or disorders mediated by tumor cell migration, primarily cancer.
  • the peptides may be used to treat diseases or disorders mediated by tumor cell proliferation, also known as neoplastic disorders.
  • the peptides may be used to treat diseases or disorders mediated by angiogenesis. Diseases associated with these aspects of tumor development including tumor cell spreading from the original site, colonisation of new tumor sites, and neovascularization can also be treated with the peptides of the invention.
  • the peptides of the invention are used as anti-cancer agents.
  • anti-cancer agent includes an agent that can inhibit cancer cell growth/proliferation, migration, and/or adhesion.
  • the peptides of the invention can be used to inhibit angiogenesis, or a combination thereof. Accordingly, the peptides of the invention are capable of acting both on cancer and on non-cancer cells (e.g., endothelial cells, fibroblasts, stromal cells).
  • neoplasm As used herein, the terms “neoplasm”, “neoplastic disorder”, “neoplasia” “cancer,” “tumor” and “proliferative disorder” refer to cells having the capacity for autonomous growth/proliferation, i.e. , an abnormal state or condition characterized by rapidly proliferating cell growth which generally forms a distinct mass that show partial or total lack of structural organization and functional coordination with normal tissue. Cancer is meant to include all types of cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness. The terms are meant to encompass hematopoietic cancers (e.g.
  • lymphomas or leukemias as well as solid cancers (e.g. sarcomas or carcinomas), including all types of pre-cancerous and cancerous growths, or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness.
  • Hematopoietic cancers are malignant tumors affecting hematopoietic structures (structures pertaining to the formation of blood cells) and components of the immune system, including leukemias (related to leukocytes (white blood cells) and their precursors in the blood and bone marrow) arising from myeloid, lymphoid or erythroid lineages, and lymphomas (relates to lymphocytes).
  • Solid cancers include sarcomas, which are malignant cancers that originate from connective tissues such as muscle, cartilage, blood vessels, fibrous tissue, fat or bone.
  • Solid cancers also include carcinomas, which are malignant cancers arising from epithelial structures (including external epithelia (e.g., skin and linings of the gastrointestinal tract, lungs, and cervix), and internal epithelia that line various glands (e.g., breast, pancreas, thyroid).
  • Examples of cancers that are particularly susceptible to treatment by the methods of the invention include leukemia, and hepatocellular cancers, sarcoma, vascular endothelial cancers, breast cancers, central nervous system cancers (e.g.
  • astrocytoma astrocytoma, gliosarcoma, neuroblastoma, oligodendroglioma and glioblastoma
  • prostate cancers lung and bronchus cancers, larynx cancers, esophagus cancers, colon cancers, colorectal cancers, gastro- intestinal cancers, melanomas, ovarian and endometrial cancer, renal and bladder cancer, liver cancer, endocrine cancer (e.g. thyroid), and pancreatic cancer.
  • cancers include but are not limited to apudoma, choristoma, branchioma, malignant carcinoid syndrome, carcinoid heart disease, carcinoma (e.g., Walker, basal cell, basosquamous, Brown-Pearce, ductal, Ehrlich tumor, non-small cell lung, oat cell, papillary, bronchiolar, bronchogenic, squamous cell, and transitional cell), histiocytic disorders, leukemia (e.g., B cell, mixed cell, null cell, T cell, T-cell chronic, HTLV-ll-associated, lymphocytic acute, lymphocytic chronic, mast cell, and myeloid), histiocytosis malignant, Hodgkin disease, immunoproliferative small, non-Hodgkin lymphoma, plasmacytoma, reticuloendotheliosis, melanoma, chondroblastoma, chondroma,
  • carcinoma e.
  • the peptides of the invention can be used to modulate angiogenesis.
  • Diseases or disorders that are mediated by angiogenesis include tumors, ocular disorders, dermatological disorders, and malignant or metastatic conditions, inflammatory diseases, osteoporosis and other conditions mediated by accelerated bone resorption, restenosis, inappropriate platelet activation, recruitment, or aggregation, thrombosis, or a condition requiring tissue repair or wound healing.
  • ocular disorders that can be treated according to the present invention are eye diseases characterized by ocular neovascularization including, but not limited to, diabetic retinopathy (a major complication of diabetes), retinopathy of prematurity (this devastating eye condition, that frequently leads to chronic vision problems and carries a high risk of blindness, is a severe complication during the care of premature infants), neovascular glaucoma, retinoblastoma, retrolental fibroplasia, rubeosis, uveitis, macular degeneration, and corneal graft neovascularization.
  • Other eye inflammatory diseases, ocular tumors, and diseases associated with choroidal or iris neovascularization can also be treated according to the present invention.
  • the disorder associated with or mediated by angiogenesis is arteriosclerosis, arthritis, psoriasis or endometriosis.
  • the peptides of the present invention can also be used to treat inflammatory diseases including, but not limited to, arthritis, rheumatism, inflammatory bowel disease, and psoriasis.
  • the peptides of the invention can be used to treat epilepsy, memory, learning, neuropsychiatric, neurological, neuromuscular, and immunological disorders, schizophrenia, bipolar disorder, sleep apnea, neurodegeneration, smooth muscle disorders.
  • the peptides of the invention can be used as antibacterial peptides, antifungal peptides antimalarial peptides antiviral peptides, bradykinin-potentiating peptides, autoimmunity targeting potassium channels blockers, immuno-modulators, and analgesic peptides .
  • Other diseases and conditions that can be treated according to the present invention include benign tumors and preneoplastic conditions, myocardial angiogenesis, hemophilic joints, scleroderma, vascular adhesions, asthma and allergy, eczema and dermatitis, graft versus host disease, sepsis, adult respiratory distress syndrome, telangiectasia, and wound granulation.
  • the invention provides peptide RK (a 17 mer), which is the first scorpion peptide with dual disintegrin activity on a1 b1 and anb3 integrins.
  • the peptide of the invention may act by interacting with a1 b1 and/or anb3 integrins.
  • Disintegrins can be used to inhibit biological processes such as platelet aggregation (can be used as antithrombotic agents for use in thrombolytic therapy by enhancing and sustaining arterial thrombolysis in conjunction with recombinant tissue plasminogen activator), cell growth, adhesion, metastasis, and neovascularization.
  • the peptides of the invention can be used for the treatment and prevention of anb3 integrins associated diseases in a mammal, which include osteoporosis, bone tumor or cancer growth, angiogenesis-related tumor growth and metastasis, tumor metastasis in bone, malignancy-induced hypercalcemia, angiogenesis-related eye diseases, Paget's disease, rheumatic arthritis, ovariectomy-induced physiological change, inflammation, coagulation diseases, and osteoarthritis.
  • the angiogenesis-related eye diseases include age-related macular degeneration, diabetic retinopathy, corneal neovascularizing diseases, ischaemia-induced neovascularizing retinopathy, high myopia, and retinopathy of prematurity.
  • Disintegrings block adhesive functions and act as platelet aggregation inhibitors.
  • the peptides of the invention can also be use in the diagnosis of cardiovascular diseases and as therapeutic agents in arterial thrombosis.
  • homodimeric, heterodimeric, and monomeric disintegrins/peptides of the disclosure may be used to modulate the adhesion, motility, and invasiveness of integrin- expressing tumor cells.
  • such proteins can be used to treat patients by inhibiting or disrupting disease processes associated with a ligand binding to a1 b1 and anb3 integrins.
  • Homodimeric, heterodimeric, and monomeric peptide disintegrins described herein may be used in methods to decrease the motility of an cd b1 and/or anb3 integrins expressing cell, the method comprising cross-linking at least two a1 b1 and anb3 integrins on the integrin expressing cells thereby inhibiting the motility of said cells.
  • Such crosslinking is believed to disrupt FAK signaling and activates tyrosine phosphorylation of FAK and CAS.
  • the crosslinking is believed to induce an alteration in cell morphology, including changes of cytoskeletal or focal adhesion structures.
  • the a1 b1 and anb3 integrins integrin expressing cell is a tumor cell.
  • Homodimeric, heterodimeric, and monomeric disintegrins described herein may be used to inhibit the adhesion of integrin expressing cells to vitronectin by exposing the cells to the homodimeric and monomeric disintegrin.
  • the homodimeric and monomeric disintegrin is believed to inhibits adhesion by binding to an integrin, in particular a1 b1 and anb3 integrins.
  • Homodimeric, heterodimeric, and monomeric disintegrins described herein may be formulated as compositions for the treatment of thrombotic diseases in mammals, alone or in conjunction with one or more thrombolytic agents.
  • such compositions have utility in treating or preventing arterial, venous and microvascular thrombosis and thromboembolism.
  • Such compositions also have utility in treating stroke, transient ischemic attacks, arteriosclerosis, atherosclerosis, pulmonary embolism, aneurisms and angina.
  • such compositions have utility in preventing or treating myocardial infarctions.
  • Homodimeric, heterodimeric, and monomeric disintegrins described herein may be used to inhibit metastasis in melanoma, carcinoma and sarcoma patients.
  • Homodimeric, heterodimeric, and monomeric disintegrins may be used to prevent metastasis in breast cancer patients and other cancers.
  • Homodimeric, heterodimeric, and monomeric disintegrins described herein may be used to treat osteoporosis.
  • Compositions and methods for treatment of osteoporosis employing an amount of a homodimeric and monomeric disintegrin effective to inhibit bone resorption by osteoclasts may be used.
  • Homodimeric, heterodimeric, and monomeric disintegrins described herein may be used to promote wound healing.
  • Homodimeric, heterodimeric, and monomeric disintegrins may inhibit cell-cell and cell-extracellular matrix interactions (including interaction with fibronectin), thus promoting wound repair, including keloid formation.
  • Compositions containing homodimeric, heterodimeric, and monomeric disintegrins may be used to prevent adhesion formation when administered to a patient in need of such treatment.
  • the peptides of the invention are not able to alter cell adhesion through a2b1 , anb6, anb5, a5b1 and a6b4 integrins, but significantly reduce the adhesive activity of a1 b1 and anb3 integrins to extracellular matrix proteins.
  • the extracellular matrix protein comprises collagen.
  • the extracellular matrix protein comprises vitronectin, fibronectin, fibrinogen, osteopoietin, and/or bone sialoprotein.
  • the activity is reduced by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 90%, or 100%.
  • the present invention also relates to RK and RK1 , as well as peptide analogs of RK or RK1 , as peptides with novel functional properties such as greater ionic channels antagonist activity and/or target selectivity.
  • Molecules that recognize certain targets, specifically with high affinity to different receptors, are useful for many clinical (e.g., diagnostic and/or therapeutic) and biotechnological applications.
  • the peptides of the invention can be used to inhibit the proliferation and/or migration of a cancer cell, or cancer cell line.
  • cancer cell line or “cancer cells” may be used interchangeably and refers to cells isolated from a tumor, metastasis or abnormal growth derived from an animal or human. These cells typically, but not always, grow rapidly in culture when supplemented with appropriate growth factors, often fetal animal serum. This term also refers to normal cells transformed into cells that display typical features of cancer cells, i.e. they divide in cell culture under trophic support and/or form tumors when administered to animals.
  • Cancer cell lines may include, but are not limited to, human prostate cancer cells (e.g., PC-3, LNCaP, DU-145), human mammary epithelial cells (e.g. MCF-7, MCF-10A, MDA-MB-438, MDA- 231 , MDA-468, T47D, SkBr3), human neuronal cells (e.g., M17, SHSY5Y, H4, U87), human acute myeloid leukemia cells (e.g., THP-1), human bone cancer cells (e.g., Saos-2 cells), human melanoma cells (e.g., 721), human glioblastoma cells (e.g., A172), human head and neck carcinoma cells (e.g., A253), human skin epithelial cells (e.g., A431), human lung carcinoma epithelial cells (e.g., A- 549), human peripheral blood mononuclear cell lymphoma (e.g., BCP-1
  • the peptides of the invention can be administered alone or in combination with other drugs (e.g., as an adjuvant) or therapeutic agents.
  • additional therapeutic agent may comprise any active ingredients suitable for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • additional therapeutic agent may comprise any active ingredients suitable for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • numerous chemotherapeutics, cytotoxic and/or cytostatic agents, especially antineoplastic drugs, immunomodulatory agents are available for combination with the peptides of the invention.
  • the additional therapeutic agent is an anti-cancer agent, for example a microtubule disruptor, an antimetabolite, a topoisomerase inhibitor, a DNA intercalator, an alkylating agent, a hormonal therapy, a kinase inhibitor, a receptor antagonist, an activator of tumor cell apoptosis, or an antiangiogenic agent.
  • an anti-cancer agent for example a microtubule disruptor, an antimetabolite, a topoisomerase inhibitor, a DNA intercalator, an alkylating agent, a hormonal therapy, a kinase inhibitor, a receptor antagonist, an activator of tumor cell apoptosis, or an antiangiogenic agent.
  • adjunctive or combined administration includes simultaneous administration of the compounds in the same or different dosage form, or separate administration of the compounds (e.g., sequential administration).
  • the peptides of the invention can be simultaneously administered with a kinase inhibitor, wherein both the compound and the kinase inhibitor are formulated together.
  • the compound can be administered in combination with kinase inhibitor, wherein both the compound and the kinase inhibitor are formulated for separate administration and are administered concurrently or sequentially.
  • the kinase inhibitor can be administered first followed by (e.g., immediately followed by) the administration of the compound of the disclosure, or vice versa.
  • concurrent or sequential administration beneficially results in both the compound of the disclosure and kinase inhibitor being simultaneously present in treated patients.
  • the cytotoxic and/or cytostatic agents, and the anti-angiogenesis agents may be any agents known to inhibit angiogenesis, the growth and/or replication of and/or kill cells, and in particular the growth/replication of cancer and/or tumor cells. Numerous agents having cytotoxic and/or cytostatic properties are known in the literature.
  • Non-limiting examples of classes of cytotoxic and/or cytostatic agents include, by way of example and not limitation, radionuclides, alkylating agents, DNA cross-linking agents, DNA intercalating agents (e.g., groove binding agents such as minor groove binders), cell cycle modulators, apoptosis regulators, kinase inhibitors, protein synthesis inhibitors, mitochondria inhibitors, nuclear export inhibitors, topoisomerase I inhibitors, topoisomerase II inhibitors, RNA/DNA antimetabolites and antimitotic agents.
  • radionuclides e.g., alkylating agents, DNA cross-linking agents, DNA intercalating agents (e.g., groove binding agents such as minor groove binders), cell cycle modulators, apoptosis regulators, kinase inhibitors, protein synthesis inhibitors, mitochondria inhibitors, nuclear export inhibitors, topoisomerase I inhibitors, topoisomerase II inhibitors, RNA/DNA antimetabolites and antimitotic agents.
  • Alkylating Agents asaley (L-Leucine, N-[N-acetyl-4-[bis-(2-chloroethyl)amino]-
  • DL-phenylalanyl]-, ethylester AZQ (1 ,4-cyclohexadiene-1 ,4-dicarbamic acid, 2, 5-bis(1- aziridinyl)-3,6-dioxo-, diethyl ester); BCNU (N,N'-Bis(2-chloroethyl)-N-nitrosourea); busulfan (1 ,4- butanediol dimethanesulfonate); (carboxyphthalato)platinum; CBDCA (cis-(1 ,1- cyclobutanedicarboxylato)diammineplatinum(ll))); CCNU (N-(2-chloroethyl)-N'-cyclohexyl-N- nitrosourea); CHIP (iproplatin; NSC 256927); chlorambucil; chlorozotocin (2-[[[[(2-chloroethyl) nitro
  • DNA Alkylating-like Agents Cisplatin; Carboplatin; Nedaplatin; Oxaliplatin; Satraplatin; Triplatin tetranitrate; Procarbazine; altretamine; dacarbazine; mitozolomide; temozolomide.
  • Alkylating Antineoplastic Agents Carboquone; Carmustine; Chlornaphazine; Chlorozotocin; Duocarmycin; Evofosfamide; Fotemustine; Glufosfamide; Lomustine; Mannosulfan; Nimustine; Phenanthriplatin; Pipobroman; Ranimustine; Semustine; Streptozotocin; ThioTEPA; Treosulfan; Triaziquone; Triethylenemelamine; Triplatin tetranitrate.
  • DNA replication and repair inhibitors Altretamine; Bleomycin; dacarbazine; Dactinomycin; Mitobronitol; Mitomycin; Pingyangmycin; Plicamycin; Procarbazine; Temozolomide; ABT-888 (veliparib); olaparib; KU-59436; AZD-2281 ; AG-014699; BSI-201 ; BGP- 15; INO-1001 ; ONO-2231.
  • Cell Cycle Modulators Paclitaxel; Nab-Paclitaxel; Docetaxel; Vincristine; Vinblastine; ABT-348; AZD-1 152; MLN-8054; VX-680; Aurora A-specific kinase inhibitors; Aurora B-specific kinase inhibitors and pan-Aurora kinase inhibitors; AZD-5438; BMI-1040; BMS-032; BMS-387; CVT-2584; flavopyridol; GPC-286199; MCS-5A; PD0332991 ; PHA-690509; seliciclib (CYC-202, R-roscovitine); ZK-304709; AZD4877, ARRY-520; GSK923295A.
  • Apoptosis Regulators AT-101 ((-)gossypol); G3139 or oblimersen (Bcl-2- targeting antisense oligonucleotide); I PI-194; I PI-565; N-(4-(4-((4'-chloro(1 , T-biphenyl)-2- yl)methyl)piperazin-1-ylbenzoyl)-4-(((1 R)-3-(dimethylamino)-1-
  • Angiogenesis Inhibitors ABT-869; AEE-788; axitinib (AG-13736); AZD-2171 ; CP-547,632; IM-862; pegaptamib; sorafenib; BAY43-9006; pazopanib (GW-786034); vatalanib (PTK-787, ZK-222584); sunitinib; SU-11248; VEGF trap; vandetanib; ABT-165; ZD-6474; DLL4 inhibitors.
  • Proteasome Inhibitors Bortezomib; Carfilzomib; Epoxomicin; Ixazomib; Salinosporamide A.
  • Protein Synthesis Inhibitors Streptomycin; Dihydrostreptomycin; Neomycin; Framycetin; Paromomycin; Ribostamycin; Kanamycin; Amikacin; Arbekacin; Bekanamycin; Dibekacin; Tobramycin; Spectinomycin; Hygromycin B; Paromomycin; Gentamicin; Netilmicin; Sisomicin; lsepamicin;Verdamicin; Astromicin; Tetracycline; Doxycycline; Chlortetracycline; Clomocycline; Demeclocycline; Lymecycline; Meclocycline; Metacycline; Minocycline; Oxytetracycline; Penimepicycline; Rolitetracycline; Tetracycline; Glycylcyclines;Tigecycline; Oxazolidinone; Eperezolid; Linezolid; Posizolid; Radezolid; Ranbezolid; Sutezolid; Tedizolid; Pepti
  • Histone deacetylase inhibitors Vorinostat; Romidepsin; Chidamide; Panobinostat; Valproic acid; Belinostat; Mocetinostat; Abexinostat; Entinostat; SB939 (pracinostat); Resminostat; Givinostat; Quisinostat; thioureidobutyronitrile (KevetrinTM); CUDC- 10; CHR-2845 (tefinostat); CHR-3996; 4SC-202; CG200745; ACY-1215 (rocilinostat); ME-344; sulforaphane.
  • Topoisomerase I Inhibitors camptothecin; various camptothecin derivatives and analogs (for example, NSC 100880, NSC 603071 , NSC 107124, NSC 643833, NSC 629971 , NSC 295500, NSC 249910, NSC 606985, NSC 74028, NSC 176323, NSC 295501 , NSC 606172, NSC 606173, NSC 610458, NSC 618939, NSC 610457, NSC 610459, NSC 606499, NSC 610456, NSC 364830, and NSC 606497); morpholinisoxorubicin; SN-38.
  • camptothecin various camptothecin derivatives and analogs (for example, NSC 100880, NSC 603071 , NSC 107124, NSC 643833, NSC 629971 , NSC 295500, NSC 249910, NSC 606985, NSC 740
  • Topoisomerase II Inihibitors doxorubicin; amonafide (benzisoquinolinedione); m-AMSA (4'-(9-acridinylamino)-3'-methoxymethanesulfonanilide); anthrapyrazole derivative ((NSC 355644); etoposide (VP-16); pyrazoloacridine ((pyrazolo[3,4,5-kl]acridine-2(6H)- propanamine, 9-methoxy-N, N-dimethyl-5-nitro-, monomethanesulfonate); bisantrene hydrochloride; daunorubicin; deoxydoxorubicin; mitoxantrone; menogaril; N,N-dibenzyl daunomycin; oxanthrazole; rubidazone; teniposide.
  • DNA Intercalating Agents anthramycin; chicamycin A; tomaymycin; DC-81 ; sibiromycin; pyrrolobenzodiazepine derivative; SGD-1882 ((S)-2-(4-aminophenyl)-7-methoxy-8- (3-(((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-5,1 1 a-dihydro-1 H-benzo[e]pyrrolo[1 ,2- a][1 ,4]diazepin-8-yl)oxy)propoxy)-1 H-benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepin-5(11 aH)-one); SG2000 (SJG-136; (11 aS, 1 1 a'S)-8,8'-(propane-1 ,3-diylbis(oxy))bis(7-methoxy-2-methylene-2,3-di
  • RNA/DNA Antimetabolites L-alanosine; 5-azacytidine; 5-fluorouracil; acivicin; aminopterin derivative N-[2-chloro-5-[[(2, 4-diamino-5-methyl-6- quinazolinyl)methyl]amino]benzoyl] L-aspartic acid (NSC 132483); aminopterin derivative N-[4- [[(2, 4-diamino-5-ethyl-6-quinazolinyl)methyl]amino]benzoyl] L-aspartic acid; aminopterin derivative N-[2-chloro-4-[[(2, 4-diamino-6-pteridinyl)methyl] amino]benzoyl] L-aspartic acid monohydrate; antifolate PT523 ((N a -(4-amino-4-deoxypteroyl)-N Y -hemiphthaloyl-L-orni
  • DNA Antimetabolites 3-HP; 2'-deoxy-5-fluorouridine; 5-HP; a-TGDR (a-2’- deoxy-6-thioguanosine); aphidicolin glycinate; ara C (cytosine arabinoside); 5-aza-2'- deoxycytidine; b-TGDR (P-2’-deoxy-6-thioguanosine); cyclocytidine; guanazole; hydroxyurea; inosine glycodialdehyde; macbecin II; pyrazoloimidazole; thioguanine; thiopurine.
  • Mitochondria Inhibitors pancratistatin; phenpanstatin; rhodamine-123; edelfosine; d-alpha-tocopherol succinate; compound 11 ⁇ ; aspirin; ellipticine; berberine; cerulenin; GX015-070 (Obatoclax®; 1 H-lndole, 2-(2-((3,5-dimethyl-1 H-pyrrol-2-yl)methylene)-3- methoxy-2H-pyrrol-5-yl)-); celastrol (tripterine); metformin; Brilliant green; ME-344.
  • Antimitotic Agents allocolchicine; auristatins, such as MMAE (monomethyl auristatin E) and MMAF (monomethyl auristatin F); halichondrin B; cemadotin; colchicine; cholchicine derivative (N-benzoyl-deacetyl benzamide); dolastatin-10; dolastatin-15; maytansine; maytansinoids, such as DM1 (A/ 2 '-deacetyl-/ ⁇ / 2 '-(3-mercapto-1-oxopropyl)-maytansine); rhozoxin; paclitaxel; paclitaxel derivative ((2'-N-[3-(dimethylamino)propyl]glutaramate paclitaxel); docetaxel; thiocolchicine; trityl cysteine; vinblastine sulfate; vincristine sulfate.
  • Hormonal Therapies anastrozole; exemestane; arzoxifene; bicalutamide; cetrorelix; degarelix; deslorelin; trilostane; dexamethasone; flutamide; raloxifene; fadrozole; toremifene; fulvestrant; letrozole; formestane; glucocorticoids; doxercalciferol; sevelamer carbonate; lasofoxifene; leuprolide acetate; megesterol; mifepristone; nilutamide; tamoxifen citrate; abarelix; prednisone; finasteride; rilostane; buserelin; luteinizing hormone releasing hormone (LHRH); Histrelin; trilostane or modrastane; fosrelin; goserelin.
  • LHRH luteinizing hormone releasing hormone
  • an antiangiogenic agent, a cytotoxic agent, a cytostatic agent comprises at least one agent chosen from endostatin, angiostatin, VEGF inhibitors, cytotoxic agents, alkaloids, antimetabolites, cancer growth inhibitors, gene therapy therapeutics, cancer vaccines, interferons, monoclonal antibodies, radiotherapy, hormonal therapy, and other supportive therapy.
  • the chemotherapy drugs are chosen from amsacrine, bleomycin, busulfan, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, crisantaspase, cycolophosphamide, cytarabine, dacarbazine, dactinomycine, daunorubicin, docetaxel, doxorubicin, epirubicin, etoposide, fludarabine, fluorouracil, gemcitabine, gliadel implants, hydroxycarbamide, idarubicin, ifosfamide, irinotecan, leucovorin, liposomal doxorubicin, liposomal daunorubicin, lomustine, melphalan, mercaptopurine, mesna, methotrexate, mitomycin, mitoxantrone, oxaliplatin, paclitaxel,
  • the peptides when used in the treatment of an infectious disease, the peptides can be administered in combination with at least one anti-bacterial agent or at least one anti-viral agent.
  • the anti-bacterial agent can be any suitable antibiotic known in the art.
  • the anti-viral agent can be any vaccine of any suitable type that specifically targets a particular virus (e.g., live-attenuated vaccines, subunit vaccines, recombinant vector vaccines, and small molecule anti-viral therapies (e.g., viral replication inhibitors and nucleoside analogs).
  • the peptides of the disclosure can be used to inhibit cancer/tumor cell proliferation, migration, and/or adhesion in vivo.
  • the peptides of the disclosure are used to inhibit tumor cell proliferation.
  • the utility of the peptides of the present invention in inhibiting tumor cell proliferation in vivo can be illustrated, for example, by their activity in vitro in the in vitro tumor cell proliferation assays described below or known in the art.
  • the link between activity in tumor cell proliferation assays in vitro and anti-tumor activity in the clinical setting has been very well established in the art.
  • the therapeutic utility of taxol, taxotere, and topoisomerase inhibitors were first demonstrated with the use of in vitro tumor proliferation assays.
  • in vivo xenograft tumor models simulate biological activity observed in humans by grafting relevant and well characterized human primary tumors or tumor cell lines into immune-deficient nude mice.
  • Studies on treatment of tumor xenograft mice with anti-cancer reagents have provided valuable information regarding in vivo efficacy of the tested reagents.
  • the peptides of the invention are used in a method of inhibiting tumor cell proliferation by contacting the tumor cell with an effective amount of the peptide.
  • the tumor cells are glioblastoma cells.
  • the tumor cells are melanoma cells.
  • the tumor cells are cells of the tumors listed elsewhere in the specification.
  • the inhibition is in vitro.
  • the inhibition is in vivo.
  • the peptide can be used alone or in combination with other drugs or therapeutic agents, many of which are exemplified elsewhere in the specification.
  • the peptides of the invention are used in a method of inhibiting tumor cell migration by contacting the cancer cells with an effective amount of the peptide.
  • the tumor cells are glioblastoma cells.
  • the tumor cells are melanoma cells.
  • the tumor cells are cells of the tumors listed elsewhere in the specification.
  • the inhibition is in vitro.
  • the inhibition is in vivo.
  • the peptide can be used alone or in combination with other drugs or therapeutic agents, many of which are exemplified elsewhere in the specification.
  • the peptides of the invention are used in a method of inhibiting tumor cell adhesion by contacting the tumor cell with an effective amount of the peptide.
  • the tumor cells are glioblastoma cells.
  • the tumor cells are melanoma cells.
  • the tumor cells are cells of the tumors listed elsewhere in the specification.
  • the inhibition is in vitro.
  • the inhibition is in vivo.
  • the peptide can be used alone or in combination with other drugs or therapeutic agents, many of which are exemplified elsewhere in the specification.
  • the peptides of the invention can be used to inhibit non cancer cell growth/proliferation, migration, and/or adhesion.
  • the cells are endothelial cells.
  • the cells are
  • the dose of the peptide administered to achieve an effective amount may vary depending upon the disorder being treated or the type of tumor cell to be inhibited.
  • a dosage to be achieved in vivo would be equivalent to an in vitro level of greater than 10- 12 M, or 10 11 M, or 10 10 M, or 10 9 M, or 10 8 M, or 10 7 M, or 10 6 M, or 10 5 M.
  • a dosage to be achieved in vivo may be equivalent to an in vitro level of 10 12 M to 10 5 M, or 10 11 M to 10 6 M, or 10 10 M to 10 7 M, or 10 9 M to 10 7 M or ranges therein.
  • the dosage used may be equivalent to an in vitro level of about 1-10000 ngml 1 , or about 10-5000 ngml 1 , or about 100-1000 ngml 1 .
  • the dosage to be achieved in vivo may be equivalent to an in vitro level of 1 mM, 2 pM, 3 pM, 4 pM, 5 pM, 6 pM, 7 pM, 1 pM, 8 pM, 9 pM, 10 pM, 20 pM, 30 pM, 40 pM, 50 pM, 70 pM, 80 pM, 90 pM, or 100 pM.
  • the equivalence between in vitro and in vivo levels is calculated or estimated by methods known to one of ordinary skill in the art.
  • the dosage achieved is a blood/plasma level of at least 1 pM, 2 pM, 3 pM, 4 pM, 5 pM, 6 pM, 7 pM, 1 pM, 8 pM, 9 pM, 10 pM, 20 pM, 30 pM, 40 pM, 50 pM, 70 pM, 80 pM, 90 pM, or 100 pM.
  • the blood level is 2-5 pM (e.g., from 2 to 5), 5-10 pM, 10-100 pM, 100-1000 pM, 50-60 pM, 60-70 pM, 70-80 pM, 80-90 pM, 90-95 pM, 95-99 pM, 99-100 pM.
  • the blood level is 1-2 pM, 2-3 pM, 3-4 pM, 4-5 pM, 5-6 pM, 6-7 pM, 7-8 pM, 8-9 pM, 9-10 pM, 10-20 pM, and all the other ranges between the values set in this paragraph.
  • this level or plasma Tmax is achieved after 1 minute, no more than 2 minutes, no more than 3 minutes, no more than 4 minutes, no more than 5 minutes, no more than 8 minutes, no more than 10 minutes, no more than 12 minutes, no more than 15 minutes, no more than 20 minutes, no more than 30 minutes, no more than 35 minutes, no more than 40 minutes, no more than 45 minutes, no more than 50 minutes, no more than 55 minutes, is between 2 minutes and 30 minutes, or is no more than 60 minutes after one administration.
  • this level or Tmax is achieved after 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 1 1 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 22 hours, 23 hours, or 24 hours.
  • this level or other desired level or plasma Tmax is achieved within 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 22 days, 23 days, or 24 days.
  • a dose may also comprise from about 1 microgram/kg body weight, about 5 microgram/kg body weight, about 10 microgram/kg body weight, about 50 microgram/kg body weight, about 100 microgram/kg body weight, about 200 microgram/kg body weight, about 350 microgram/kg body weight, about 500 microgram/kg body weight, about 1 milligram/kg body weight, about 5 milligram/kg body weight, about 10 milligram/kg body weight, about 50 milligram/kg body weight, about 100 milligram/kg body weight, about 200 milligram/kg body weight, about 350 milligram/kg body weight, about 500 milligram/kg body weight, to about 1000 mg/kg body weight or more per administration, and any range derivable therein.
  • a range of about 5 mg/kg body weight to about 100 mg/kg body weight, about 5 microgram/kg body weight to about 500 milligram/kg body weight, etc. can be administered, based on the numbers described above.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 5.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the peptide molecule).
  • An initial higher loading dose, followed by one or more lower doses may be administered.
  • the dosages may be administered daily, every other day, continuously over one day or more.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • the dosage may comprise from about 0.00001 to 500 mg/kg/day, or from about 0.0001 to 300 mg/kg/day, or from about 0.003 to 100 mg/kg/day, or from about 0.03 to 30 mg/kg/day, or from about 0.1 mg/kg/day to 10 mg/kg/day, or from about 0.3 mg/kg/day to 3 mg/kg/day.
  • the dosage is 20 mg per peptide per day. In another embodiment, the dosage is 10 mg/peptide/day. In another embodiment, the dosage is 30 mg/peptide/day. In another embodiment, the dosage is 40 mg/peptide/day. In another embodiment, the dosage is 60 mg/peptide/day.
  • the dosage is 80 mg/peptide/day. In another embodiment, the dosage is 100 mg/peptide/day. In another embodiment, the dosage is 150 mg/peptide/day. In another embodiment, the dosage is 200 mg/peptide/day. In another embodiment, the dosage is 300 mg/peptide/day. In another embodiment, the dosage is 400 mg/peptide/day. In another embodiment, the dosage is 600 mg/peptide/day. In another embodiment, the dosage is 800 mg/peptide/day. In another embodiment, the dosage is 1000 mg/peptide/day.
  • the dosage is 20 pg per peptide per day. In another embodiment, the dosage is 10 pg/peptide/day. In another embodiment, the dosage is 30 pg/peptide/day. In another embodiment, the dosage is 40 pg/peptide/day. In another embodiment, the dosage is 60 pg/peptide/day. In another embodiment, the dosage is 80 pg/peptide/day. In another embodiment, the dosage is 100 pg/peptide/day. In another embodiment, the dosage is 150 pg/peptide/day. In another embodiment, the dosage is 200 pg/peptide/day. In another embodiment, the dosage is 300 pg/peptide/day.
  • the dosage is 400 pg/peptide/day. In another embodiment, the dosage is 600 pg/peptide/day. In another embodiment, the dosage is 800 pg/peptide/day. In another embodiment, the dosage is 1000 pg/peptide/day. In another embodiment, the dosage is 1500 pg/peptide/day. In another embodiment, the dosage is 2000 pg/peptide/day.
  • the dosage is 10 pg/peptide/dose. In another embodiment, the dosage is 30 pg/peptide/dose. In another embodiment, the dosage is 40 pg/peptide/dose. In another embodiment, the dosage is 60 pg/peptide/dose. In another embodiment, the dosage is 80 pg/peptide/dose. In another embodiment, the dosage is 100 pg/peptide/dose. In another embodiment, the dosage is 150 pg/peptide/dose. In another embodiment, the dosage is 200 pg/peptide/dose. In another embodiment, the dosage is 300 pg/peptide/dose. In another embodiment, the dosage is 400 pg/peptide/dose. In another embodiment, the dosage is 600 pg/peptide/dose.
  • the dosage is 800 pg/peptide/dose. In another embodiment, the dosage is 1000 pg/peptide/dose. In another embodiment, the dosage is 1500 pg/peptide/dose. In another embodiment, the dosage is 2000 pg/peptide/dose.
  • the dosage is 10-20 pg/peptide/dose. In another embodiment, the dosage is 20-30 pg/peptide/dose. In another embodiment, the dosage is 20-40 pg/peptide/dose. In another embodiment, the dosage is 30-60 pg/peptide/dose. In another embodiment, the dosage is 40-80 pg/peptide/dose. In another embodiment, the dosage is 50-100 pg/peptide/dose. In another embodiment, the dosage is 50-150 pg/peptide/dose. In another embodiment, the dosage is 100-200 pg/peptide/dose. In another embodiment, the dosage is 200-300 pg/peptide/dose. In another embodiment, the dosage is 300-400 pg/peptide/dose.
  • the dosage is 400-600 pg/peptide/dose. In another embodiment, the dosage is 500- 800 pg/peptide/dose. In another embodiment, the dosage is 800-1000 pg/peptide/dose. In another embodiment, the dosage is 1000-1500 pg/peptide/dose. In another embodiment, the dosage is 1500-2000 pg/peptide/dose.
  • the dosage is 10 mg/peptide/dose. In another embodiment, the dosage is 30 mg/peptide/dose. In another embodiment, the dosage is 40 mg/peptide/dose. In another embodiment, the dosage is 60 mg/peptide/dose. In another embodiment, the dosage is 80 mg/peptide/dose. In another embodiment, the dosage is 100 mg/peptide/dose. In another embodiment, the dosage is 150 mg/peptide/dose. In another embodiment, the dosage is 200 mg/peptide/dose. In another embodiment, the dosage is 300 mg/peptide/dose. In another embodiment, the dosage is 400 mg/peptide/dose. In another embodiment, the dosage is 600 mg/peptide/dose. In another embodiment, the dosage is 800 mg/peptide/dose. In another embodiment, the dosage is 1000 mg/peptide/dose.
  • the dosage is 10-20 mg/peptide/dose. In another embodiment, the dosage is 20-30 mg/peptide/dose. In another embodiment, the dosage is 20-40 mg/peptide/dose. In another embodiment, the dosage is 30-60 mg/peptide/dose. In another embodiment, the dosage is 40-80 mg/peptide/dose. In another embodiment, the dosage is 50- 100 mg/peptide/dose. In another embodiment, the dosage is 50-150 mg/peptide/dose. In another embodiment, the dosage is 100-200 mg/peptide/dose. In another embodiment, the dosage is 200- 300 mg/peptide/dose. In another embodiment, the dosage is 300-400 mg/peptide/dose. In another embodiment, the dosage is 400-600 mg/peptide/dose. In another embodiment, the dosage is 500-800 mg/peptide/dose. In another embodiment, the dosage is 800-1000 mg/peptide/dose.
  • Initial dosages can also be estimated from in vivo data, e.g., animal models, using techniques that are well known in the art. One having ordinary skill in the art could readily optimize administration to humans based on animal data.
  • the dosage amount and interval may be adjusted individually to provide plasma levels of the peptides that are sufficient to maintain therapeutic effect.
  • Usual patient dosages for administration by injection range from about 0.1 to 50 mg/kg/day, typically from about 0.5 to 1 mg/kg/day.
  • Therapeutically effective plasma levels may be achieved by administering multiple doses each day. Levels in plasma may be measured, for example, by HPLC.
  • the plasma concentration of the peptides is about 0.05 to 0.9 ng/ml after about 15 minutes after application, or about 0.1 to 1.4 ng/ml after about 30 minutes, or about 0.1 to 1.6 ng/ml after about 1 hour, or about 0.1 to 1.4 ng/ml after about 1.5 hours, or about 0.1 to 1.3 ng/ml after about 2 hours, or less than about 0.7 ng/ml after 5 hours, or less than about 0.2 ng/ml after 10 hours.
  • the plasma concentration of the peptides is about 1-20 mM after about 15 minutes after application, or about 1-20 mM after about 30 minutes, or about 1-20 pM after about 1 hour, or about 1-20 pM after about 1.5 hours, or about 1-20 pM after about 2 hours, or less than about 1-20 pM after 5 hours, or less than about 1-20 pM after 10 hours.
  • the dose may be administered all at once, such as with an oral formulation in a capsule, or slowly over a period of time, such as with an intravenous administration.
  • the administering period can be a matter of minutes, such as about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120 or more minutes, or a period of hours, such as about 0.5, 1 , 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5 or more hours.
  • the administration of the dose may be interrupted, such as where the dose is administered via intravenous infusion and the dose is divided into two or more infusion bags. Under such circumstances, the administration of the dose may be interrupted while the infusion bags are changed.
  • the peptides of the invention can also be used for tumor visualization by single-photon emission computed tomography (SPECT), which is based on g-rays, and the peptides of the invention are here linked to radionuclides such as 99mTc, 177Lu, 1231 and 1 1 11n.
  • SPECT single-photon emission computed tomography
  • the positron-emitting radioisotopes 68Ga, 1241 or 89Zr are used for positron emission tomography (PET) purposes.
  • the peptides of the invention can be conjugated with bifunctional chelating agents which possess a metal binding moiety for sequestration of the metallic radionuclide and are generally DPTA (acyclic)-, DOTA (macrocyclic), or NOTA-based.
  • the chelating agents are equipped with a chemically reactive functional group for attachment to the peptides of the invention, which can occur in several ways well known to one of ordinary skill in the art.
  • the peptides of the invention can also be conjugated to near-infrared fluorophores such as IRDye800CW to perform optical tumor imaging, a technique that is cost-effective, flexible, sensitive and fast.
  • suitable labels include an affinity label (e.g., biotin, avidin), a spin label, an enzyme, a fluorescent group, or a chemiluminescent group.
  • affinity label e.g., biotin, avidin
  • spin label e.g., an enzyme, a fluorescent group, or a chemiluminescent group.
  • an article of manufacture containing materials that comprise the peptides of the invention and are useful for the treatment, prevention and/or diagnosis of the disorders described above comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is a peptide of the invention.
  • the label or package insert indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises peptide of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution.
  • BWFI bacteriostatic water for injection
  • Ringer's solution such as phosphate
  • the molecular docking model between peptide RK and integrin described in this disclosure is also an embodiment of the invention.
  • the molecular docking model between peptide RK and integrins is used to design or select/screen for organic drugs or other peptides that can be used in any of the other methods described herein (e.g., methods of treatment).
  • Scorpion venoms is a rich soup of peptide compounds, known for their biological properties. Many of them were found to act on different cancer hallmarks [21] Few of the scorpion peptides are able to act on cell proliferation, migration and angiogenesis.
  • Margatoxin, Iberiotoxin and Charybdotoxin were reported to inhibit cell proliferation while TM601 , a synthetic analogue of Chlorotoxin is capable to inhibit angiogenesis [22]
  • These scorpion peptides have the common feature of presenting multiple disulfide bridges (at least 3) and a number of composing residues up to 30 amino acids [23]
  • the inventors describe a new short peptide, RK1 from Buthus occitanus tunetanus with 1467.67 KDa consisting of 14 amino acids.
  • the peptide does not belong to any of the characterized families. It is also particular in containig one disulfide bridge formed between the C3 and the C12 amino acid.
  • Toxins and peptides derived from scorpion venoms generally have direct effects on the ion channels (K+, Na+, Ca2+%)
  • the few peptides that have an anti-tumor effect are generally cytotoxic.
  • two peptides named neopladine 1 and neopladine 2, isolated from Tityus discrepans scorpion venom were reported to be effective in inducing apoptosis and necrosis of SKBR3 breast cancer cells
  • the inventors assessed its toxicity on Tumor cells U87 and IGR39. After 48 h treatment, RK1 has no cytotoxic effect up to a concentration of 8 mM. Therefore for the further tests, the inventors operated under cytotoxic concentrations to study the specific effect of RK1.
  • RK1 has an anti-tumor effect by inhibiting the proliferation and migration of gliomatous and melanomatous cells on the other hand, RK1 strongly inhibits neoangiogenesis.
  • Our peptide has no toxcity, in vitro and in vivo, which can decrease the harmful toxic effects of chemotherapy and radiotherapy. Small size of RK1 hence the possibility of generating less immunogenicity and its capacity to form a disulfide bond which might increase its stability in physiological conditions.
  • RK1 is a very short peptide (14 aa) that has significant anti-tumor activity and can be classified as one of the few anti-cancer scorpion venom peptides that do not have cytotoxic activity.
  • the peptide was analyzed by MALDI-TOF mass spectrometer (Perspective Biosystems, Inc., Framingham, MA). The sample was dissolved in CH3CN/H20 (30/70) with 0.3% trifluoroacetic acid to obtain a concentration of 1-10 pmol. pL-1.
  • the matrix was prepared as follows: alpha-cyanohydroxycinnamic acid was dissolved in 50% CH3CN in 0.3% trifluoroacetic acid/H20 to obtain a saturated solution. Peptide solution was placed on the sample plate, and 0.5 pL of the matrix solution was added. This mixture was allowed to dry. Mass spectra were recorded in linear mode, calibrated with suitable standards and were analyzed by GRAMS/386 software.
  • the peptide stock was diluted in distilled water and kept at -20°C, and quantified by a Nano DropND-1000 V3.5.2 Spectrophotometer at 280 nm.
  • the reduced peptide was solubilized in 0.2 M Tris/HCI, pH 8, at 5 mM and stirred under air to allow folding (48 h, 25 °C). 2.6.
  • Cell viability test and in vivo toxicity test
  • MTT 3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide
  • MTT solution was added to the culture medium 4 h before the end of treatment.
  • Cells were incubated with RK1 for 72 h, then fixed with 1 % glutaraldehyde, stained with a solution of 0.1 % crystal violet, and lysed with 1 % SDS. Absorbance was then measured at 560 nm. A control was used in the same conditions but without RK1.
  • RK1 is a short peptide composed of 14 amino acid residues containing two cystein residues (Fig. 1 E).
  • RK1 Before testing the biological effect of RK1 , the inventors assessed the peptide’s toxicity on Tumor cells U87 and IGR39. After 48 h treatment, RK1 has no cytotoxic effect up to a concentration of 8 mM. Therefore, for the further tests, the inventors operated under cytotoxic concentrations to study the specific effect of RK1 Evaluation of the toxicity of RK1 and RK1 s revealed that both peptides do not exhibit any neurotoxicity up to a 2pg/20g mice or 100 pg/kg body weight as determined by intracerebroventricular injection. RK1s was tested on the viability of glioblastoma (U87) and melanoma (IGR-39) cell lines using the MTT assay. At 100 pM concentration, RK1 s does not affect the viability of the two cell lines after 24h, 48h or 72h incubation time (Fig. 2A).
  • RK1 was shown to inhibit ex-vivo vascular growth by 50 % at 4 mM dose and by 60 % at 8 mM.
  • RK1 has an anti-tumor effect by inhibiting the proliferation and migration of gliomatous and melanomatous cells.
  • RK1 strongly inhibits neoangiogenesis, which means that the peptide is capable of acting on endothelial cells.
  • Our peptide has no toxicity, in vitro and in vivo, which can decrease the harmful toxic effects of chemotherapy and radiotherapy.
  • Small size of RK1 hence the possibility of generating less immunogenicity and its capacity to form a disulfide bond which might increase its stability in physiological conditions.
  • RK1 is a very short peptide (14 aa) that has significant anti-tumor activity and can be classified as one of the few anti-cancer scorpion venom peptides that do not have cytotoxic activity.
  • RK1 is the first very short peptide from Buthus occitanus tunetanus that, unexpectedly, inhibits three key aspects of cancer development, namely tumor cell migration, proliferation, and angiogenesis.
  • RK A novel peptide, named RK was also isolated and characterized from the Buthus occitanus tunetanus scorpion venom targeting the cell adhesion activity by acting on integrins. To our knowledge, this is the first disintegrin-like peptide purified from a scorpion venom. Interestingly, RK peptide is unique among the variety of known peptides purified from scorpion venoms in terms of its structure. It is a small peptide of 17 amino acids (IDCGTVMIPSECDPKSS; SEQ ID NO:1) containing a single disulfide bond. RK may represent the first member of a new group of scorpion peptides.
  • RK did not show any toxicity a dose of 100 pg per kg mice body weight. It is also non-cytotoxic until 100 pM concentration. RK causes the detachment of three cell lines on different ECM proteins. The ineffectiveness of the RK on the non-specific substratum poly-L-lysine coated plates suggests that the inhibition effect of tumoral cells adhesion, by RK peptide, is integrin dependent. High levels of anb3, a5b1 and a3b1 are expressed in U87 [32], while IGR-39 highly expresses a2b1 and anb5 integrins and modestly expresses anb3 [33]
  • lebectin a C-type lectin.
  • lebectin is only active on fibronectin and fibrinogen matrix types but not on collagen I and IV [35]
  • RK is the first biomolecule demonstrating the dual activity on a1 b1 , which binds to collagen IV, and anb3 which binds to all ECM macromolecules presenting the RGD motif (vitronectin, fibronectin, fibrinogen, osteopentin, and bone sialoprotein) [39]
  • RGD motif vitronectin, fibronectin, fibrinogen, osteopentin, and bone sialoprotein
  • Fg fibrinogen
  • Vn vitronectin
  • Fn fibronectin
  • a1 K562 K562 cells transfected with specific ai subunits of integrins. Ref, references.
  • the free energy landscape shows a dense populated global minimum with a large bottom.
  • a1 b1 integrin (Fig. 11).
  • the latter has the property to undergo a structural rearrangement upon the binding of collagen IV notably for the C loop as concluded by the crystal and NMR solution of the integrin [45-46-47-48]
  • the binding to a1 b1 could then be ensured favorably enough even if the peptide structure shifts significantly from the global minimum conformation which corresponds to a low drifts in the energy of the system.
  • This property also, imply that the RK peptide must preserve a minimum specific set of contacts with high structural stability, in order to interact with the integrin.
  • the RMSF profile suggests that the central segment of the peptide, limited by the positions of the two cystein residues, are stable during the molecular dynamics simulation.
  • the segment V6, M7 and I8 of the peptide might be critical to ensure the minimal contacts required for the interaction with a1 b1 regarding also the results from the docking (Fig. 11).
  • the local conformation of the segment corresponding to a turn-like structure that remains stable during the 300 ns of molecular dynamics simulation. It allows to the M7 side chain to preserve a high level of solvent exposure but also to minimize the structural constraints with other atoms of the peptide.
  • RK was substantially purified from the Buthus occitanus tunetanus scorpion venom by a first stage of gel filtration on a Sephadex G-50 column chromatography as previously described
  • the toxic fraction (BotG50) obtained from this separation was purified by high performance liquid chromatography (HPLC) using a semi preparative C8 column.
  • the fraction eluting at 19-22 min (Fig. 5A)
  • the component eluted at 33.5 min (Fig. 5B) was homogeneous, as indicated by mass spectrometry analysis and amino acid sequencing. It is designated RK.
  • An analytical HPLC run of RK showed a single symmetric peak (Fig. 5C).
  • RKs has the highest activity on PC12 cell adhesion to collagen IV. Inhibition of adhesion occurs in a dose-dependent manner with an IC 5 o value of 4.84 pM. RKs blocked notably the adhesion of human glioblastoma cells U87 cells to fibrinogen and fibronectin with IC 5 o values of 10 pM and 10.33 pM respectively (Fig. 8A). However the inhibitory effect of RKs on IGR39 cells on fibrinogen and fibronectin does not exceed 40% (Fig. 8B).
  • RKs activity is selective to a1 b1 and anb3 integrins
  • RKs was not able to alter cell adhesion through a2b1 , anb6, anb5, a5b1 and a6b4 integrins, but significantly reduced the adhesive function of a1 b1 and anb3 integrins.
  • the second one is the segment flanked by the two cystein residues (C3GTVMIPSEC12) which harbors a one-turn a helix consisting of residues 9-12.
  • the third sub- segment is the C-terminal end corresponding to the DPKSS (aa 1 1-16 of SEQ ID NO: 1) sequence (Fig. 10A). Except for the two cystein residues, all the amino acids of RK are solvent exposed.
  • the Accessible surface area calculated for each residue using GETAREA shows a minimum of exposure for E11 (53%) and a maximum (100%) for T5, P8, S16 and S17.
  • the RMSD Root Mean Square Deviation
  • the first phase is very short consisting of an equilibration ending at the 8 th nanosecond of the simulation.
  • the second plateau phase lasts between the 8 th and the 52 nd nanosecond where the RMSD fluctuate approximately around an average value of 3.8 A relative to the starting structure.
  • the peptide preserves the overall conformation of the initial structure, mainly the one turn a helix.
  • Residues 12-15 are the least flexible with RMSF (Root Mean Square Fluctuation) values ranging between 2.7 and 4.8 A.
  • the inventors established the energy landscape of the peptide based on the radius of gyration and the RMSD values as reaction coordinates.
  • the Fig. 10C shows that the peptide describes mainly two low energy wells. The first one is the largest in which the conformation with the lowest energy preserves the one turn a helix. The well is highly populated and corresponds to a global minima.
  • the second well is not as much populated as the first well in which the conformations present low energy values and corresponds to local minima.
  • the two wells are separated by two other metastable local minima.
  • the well representing the global minima of the free energy landscape of RK structure served to construct the docking ensemble of the ligand.
  • the clustering of the RK structure models leads to an ensemble of 13 structures.
  • the receptor structure used in this study is the domain I of a1 integrin (Dial). It consists of an inserted segment near the N-terminus found only in a1 , a2 and a12 integrin sequences. For instance, in contrast to a v s the MIDAS (metal-ion-dependent adhesion site) is harbored in Dial for the a1 integrin by which it interacts with the extracellular collagen molecules. There are eleven structures in NMR ensemble of Dial (PDB code: 2M32).
  • the inventors added three other conformations resulting from the output of ENCoM server after submitting the crystal unbound structure of Dial (PDB code: 1 PT6). As a result, a total number of 14 receptor conformers were processed in this study. Consequently, the inventors affected a number of 182 (13 for the ligand vs. 14 for the receptor) docking and a total of 364000 complexes were evaluated using the ZRANK scoring function.
  • the residue K 15 of RK is able to establish a stable salt bridge with E 285 of Dial (Fig. 11 B), with an average distance of 2.7 A during the 10 ns of molecular dynamics.
  • K 15 also establishes a stable hydrogen bond with D 13 with an occupancy of 57% during the 10 ns simulation time (Fig. 11 C). It seems that this bond, reduces the rotameric flexibility and restraints K 15 side chain to a close distance to E 285 side chain which might contribute in a more stabilization of the salt bridge formation between the two amino acids.
  • K 15 has the role of a bridging amino acid between residues D 13 of RK and E 285 of Dial
  • the only interaction of this type which is reasonably stable (20% occupancy of the total snapshots from the trajectory) is made between C 3 of RK and G 3I8 residue from the C loop segment of Dial .
  • PC12 neuronal cell line expresses two beta 1 -class integrins-alpha 1 beta 1 and alpha 3 beta 1 -that recognize different neurite outgrowth-promoting domains in laminin. Neuron. 1990, 5, 651-62.
  • Leberagin-C A disintegrin-like/cysteine-rich protein from Macrovipera lebetina transmediterranea venom, inhibits alphavbeta3 integrin-mediated cell adhesion. Matrix Biol. 2010, 29, 1 17-126.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention is related to novel scorpion venoms and methods of using the same in various treatment applications, including, but not limited to inhibiting cell migration, cell proliferation, and angiogenesis, as well as to treat cancer, pathological angiogenesis, and integrin-mediated diseases. The invention encompasses a short 14 amino acid peptide (calledRK1), purified from the venom of Buthus occitanus tunetanus, with the particular capabilities,among different other scorpion peptides, to inhibit cell proliferation, migration and angiogenesis of U87 (Glioblastoma) and IGR39 (Melanoma). Moreover, RK1 is a first peptide derived from scorpion venom exhibiting a potential anti-tumoral activity with no manifest toxicity.

Description

ANTI-CANCER AND DISINTEGRIN SCORPION VENOMS
FIELD
[1 ] The invention is related to novel scorpion venoms and methods of using the same to inhibit cell migration, cell proliferation, and angiogenesis, as well as to treat cancer, pathological angiogenesis, and integrin-mediated diseases.
BACKGROUND
Description of Related Art
[2] Curing cancer has undoubtedly become one of the largest challenges in the medical world. According to the World Health Organization (WHO), cancer killed nearly 8.8 million people in 2015, accounting more than 13% of the world's deaths that year. Despite the billions of dollars invested in research, the current treatments (chemotherapy and radiotherapy) are insufficient. According to the WHO, doctors manage to treat 7.3% more cancers than in 1950, while by 2030, twice as many people would die from this disease.
[3] Cancer cells share characteristics that make it possible to identify them, despite the great diversity of cancers. They are a genetically unstable type cell, able to explore functions of the entire genome and to use any migration or proliferative advantage to select and transmit it to its offspring. The tumor masses oscillate between proliferation and apoptosis. The development of new blood vessels in the tumor also facilitates metastasis formation, Cell migration and proliferation in response to pro-angiogenic factors. Cell migration, proliferation and angiogenesis are key phenomena in tumor growth. They give rise to numerous cell interactions with each other and with their environment, leading to non-trivial collective effects.
[4] Angiogenesis is required to maintain the continuous growth of malignant cells by providing with nutrients and oxygen and allow them to discard the wastes. The ability of tumor cells to induce the neovascularization is a significant step in tumorigenesis. The vast majority of tumor cells must initiate tumor angiogenesis by producing proangiogenic diffusion-promoting tumor substances to form detectable tumors. For that, blocking the action of proangiogenic substances can stop tumor growth. For more than twenty years, Cubans have been treating cancer patients with blue scorpion venom. The results are not always so great, but thousands of patients say their pain has been relieved, their muscle strength is increased, and their energy is boosted by taking this medication. In fact, many of the scorpion venom peptides are active on different tumoral metastasis stages. For example, Charybdotoxin (ChTx) and Iberiotoxin are capable to inhibit the proliferation and migration of glioma and melanoma cells, respectively [1 , 2] The few peptides from scorpion venom that have an anti-tumor effect are generally cytotoxic.
[5] In the last decades, it was demonstrated that different types of integrins, are highly expressed by many types of tumors and play a role in tumor angiogenesis, growth, and mestastasis. As a result, these proteins emerged as interesting biomarkers to detect cancers and as potential pharmacological targets to treat them.
[6] Integrins are transmembrane heterodimeric proteins of non-covalently associated a and b subunits, are implicated in all the processes of carcinogenesis [3] The association between their a and b subunits forms adhesion receptors which binds to the extracellular matrix and provide critical adhesive and signalling functions [4] Integrins are capable to affect cellular functions [5] such as cytoskeleton organization, transduction of intracellular signals [6], cell differentiation, growth, and apoptosis [7-8] Different subtypes of integrins have been characterized on the basis of their interactions with different motifs of the extracellular matrix implicated in cell adhesion [9] These motifs coordinate the biological responses between endothelial cells, tumor cells and the extracellular matrix [10] In fact, these integrins were first identified on the basis of their ability to recognize the RGD amino acid sequence [11-12], and are implicated in cell proliferation, invasion and viability. Their ligand binding function is also dependent on the presence of the metal ion (Mg2+, Mn2+ and Ca2+) [13] Integrins bind to collagen by using their al Domain, a1 b1 and a2b1 represent the most known collagen receptors which are members of integrin family and are structurally very similar [14] Their ligand binds to a Mg2+ ion in the Metal Ion-Dependent Adhesion Site (MIDAS) [15] a1 b1 integrin is the principal collagen IV receptor [16] but its contribution in tumor formation and progression is poorly defined compared to anb3 and a5b1. The critical role of a1 b1 integrin in tumorigenicity was demonstrated with works elaborated in colon cancer cells that associate with talin and paxillin, resulting in promotion of cancer cell invasion [17]
[7] The ability of multiple polypeptides to inhibit the integrin activity was previously discovered for different molecules purified mainly from snake venoms called disintegrins. Disintegrins were proven to have antitumour effects involving angiogenesis and cancer metastatic dissemination. The functional classification of disintegrins depends on their ability to interact with specific integrins [18], which is determined by the presence of a particular integrin-binding motif localized in the hairpin loop, unless they are present in the same fold. Functionally, disintegrins can be divided into three classes containing RGD, MLD, and R/KTS motifs have been identified [19] RGD-disintegrins block anb3, anb1 , a5b1 and a8b1 integrins. MLD inhibits the physiological functions of a3b1 , a6b1 , anbq, a7b1 , a4b1 , a4b7 and a9b1 integrins [20] R/KTS-disintegrins are a potent and selective inhibitors of a1 b1 integrin [21]
[8] Scorpion peptides are well known for their pharmaceutical potential on different targets. These targets are mainly ion channels, which were found to be highly expressed in many diseases including cancer, auto-immune pathologies and Alzheimer. However, to date, disintegrin activity has only been found in snake venom molecules, not scorpion. Scorpion toxins have been the subject of many studies which explore their pharmacological potential toward diverse molecular targets, known to monitor key mechanisms in cancer such as proliferation, migration and angiogenesis. The few peptides from scorpion venom that have an anti-tumor effect are generally cytotoxic.
[9] Scorpion peptides are disulfide bond-rich molecules presenting a sequence length ranging between 23 and about 80 amino acids. The majority of scorpion peptides have the sequence signature of the cystine stabilized a/b (CSa/b) motif [25] Some scorpion CSa/b peptides display a remarkable specificity for certain subtypes of ion channels [26] Scorpion venom contains also a significant number of peptides, without disulfide bridge, that exhibit antimicrobial, immunomodulatory, Bradykinin-potentiating and/or hemolytic activities [27]
[10] Only a few purified toxins seem to be endowed by the anticancer effects. The Chlorotoxin (CTX) isolated from Leiurus quinquestriatus scorpion venom [22], that have been shown to bind specifically to glioma cell surfaces as a specific chloride channel blocker and is currently in phase II of human trials [23] Today there are nearly 970 scorpion peptides (available in public databases) described by the animal toxin annotation project uniprot [24] The extensive screening of many scorpion venoms showed a great diversity in their composition with different pharmacological potentials. Recently, and for therapeutic purposes, special emphasis was given to bioactive peptides which have anticancer activity.
SUMMARY
I. The RK1 Peptide
[1 1] In one embodiment, the inventors present the first description of a first short 14 amino acid peptide (called RK1), purified from the venom of Buthus occitanus tunetanus, with the particular capabilities, among different other scorpion peptides, to inhibit cell proliferation, migration and angiogenesis of U87 (Glioblastoma) and IGR39 (Melanoma). Moreover, RK1 is a first peptide derived from scorpion venom exhibiting a potential anti-tumoral activity with no manifest toxicity. Our results suggest that, in terms of its primary structure, RK1 is unique compared to a variety of known peptides purified from scorpion venoms. In addition, RK1 is the first natural peptide able to abolish completely the proliferation of cancer cells. The chicken chorioallantoic membrane model revealed that RK1 strongly inhibits ex-vivo vascular growth.
[12] Interestingly, RK1 is a small peptide of 14 amino acids containing a single disulfide bond. RK1 may represent the first member of a new group of scorpion peptides and can be classified as one of the first anti-cancer scorpion venom peptides that do not have cytotoxic activity.
[13] RK1 could open new perspectives for the pharmaceutical application of short scorpion venom peptides in anticancer activity and may represent the first member of a new group of scorpion peptides.
II. The RK peptide
[14] In another embodiment, the inventors isolated and characterized RK, from the Buthus occitanus tunetanus scorpion venom targeting the cell adhesion activity by acting on integrins. To our knowledge, this is the first disintegrin-like peptide purified from a scorpion venom. Interestingly, RK peptide is unique among the variety of known peptides purified from scorpion venoms in terms of its structure. It is a small peptide of 17 amino acids (IDCGTVMIPSECDPKSS; SEQ ID NO:1) containing a single disulfide bond. RK may represent the first member of a new group of scorpion peptides.
[15] There are only 23 scorpion peptides published so far whose primary structure comprises lesser than 20 residues with different pharmacological activities [30] They all possess variable pharmacological properties. Their sequence covers a range of length anywhere between 4 and 17 amino acids. Among them, only PIMT and Pit toxins share the same sequence length than RK [31] It was therefore not trivial to assign RK to any of the pre-existing families because the peptide has no obvious structural or sequence homology with these other peptides.
[16] Disintegrin activity had only been characterized for snake venom molecules and not scorpion. Herein, the inventors present the first description of a short 17 amino acid peptide (termed RK), purified from the venom of Buthus occitanus tunetanus, able to inhibit the cell adhesion of U87 (Glioblastoma), IGR39 (Melanoma) and PC12 (Rat pheochromocytoma) to different extracellular matrix (ECM) receptors. Anti-integrin antibody assay suggests that RK interacts with both aibi and anb3 with a more pronounced effect for the former. This is the first report of a peptide with such dual activity. The molecular docking study shows that RK involves mainly two segments in its structure to interact with the aibi integrin. M7 of RK peptide seems to play an important role due to its ability to occupy a small hydrophobic pocket on the surface of the integrin. h and D2 are capable to interact with the C-loop of the receptor, while K15 establishes a salt bridge interaction with its E28s. The molecular modeling study, suggests the key contribution of the EHCI 2DI3 segment in the interaction with anb3 integrin. Our results highlight a new class of disintegrin molecules which opens a interesting perspectives for the pharmaceutical application of short scorpion venom peptides in cancer treatment.
[17] The present invention is directed to novel scorpion peptides with various biological activities and their uses in the treatment of various disorders. Several of the various features of the invention will be described hereinafter. It is to be understood that the invention is not limited in its application to the details set forth in the following embodiments, claims, description and Figures. The invention is capable of other embodiments and of being practiced or carried out in numerous other ways.
1. An isolated peptide selected from the group consisting of peptides whose amino acid sequence comprises or consists of:
IDCSKVNLTAECSS (peptide RK1 ; SEQ ID NO:2);
IDCGTVMIPSECDPKS (peptide RK; SEQ ID NO:3);
a sequence has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 91 %, at least about 92%, at least about 93%, at least about 94%, at last about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity to RK1 ; and
a sequence has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 91 %, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity to RK.
2. The peptide according to embodiment 1 , comprising at least one substitution modification, insertion, or deletion modification relative to IDCSKVNLTAECSS (SEQ ID NO:2) and IDCGTVMIPSECDPKSS (SEQ ID NO: 1).
3. The peptide according to any one of embodiments 1-2, wherein the sequence comprises: a.) the segment V6, M7, and l8 of the RK peptide, and/or SnECDPKSi6 (aa 1 1-16 of SEQ
ID NO:3), and/or Ei2CDPKi5 (aa 12-16 of SEQ ID NO:3), and/or DI 4PKI6 (aa 14-16 of
SEQ ID NO:3), of the RK peptide and, optionally, still binds a1 b1 integrin; or b.) DCSK (aa 2-5 of SEQ ID NO:2), DCSKXXXXXECS (SEQ ID NO:4), and/or
DCSKXXXXXECDS (SEQ ID NO:5) of the RK1 peptide.
4. The peptide according to any one of embodiments 1-3, wherein the sequence comprises residues EI 2CDPKI5 (aa 12-16 of SEQ ID NO:3) of the RK peptide. The peptide according to any one of embodiments 1-4, wherein the peptide is no more than 20 amino acids long.
The peptide according to any one of embodiments 1-5, wherein the peptide is 17 amino acids long.
The peptide according to any one of embodiments 1-6, wherein the peptide is 14 amino acids long.
The peptide according to any one of embodiments 1-7, wherein the amino acid sequence is I DCGTVMI PSECDP (aa 1-14 of SEQ ID NO: 1).
The peptide according to any one of embodiments 1-8, wherein the amino acid sequence is IDCSKVNLTAECSS (RK1 ; SEQ ID NO:2) or I DCGTVMI PSECDPKSS (RK; SEQ ID NO:3).
The peptide according to any one of embodiments 1-9, wherein the peptide is isolated from Buthus occitanus tunetanus scorpion venom.
The peptide according to any one of embodiments 1-10, wherein the peptide is produced synthetically.
The peptide according to any one of embodiments 1-1 1 , wherein the peptide comprises a disulfide bond.
The peptide according to any one of embodiments 1-12, comprising an N-terminal and/or C-terminal modification.
The peptide according to of any one of embodiments 1-13, wherein the peptide is amidated at its C-terminal end.
The peptide according to any one of embodiments 1-14, wherein the peptide is conjugated to a half-life extending moiety.
The peptide according to any one of embodiments 1-15, wherein the peptide is linked to a polymer.
The peptide according to any one of embodiments 1-16, wherein the polymer is chosen from polyalkylethers (e.g. polyethylene glycol and polypropylene glycol), polylactic acid, polyglycolic acid, polyoxyalkenes, polyvinylalcohol, polyvinylpyrrolidone, cellulose and cellulose derivatives, dextran, and dextran derivatives, polyvinyl pyrrolidones, glycopeptides, and polyamino acids.
The peptide according to any one of embodiments 1-17, wherein the peptide is linked to a coupling partner. The peptide according to any one of embodiments 1-18, wherein the coupling partner is chosen from an effector molecule, a label, a drug, a toxin, a carrier, and a transport molecule.
The peptide according to any one of embodiments 1-19, wherein the peptide is modified by the addition of cysteine or biotin.
The peptide according to any one of embodiments 1-20, wherein the peptide is modified by a moiety to facilitate crosslinking.
The peptide according to any one of embodiments 1-21 , wherein the moiety is chosen from benzophenone, maleimide, and activated esters.
The peptide according to any one of embodiments 1-22, wherein the peptide is not able to alter cell adhesion through a2b1 , anbq, anb5, a5b1 and a6b4 integrins, but significantly reduces the adhesive activity of a1 b1 and anb3 integrins to extracellular matrix proteins. The peptide according to any one of embodiments 1-23, wherein the extracellular matrix protein comprises collagen.
The peptide according to any one of embodiments 1-24, wherein the extracellular matrix protein comprises vitronectin, fibronectin, fibrinogen, osteopoietin, and/or bone sialoprotein.
The peptide according to any one of embodiments 1-25, wherein the peptide competes with a synthetic RGD peptide for binding to aibi and/or anb3 integrins
A composition comprising the peptide according to any one of embodiments 1-26 and a pharmaceutically acceptable carrier, diluent, or excipient.
The composition of embodiment 27, wherein the composition is a sustained release formulation or is in a sustained release carrier.
The composition according to any one of embodiments 27-28, wherein the composition is formulated to be administered in microspheres, liposomes, or one of other microparticulate delivery systems.
The composition according to any one of embodiments 27-29, wherein the sustained release carrier comprises a semipermeable polymer matrix.
The composition according to any one of embodiment 27-30 , wherein the semipermeable polymer matrix comprises one or more of polylactides copolymers of L-glutamic acid and gamma ethyl-L-glutamate, poly (2-hydroxyethyl-methacrylate), or ethylene vinyl acetate. A method of treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31. The method of embodiment 32, wherein the peptide is administered at a dosage ranging from 0.00003 mg/kg/day to 30 mg/kg/day.
The method according to any one of embodiments 32-33 or 45-60, wherein the peptide is administered at a dosage ranging from 0.003 mg/kg/ day to 3 mg/kg/day.
The method according to any one of embodiments 32-34 or 45-60, wherein the peptide is administered at a dosage ranging from 0.03 mg/kg/day to 0.3 mg/kg/day.
The method according to any one of embodiments 32-35 or 45-60, wherein the peptide is administered at a dosage ranging from 0.1 to 50 mg/kg/day, typically from about 0.5 to 1 mg/kg/day.
The method according to any one of embodiments 32-25 or 45-60, wherein the dosage of the peptide to be achieved in vivo may be equivalent to an in vitro level of 1 mM, 2 pM, 3 pM, 4 pM, 5 pM, 6 pM, 7 pM, 1 pM, 8 pM, 9 pM, 10 pM, 20 pM, 30 pM, 40 pM, 50 pM, 70 pM, 80 pM, 90 pM, or 100 pM.
The method according to any one of embodiments 32-25 or 45-60, wherein the dosage of the peptide to be achieved in vivo may be a blood/plasma level of at least 1 pM, 2 pM, 3 pM, 4 pM, 5 pM, 6 pM, 7 pM, 1 pM, 8 pM, 9 pM, 10 pM, 20 pM, 30 pM, 40 pM, 50 pM, 70 pM, 80 pM, 90 pM, or 100 pM.
The method according to any one of embodiments 32-38 or 45-60, wherein the peptide is administered intravenously continuously for more than one day.
The method according to any one of embodiments 32-39 or 45-60, wherein the composition is administered orally, parenterally, topically, by inhalation, intranasally, or recta I ly.
The method according to any one of embodiments 32-40 or 45-60, wherein the composition is administered intravenously, oral, sublingual, intranasal, intraocular, rectal, transdermal, mucosal, topical or parenteral.
The method according to any one of embodiments 32-41 or 45-60, wherein the composition comprises at least one additive chosen from a pharmaceutically acceptable excipients, carriers, preservatives, buffers, stabilizers, antioxidants, or other additives. The method according to any one of embodiment 32-42 or 45-60, wherein the composition is in a form chosen from a tablet, capsule, powder, or liquid.
The method according to any one of embodiment 32-43 or 45-60, wherein the liquid comprises at least one additive chosen from liquid carriers, petroleum, animal oils, vegetable oils, mineral oils, synthetic oils, physiological saline solutions, saccharide solutions, and glycols. The method according to any one of embodiment 32-44 or 45-60, wherein the cancer is selected from glioblastoma, melanoma, leukemia, and hepatocellular cancers, sarcoma, vascular endothelial cancers, breast cancers, central nervous system cancers (e.g. astrocytoma, gliosarcoma, neuroblastoma, oligodendroglioma and glioblastoma), prostate cancers, lung and bronchus cancers, larynx cancers, esophagus cancers, colon cancers, colorectal cancers, gastro-intestinal cancers, melanomas, ovarian and endometrial cancer, renal and bladder cancer, liver cancer, endocrine cancer (e.g. thyroid), and pancreatic cancer.
A method of inhibiting angiogenesis in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31.
A method for treatment of an integrin-associated disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31.
The method of embodiment 45 wherein the integrin-associated disease is osteoporosis, bone tumor or cancer growth, angiogenesis-related tumor growth and metastasis, tumor metastasis in bone, malignancy-induced hypercalcemia, angiogenesis-related eye diseases, Paget's disease, rheumatic arthritis, ovariectomy-induced physiological change, inflammation, coagulation diseases, or osteoarthritis.
The method according to any one of embodiments 32-48, wherein the method further comprises administering to the subject a therapeutically effective amount of one or more other therapeutic agents.
The method according to embodiment 49, wherein said therapeutic agent is selected from the group consisting of an antiangiogenic agent, a cytotoxic agent, a cytostatic agent, and an immunomodulatory agent.
The method according to any one of embodiment 32-50, wherein the composition is administered in combination with radiotherapy.
The method according to embodiment 50, wherein the at least one an antiangiogenic agent, a cytotoxic agent, a cytostatic agent comprises at least one agent chosen from endostatin, angiostatin, VEGF inhibitors, cytotoxic agents, alkaloids, antimetabolites, cancer growth inhibitors, gene therapy therapeutics, cancer vaccines, interferons, monoclonal antibodies, radiotherapy, hormonal therapy, and other supportive therapy. The method according to any one of embodiments 32-52, wherein the therapeutic agent is selected from Alkylating Agents, DNA Alkylating-like Agents, Alkylating Antineoplastic Agents, DNA replication and repair inhibitors, Cell Cycle Modulators, Apoptosis Regulators, Angiogenesis Inhibitors, Proteasome Inhibitors, Kinase Inhibitors, Protein Synthesis Inhibitors, Histone deacetylase inhibitors, Topoisomerase I Inhibitors, Topoisomerase II Inihibitors, DNA Intercalating Agents, RNA/DNA Antimetabolites, DNA Antimetabolites, Mitochondria Inhibitors, Antimitotic Agents, Nuclear Export Inhibitors, and Hormonal Therapies.
The method according to any one of embodiments 32-53, wherein therapeutically effective means that the administration of the composition to the subject results in any demonstrated clinical benefit compared with no therapy (when appropriate) or to a known standard of care.
The method according to any one of embodiment 54, wherein clinical benefit is assessed based on objective response rate (ORR) (determined using RECIST version 1.1), duration of response (DOR), progression-free survival (PFS), and/or overall survival (OS).
A method of decreasing binding of aibi and/or anb3 integrins expressing cells to extracellular molecules, comprising contacting the cells with according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31.
A method of screening for inhibition of tumor cell growth, comprising administering a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31 to a subject, and determining whether the peptide reduces tumor cell growth.
A method of inhibiting tumor cell proliferation, comprising contacting the tumor cell with an effective amount of a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31.
A method of inhibiting tumor cell migration, comprising contacting the tumor cell with an effective amount of a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31.
The method according to any one of embodiments 55-59, wherein the tumor cells are glioblastoma cells or melanoma cells.
A method of designing or screening for a disintegrin peptide or other integrin inhibitor, wherein the method comprises using the model described in the figures as a basis for the design or screening.
A peptide according to any one of embodiments 1-26 as a medicament. A composition according to any one of embodiment 27-31 as a medicament.
A peptide according to any one of embodiments 1 -26 for the treatment of cancer, inhibiting tumor cell migration, inhibiting tumor cell proliferation, decreasing binding of aibi and/or anb3 integrins expressing cells to extracellular molecules, treatment of an integrin- associated disease, and/or inhibiting angiogenesis.
A composition according to any one of embodiment 27-31 for the treatment of cancer, inhibiting tumor cell migration, inhibiting tumor cell proliferation, decreasing binding of aibi and/or anb3 integrins expressing cells to extracellular molecules, treatment of an integrin- associated disease, and/or inhibiting angiogenesis.
The use of a peptide according to any one of embodiments 1-26 or a composition according to anyone of embodiments 27-31 for the preparation of a drug for the treatment of cancer, inhibiting tumor cell migration, inhibiting tumor cell proliferation, decreasing binding of aibi and/or anb3 integrins expressing cells to extracellular molecules, treatment of an integrin-associated disease, and/or disorders mediated by/associated with angiogenesis.
The use of a peptide according to any one of embodiments 1-26 or a composition according to anyone of embodiments 27-31 for the preparation of a drug for the treatment of epilepsy, memory, learning, neuropsychiatric, neurological, neuromuscular, and immunological disorders, schizophrenia, bipolar disorder, sleep apnea, neurodegeneration, smooth muscle disorders, bacterial diseases, fungal diseases, malaria, viral diseases, Immuno-modulator-responsive disorders or pain.
The peptide according to any one of embodiments 1-26, or a composition according to anyone of embodiments 27-31 for use as a medicament.
The peptide according to any one of embodiments 1-26, or a composition according to anyone of embodiments 27-31 for use as a medicinal product intended for the treatment of cancer, inhibiting tumor cell migration, inhibiting tumor cell proliferation, decreasing binding of aibi and/or anb3 integrins expressing cells to extracellular molecules, treatment of an integrin-associated disease, disorders mediated by/associated with angiogenesis, epilepsy, memory, learning, neuropsychiatric, neurological, neuromuscular, and immunological disorders, schizophrenia, bipolar disorder, sleep apnea, neurodegeneration, smooth muscle disorders, bacterial diseases, fungal diseases, malaria, viral diseases, Immuno-modulator-responsive disorders, or pain.
A kit comprising a peptide according to any one of embodiments 1-26 or a composition according to any one of embodiments 27-31. BRIEF DESCRIPTION OF DRAWINGS
[18] Fig. 1A and B. Purification of RK1 from the venom Buthus occitanus tunetanus.
[19] (A) Purification of BotG50 on semi preparative C8 reverse phase HPLC column. Fractions 19-22 min were collected.
[20] (B) Separation of fraction 19-22 min on C18-RP-HPLC, fraction eluted at 37.5 min was collected. C. Final purification step to ensure the purity of peak eluting at 37.5 min (RK1). (D) MALDI- TOF mass spectrometry analysis shows a sharp peak at a molecular mass of 1467.67 Da. (E) Amino acid sequence of RK1.
[21] Fig. 2A and B. Effect of RK1 on tumour cell viability and proliferation
[22] (A). Cell viability of IGR39 and U87 cells incubated for 72 h with and without RK1. MTT solution was then added for 4 h. The MTT solution was removed and replaced with 100 pl_ of DMSO into each well in order to dissolve the precipitated formazan crystals 100 mI SDS 1 %. Finally, the absorbance was measured at 560 nm.
[23] (B). Proliferation of IGR39 and U87 cells incubated for 4 days with different concentrations of RK1 (2 and 4 mM). After 4 days of incubation, the wells are washed with PBS and the cells were fixed with 1 % glutaraldehyde every day, stained with 0.1 % crystal violet and quantified by absorbency at 560 nm.
[24] Fig. 3A and B. Effect of RK1 on tumour cell migration
[25] (A) IGR39 and U87 cells were incubated with RK1 (2 and 4mM) for 16 h. Photomicrographs showing cell migration into the denuded area in the absence and presence of RK1.
[26] (B) Quantification of the covered surface by IGR 39 and U87 cells in the absence and in the presence of RK1 was calculated using ImageJ. Values shown are mean (±SD). The histogram is representative of three independent experiments. ***Value significantly different versus corresponding untreated cells (P < 0.001).
[27] Fig. 4A and B. Chicken chorioallantoic membrane assay CAM
[28] (A) The CAM models were prepared using 8-day-old chick embryos treated as described in materials and methods. Filter disks were soaked in a 0.9% NaCI alone; b. 2 mM of RK1 ; c. 4 mM of RK1 ; d. 8 mM of RK1. After incubation for 72h, CAMs were photographed with a digital camera. Each group contained four CAMs and the experiment was repeated three times
[29] (B) The quantitative measurement of total vessel length was performed on 50% of the total CAM surface. [30] Fig. 5A-D. Purification of RK from the venom Buthus occitanus tunetanus. (A) Purification of BotG50 on semi preparative C8 reverse phase HPLC column. Fractions 19-22 min were collected. (B) Separation of fraction 19-22 min on C18-RP-HPLC, fraction eluted at 33.5 min was collected. (C) Final purification step to ensure the purity of peak eluting at 33.5 min (RK). (D) Amino acid sequence of RK (SEQ ID NO:1).
[31] Fig. 6A-C. Cell viability U87 (A), IGR39 (B) and PC12 (C) cells incubated for 72 h with different concentrations of RK. MTT solution was then added for 4 h. The MTT solution was removed and replaced with 100 pl_ of DMSO into each well in order to dissolve the precipitated formazan crystals 100 mI SDS 1 %. Finally, the absorbance was measured at 560 nm. All data shown are mean (±SD) from 3 experiments (n=3) performed in triplicate at different times.
[32] Fig. 7A-C. RK inhibits cell adhesion Glioblastoma cells U87 (A), Melanoma cells IGR39 (B) and Rat pheochromocytoma cells PC12 (C) Were then added to 96-well microtiter plates coated with different ECM proteins (type IV collagen, fibrinogen (Fg), fibronectin (Fn), laminin I (Lnl)) or poly-L-lysine and allowed to adhere for 1 h at 37 °C. After washing, adherent cells were stained with crystal violet, solubilized by SDS and absorbance was measured at 560 nm. All data shown are mean (±SD) from3 experiments (n=3) performed in triplicate at different times.
[33] Fig. 8A-C. RK inhibits integrin-mediated functions in tumour cells. After washing, adherent cells were stained with crystal violet, solubilized by SDS and absorbance was measured at 560 nm. Glioblastoma cells U87 (A), Melanoma cells IGR39 (B) and Rat pheochromocytoma cells PC12 (C), were preincubated with 5, 10, 15, 20 mM and 30 mM of RK for 30 min at room temperature and then added to wells coated with 10 pg/ml fibronectin (Fn), 50 pg/ml fibrinogen (Fg) or 10 pg/ml type IV collagen, (Col IV) and allowed attach for 1 h at 37 °C. Data shown are means (±SD) from 3 experiments (n=3) performed in triplicate at different times.
[34] Fig. 9A and B. Effect of RK on various integrins in cell adhesion assays (A) Adhesion assays were performed with various cell/ECM protein pairs involving unique integrins: aibi (PC12/type I collagen), a2bi (HT1080/type I collagen), a6b4 (HT29-D4/laminin), anb6 (HT29- D4/fibronectin), anb3 (HT29 D4/fibrinogen), anbd (HT29-D4/vitronectin) and a5bi (K562/fibronectin). Cells were preincubated without or with 15mM of RK for 30 min at room temperature. Cells were then added to 96-well microtiter plates coated with 5 pg/ml fibronectin, vitronectin or laminin-1 , with 10 pg/ml type I collagens or with 50 pg/ml fibrinogen and allowed to adhere for 1.30 or 2 h at 37°C. After washing, adherent cells were stained with crystal violet, solubilized by SDS and absorbance was measured at 560 nm. (B) Cells incubated without or with 10 pg/ml antibodies against aibi, anb3, a5bi or b3, with 1 mM GRGDSP peptide (RGD peptide), all experiments were performed in triplicate. Data shown are means (±SD) from 3 experiments (n=3) performed in triplicate at different times.
[35] Fig. 10A-C. Molecular modeling and molecular dynamics of RK peptide (A) We present the predicted model returned by the PEP-FOLD server. The Ca atoms of each of the residues are indicated in spheres. The disulfide bond between the 3rd and the 12th amino acid is colored in red. (B) RMSD time evolution (upper panel) and RMSF profile (lower panel) of RK peptide calculating for a molecular dynamics trajectory of 300 ns. (C) Potential of mean forces of the RK peptide calculated from the molecular dynamics simulation. The reaction coordinates consist of the radius of gyration (Rg) and the RMSD values relative to an average structure computed from the 300 ns simulation time.
[36] Fig. 1 1A-C. Peptide-Protein docking of RK with a1 b1 (A) RMSD time evolution of the best retained complex between RK and a1 b1 simulated for 10 ns. (B) Interaction of RK peptide (red) with the collagen IV binding site on the a1 b1. (C) The key interaction with the integrin are expressed in more details. The M7 residue is capable to interact with a small hydrophobic pocket on the surface of the integrin formed by the residues in yellow. We also demonstrate the interaction of 11 and I2 with the C-loop and the salt bridge between K15 and E285.
[37] Fig. 12A-C. Analysis of the interaction of RK with anb3 (A) We fitted the atoms of the ECD segment from each snapshot the molecular dynamics trajectory of RK to those of the RGD coordinates from the 1 L5G PDB structure. The ROSETTA clash score was then calculated. The reaction coordinates consists of the distance between E11 CA and D13 CG, and the angle between En CA, C12 CA and D13 CG. (B) The conformation of RK with the least RMSD value is presented along with RGD peptide (yellow sticks). (C) The complex of RK with anb3 was constructed by fitting the conformation corresponding to the lowest clash score of the ECD segment with the RGD coordinates of the cilengetide from the PDB structure 1 LG5.
DETAILED DESCRIPTION
I. Definitions
[38] Unless specifically stated or obvious from context, as used herein, the terms "a", "an", and "the" are understood to be singular or plural.
[39] Furthermore, "and/or" where used herein is to be taken as specific disclosure of each of the two specified features or components with or without the other. Thus, the term "and/or" as used in a phrase such as "A and/or B" herein is intended to include "A and B," "A or B," "A," (alone) and "B" (alone). Likewise, the term "and/or" as used in a phrase such as "A, B, and/or C" is intended to encompass each of the following embodiments: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
[40] Unless specifically stated or obvious from context, as used herein, the term "about" is understood as within a range of normal tolerance in the art, for example within 2 standard deviations of the mean. About can be understood as within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1 %, 0.5%, 0.1 %, 0.05%, or 0.01 % of the stated value. Unless otherwise clear from context, all numerical values provided herein are modified by the term about.
[41] Any compositions or methods provided herein can be combined with one or more of any of the other compositions and methods provided herein.
[42] In this disclosure, "comprises," "comprising," "containing" and "having" and the like can have the meaning ascribed to them in U.S. Patent law and can mean "includes," "including," and the like; "consisting essentially of" or "consists essentially" likewise has the meaning ascribed in U.S. Patent law and the term is open-ended, allowing for the presence of more than that which is recited so long as basic or novel characteristics of that which is recited is not changed by the presence of more than that which is recited, but excludes prior art embodiments.
[43] As used herein, the terms "determining", "assessing", "assaying", "measuring" and "detecting" refer to both quantitative and qualitative determinations, and as such, the term "determining" is used interchangeably herein with "assaying," "measuring," and the like. Where a quantitative determination is intended, the phrases "determining an amount" of an analyte and the like can be used. Where a qualitative and/or quantitative determination is intended, the phrase "determining a level" of an analyte or "detecting" an analyte is used.
[44] By "reference" is meant a standard of comparison. The standard can be an established method in the art. A control reference method is a reference method in which all of the parameters are identical to those of the method being compared with exception of the variable being tested.
[45] By "subject" is meant a mammal, including, but not limited to, a human or non human mammal, such as a bovine, equine, canine, ovine, or feline.
[46] Ranges provided herein are understood to be shorthand for all of the values within the range. For example, a range of 1 to 50 is understood to include any number, combination of numbers, or sub-range from the group consisting 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 ,
12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37,
38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, or 50. [47] "Binding affinity" generally refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g., of an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein, "binding affinity", "bind to", "binds to" or "binding to" refers to intrinsic binding affinity that reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody Fab fragment and antigen). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD).
[48] Standard assays for quantifying binding and determining binding affinity are known in the art and include, e.g., equilibrium dialysis, equilibrium binding, gel filtration, surface plasmon resonance, the use of a labeled binding partners, ELISAs and indirect binding assays (e.g., competitive inhibition assays). For example, as is well known in the art, the dissociation constant of a protein can be determined by contacting the protein with a binding partner and measuring the concentration of bound and free protein as a function of its concentration.
[49] As used herein, the term “linked” identifies a covalent linkage between two different groups (e.g., nucleic acid sequences, polypeptides, polypeptide domains) that may have an intervening atom or atoms between the two groups that are being linked. As used herein, “directly linked” identifies a covalent linkage between two different groups (e.g., nucleic acid sequences, polypeptides, polypeptide domains) that does not have any intervening atoms between the two groups that are being linked.
[50] As used herein, "disintegrin" refers to a class of cysteine-rich proteins that are potent soluble ligands of integrins and which are involved in regulating many processes such as cell-cell and cell-extracellular matrix adhesion, migration and invasion, cell cycle progression, differentiation and cell type speciation during development of many metazoan organisms, cell death and apoptosis. The tri-peptide motif RGD (Arg-Gly-Asp) is conserved in most monomeric disintegrins and is located at the tip of a flexible loop, the integrin-binding loop, which is stabilized by disulfide bonds and protruding from the main body of the polypeptide chain. All disintegrins purified from snake venoms bind to the fibrinogen receptor, integrin <¾,b3 the binding of which results in the inhibition of fibrinogen-dependent platelet aggregation. Most disintegrins also bind to anb3 (a vitronectin receptor) and anbi (a fibronectin receptor) in an RGD-dependent manner.
[51] "Disease" refers to any condition, infection, disorder, or syndrome that requires medical intervention or for which medical intervention is desirable. Such medical intervention can include treatment, diagnosis, and/or prevention.
[52] "Treatment," covers any administration or application of remedies for disease in a mammal, including a human, and includes inhibiting the disease, arresting its development, or relieving the disease, for example, by causing regression, or restoring or repairing a lost, missing, or defective function; or stimulating an inefficient process. The term includes obtaining a desired pharmacologic and/or physiologic effect, covering any treatment of a pathological condition or disorder in a mammal, including a human. The effect may be prophylactic in terms of completely or partially preventing a disorder or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disorder and/or adverse affect attributable to the disorder. Thus, the invention provides both treatment and prophylaxis. It includes (1) preventing the disorder from occurring or recurring in a subject who may be predisposed to the disorder but is not yet symptomatic, (2) inhibiting the disorder, such as arresting its development, (3) stopping or terminating the disorder or at least its associated symptoms, so that the host no longer suffers from the disorder or its symptoms, such as causing regression of the disorder or its symptoms, for example, by restoring or repairing a lost, missing or defective function, or stimulating an inefficient process, or (4) relieving, alleviating, or ameliorating the disorder, or symptoms associated therewith, where ameliorating is used in a broad sense to refer to at least a reduction in the magnitude of a parameter, such as inflammation, pain, and/or tumor size.
[53] As used herein, an“effective amount” means the amount of an agent that is effective for producing a desired effect in a subject. The term“therapeutically effective amount” denotes that amount of a drug or pharmaceutical agent that will elicit therapeutic response of an animal or human that is being sought. The actual dose which comprises the effective amount may depend upon the route of administration, the size and health of the subject, the disorder being treated, and the like.
[54] Therapeutically effective can mean to result in at least one anti-tumor effect chosen from reduction of tumor cell growth and reduction of tumor cell proliferation in the subject.
[55] By therapeutically effective in treating cancer is meant that the use of the peptide of the invention to treat cancer in a patient results in any demonstrated clinical benefit compared with no therapy (when appropriate) or to a known standard of care. Clinical benefit can be assessed by any method known to one of ordinary skill in the art. In one embodiment, clinical benefit is assessed based on objective response rate (ORR) (determined using RECIST version 1.1), duration of response (DOR), progression-free survival (PFS), and/or overall survival (OS). In some embodiments, a complete response indicates therapeutic benefit. In some embodiments, a partial response indicates therapeutic benefit. In some embodiments, stable disease indicates therapeutic benefit. In some embodiments, an increase in overall survival indicates therapeutic benefit. In some embodiments, therapeutic benefit may constitute an improvement in time to disease progression and/or an improvement in symptoms or quality of life. In other embodiments, therapeutic benefit may not translate to an increased period of disease control, but rather a markedly reduced symptom burden resulting in improved quality of life. As will be apparent to those of skill in the art, a therapeutic benefit may be observed using the peptide of the invention alone (monotherapy) or adjunctive to, or with, other anti-cancer therapies and/or targeted or non- targeted anti-cancer agents. Various methods for assessing therapeutic benefit are well known in the art.
[56] In some embodiments, therapeutic effect for determination of a particular dosage regimen is assessed using standard clinical tests designed to measure the response to a new treatment for cancer. To assess the therapeutic benefits of the peptides described herein one or a combination of the following tests can be used: (1) the Response Evaluation Criteria In Solid Tumors (RECIST) version 1.1 (for details, see Example 16), (2) the Eastern Cooperative Oncology Group (ECOG) Performance Status, (3) immune-related response criteria (irRC), (4) disease evaluable by assessment of tumor antigens, (5) validated patient reported outcome scales, and/or (6) Kaplan-Meier estimates for overall survival and progression free survival.
[57] The term “pharmaceutically acceptable carrier” as used herein may refer to compounds and compositions that are suitable for use in human or animal subjects, as for example, for therapeutic compositions administered for the treatment of a disorder or disease of interest.
[58] The term“pharmaceutical composition” is used herein to denote a composition that may be administered to a mammalian host, e.g., orally, parenterally, topically, by inhalation spray, intranasally, or rectally, in unit dosage formulations containing conventional non-toxic carriers, diluents, adjuvants, vehicles and the like.
[59] The term “parenteral” as used herein, includes subcutaneous injections, intravenous, intramuscular, intracisternal injection, or infusion techniques.
[60] "Inhibit" as used herein may mean prevent, suppress, repress, reduce or eliminate.
[61] Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, IC50 and so forth used in the specification and claims are to be understood as being modified in all instances by the term "about." Accordingly, unless indicated to the contrary, the numerical parameters set forth in the present specification and attached claims are approximations. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should at least be construed in light of the number of significant Fig.s and by applying ordinary rounding techniques. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the invention are approximations, the numerical values set in the examples, Tables and Fig.s are reported as precisely as possible. Any numerical values may inherently contain certain errors resulting from variations in experiments, testing measurements, statistical analyses and such.
[62] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
II. PEPTIDES
[63] In one embodiment, the invention provides peptide RK1 (IDCSKVNLTAECSS; SEQ ID NO:2, a 14 mer), which is the first very short peptide from Buthus occitanus tunetanus that inhibits tumor cell migration, proliferation and angiogenesis.
[64] In another embodiment, the invention provides peptide RK (IDCGTVMIPSECDPKSS; SEQ ID NO: 1 , a 17 mer), which is the first scorpion peptide with dual disintegrin activity on a1 b1 and anb3 integrins.
[65] In one embodiment the peptide is isolated from Buthus occitanus tunetanus scorpion venom. Methods for isolation are exemplified in the EXAMPLES.
[66] In one embodiment, the peptide is produced synthetically.
[67] In one embodiment, the peptides of the invention include modifications of the RK1 and RK peptides according to which the resulting peptides are not natural. In some embodiments, these are modifications to the actual sequence or to the chemistry of the amino acids (e.g., non-natural amino acids), as exemplified below. In some embodiments, these modifications are additions or conjugations of the peptides with other molecules, as exemplified below. In some embodiments, the modifications are the result of the formulation of the peptides with compounds that affect the properties of the peptides, be it solubility, half-life, antigenicity, biological activity, stability, or any other physical and/or chemical change to the native peptide.
[68] In one embodiment, the invention provides a peptide whose amino acid sequence has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to IDCSKVNLTAECSS or IDCGTVMIPSECDPKSS (RK1 ;SEQ ID NO:2, or RK; SEQ ID NO: 1). Peptides of the invention may be able to inhibit tumor cell migration, proliferation, and/or angiogenesis. Peptides of the invention may also have dual disintegrin activity on a1 b1 and anb3 integrins.
[69] In a preferred embodiment, the amino acid sequence of the peptide is IDCSKVNLTAECSS (RK1 ; SEQ ID NO:2). In another embodiment, the amino acid sequence of the peptide is IDCGTVMIPSECDPKSS (RK; SEQ ID NO: 1).
[70] In one embodiment, the sequence comprises a disulfide bond.
[71] In a preferred embodiment, the sequence comprises at least one substitution modification, insertion, or deletion modification relative to IDCSKVNLTAECSS (SEQ ID NO:2) and IDCGTVMIPSECDPKSS (SEQ ID NO: 1).
[72] In one embodiment, the peptide is not able to alter cell adhesion through a2b1 , anb6, anb5, a5b1 and a6b4 integrins, but significantly reduces the adhesive activity of a1 b1 and anb3 integrins to extracellular matrix proteins. In one embodiment, the extracellular matrix protein comprises collagen. In other embodiments, the extracellular matrix protein comprises vitronectin, fibronectin, fibrinogen, osteopoietin, and/or bone sialoprotein.
[73] In one embodiment, the peptide competes with a synthetic RGD peptide for binding to aibi and/or anb3 integrins. Competition assays are well known in the art.
[74] In a preferred embodiment, the sequence comprises the segment V6, M7, and I8 of the RK peptide and (and still binds a1 b1 integrin).
[75] In a preferred embodiment, the sequence comprises residues SnECDPKSi6 (aa 11-16 of SEQ ID NO:3), Ei2CDPKi5 (aa 12-15 of SEQ ID NO:3), or Di4PKi6 (aa 14-16 of SEQ ID NO:3), of the RK peptide, or DCSK (aa 2-5 of SEQ ID NO:2), DCSKXXXXXECS (SEQ ID NO:4) or DCSKXXXXXECDS (SEQ ID NO:5) of the RK1 peptide.
[76] Peptide K can be divided into three subsegments using the computation study and molecular model described in the EXAMPLES. In one embodiment, the peptide comprises at least one of the RK segments. In one embodiment, the peptide comprises two of these RK segments. In one embodiment, the peptide comprises all segments. In some embodiments, the sequence of one or more of the segments has been modified.
[77] In one embodiment, the sequence of the peptide is designed while considering the molecular docking studies and results described in this disclosure.
[78] In one embodiment, the invention comprises the application of the information gathered using any one or more of the computational studies, molecular dynamic studies, a molecular docking model described in this disclosure, particularly in designing disintegrins and peptides of the invention. [79] As is used herein, the terms "at least 70% identical" or "at least 70% identity" means that a polypeptide or peptide sequence or a polynucleotide sequence shares 70%-100% sequence identity with a reference sequence. This range of identity is inclusive of all whole numbers (e.g., 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) or partial numbers (e.g., 72.15, 87.27%, 92.83%, 98.11 %-to two significant Fig.s) embraced within the recited range numbers, therefore forming a part of this description. For example, an amino acid sequence with 200 residues that share 85% identity with a reference sequence would have 170 identical residues and 30 non-identical residues. Similarly, an amino acid sequence with 235 residues may have 200 residues that are identical to a reference sequence, thus the amino acid sequence will be 85.1 1 % identical to the reference sequence. Similarly, the terms "at least 80%, " "at least 90%, " "at least 95%" and "at least 99%" and the like are inclusive of all whole or partial numbers within the recited range. As is used herein, the terms "greater than 95% identical" or "greater than 95% identity" means that a sequence shares 95.01 %-100% sequence identity with a reference sequence. This range is all inclusive. Differences in identity can be due to additions, deletions or substitutions of residues in a first sequences compared to a second sequence.
[80] The percent identity can be measured using sequence comparison software or algorithms or by visual inspection. Various algorithms and software are known in the art that can be used to obtain alignments of amino acid or nucleotide sequences (see e.g., Karlin et al., 1990, Proc. Natl. Acad. Sci. USA, 87:2264-2268, as modified in Karlin et al., 1993, Proc. Natl. Acad. Sci. USA, 90:5873-5877, and incorporated into the NBLAST and XBLAST programs (Altschul et al., 1991 , Nucleic Acids Res., 25:3389-3402). In certain embodiments, Gapped BLAST can be used as described in Altschul et al., 1997, Nucleic Acids Res. 25:3389-3402. BLAST-2, WU- BLAST-2 (Altschul et al., 1996, Methods in Enzymology, 266:460-480), ALIGN, ALIGN-2 (Genentech, South San Francisco, Calif.) or Megalign (DNASTAR).
[81] In some embodiments, the peptide is 20 amino acids long or less. In some embodiments, the peptide is between 20 and 15 amino acids long. In some embodiments, the peptide is 14 amino acids long or less. In some embodiments, the peptide is 17 amino acids long or less.
[82] In one embodiment, the peptide comprises a modification relative to IDCSKVNLTAECSS (SEQ ID NO:2) or IDCGTVMIPSECDPKSS (SEQ ID NO: 1). In some embodiments, the comprising at least one substitution modification, insertion modification, or deletion modification (i.e. sequence modifications) relative to IDCSKVNLTAECSS (SEQ ID NO:2) and IDCGTVMIPSECDPKSS (SEQ ID NO: 1). [83] Substitutes for an amino acid within the peptide sequence may be selected from other members of the class to which the amino acid belongs. For example, the nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine. Amino acids containing aromatic ring structures include phenylalanine, tryptophan, and tyrosine. The polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine. The positively charged (basic) amino acids include arginine and lysine. The negatively charged (acidic) amino acids include aspartic acid and glutamic acid. For example, one or more amino acid residues within the sequence can be substituted by another amino acid of a similar polarity, which acts as a functional equivalent, resulting in a silent alteration.
[84] A conservative change generally leads to less change in the structure and function of the resulting protein. A non-conservative change is more likely to alter the structure, activity, or function of the resulting protein. For example, the peptide of the present disclosure comprises one or more of the following conservative amino acid substitutions: replacement of an aliphatic amino acid, such as alanine, valine, leucine, and isoleucine, with another aliphatic amino acid; replacement of a serine with a threonine; replacement of a threonine with a serine; replacement of an acidic residue, such as aspartic acid and glutamic acid, with another acidic residue; replacement of a residue bearing an amide group, such as asparagine and glutamine, with another residue bearing an amide group; exchange of a basic residue, such as lysine and arginine, with another basic residue; and replacement of an aromatic residue, such as phenylalanine and tyrosine, with another aromatic residue.
[85] Particularly preferred amino acid substitutions include: a) Ala for Glu or vice versa, such that a negative charge may be reduced; b) Lys for Arg or vice versa, such that a positive charge may be maintained; c) Ala for Arg or vice versa, such that a positive charge may be reduced; d) Glu for Asp or vice versa, such that a negative charge may be maintained; e) Ser for Thr or vice versa, such that a free --OH can be maintained; f) Gin for Asn or vice versa, such that a free NH2 can be maintained; g) lie for Leu or for Val or vice versa, as roughly equivalent hydrophobic amino acids; h) Phe for Tyr or vice versa, as roughly equivalent aromatic amino acids; and i) Ala for Cys or vice versa, such that disulphide bonding is affected.
[86] Furthermore, if desired, nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the peptide sequence. Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids, 2,4- diaminobutyric acid, a-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, g- Abu, e-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, b-alanine, fluoro-amino acids, designer amino acids such as b-methyl amino acids, Ca-methyl amino acids, Na-methyl amino acids, and amino acid analogs in general. Furthermore, the amino acid can be D (dextrorotary) or L (levo rotary).
[87] In some embodiments, the peptide comprises an N-terminal and/or C-terminal modification. The modifications can comprise the addition or replacement of the termini with amino acid analogs. Peptidomimetics can be incorporated into a peptide to induce or favor specific secondary structures. A desamino or descarboxy residue can be incorporated at the terminal ends of the peptide, so that there is no terminal amino or carboxyl group, to decrease susceptibility to proteases or to restrict conformation. C-terminal functional groups include amide, amide lower alkyl, amide di (lower alkyl), lower alkoxy, hydroxy, and carboxy, and the lower ester derivatives thereof, and the pharmaceutically acceptable salts thereof.
[88] The peptide might also comprise other modifications. For example, the peptide may be glycosylated, phosphorylated, sulfated, glycosylated, animated, carboxylated, acetylated. The C-terminal may be modified with addition of peptide alcohols and aldehydes, addition of esters, addition of p-nitorailine and thioesteres and multipelantigens peptides. The N-terminal and side chains may be modified by PEGylation, acetylation, formylation, addition of a fatty acid, addition of benzoyl, addition of bromoacetyl, addition of pyroglutamyl, succinylation, addition of tetrabutyoxycarbonyl and addition of 3-mercaptopropyl, acylations (e.g. lipopeptides), biotinylation, phosphorylation, sulfation, glycosylation, introduction of maleimido group, chelating moieties, chromophores and flurophores.
[89] The peptide may be conjugated to a fatty acid, e.g. the peptide is myristoylated. For example, a fatty acid may be conjugated to the N-terminus of the peptide, such fatty acids include caprylic acid (C8), capric acid (C10), lauric acid (C12), myristic acid (C14), palmitic acid (C16) or stearic acid (C18) etc. Furthermore cysteines in peptides can be palmitoylated.
[90] The peptide may be conjugated or linked to another peptide, such as a carrier peptide. The carrier peptide may facilitate cell-penetration, such as antennapedia peptide, penetratin peptide, TAT, tranportan or polyarginine.
[91] The peptides may be cyclic. The peptide disclosed herein may be cyclized by adding a single or multiple disulfide bridges, adding a single or multiple amide bonds between the N- and C-terminus, heat to tail cyclization, side chain cyclization (e.g. lactam bridge, thioester), hydrocarbon-stabled peptides. [92] The peptide may be labeled with heavy isotope labeling, e.g. 15N, 13C, FITC, conjugation to a carrier protein, conjugation to imaging agent, FRET substrates with a fluorophore/quencher pair, peptide-DNA conjugation, peptide-RNA conjugation and peptide- enzyme labeling.
[93] The peptide may be within a fusion protein such as fused to a polypeptide or peptide which promotes oligomerization, such as a leucine zipper domain; a polypeptide or peptide which increases stability or to increase half-life, such as an immunoglobulin constant region; and a polypeptide which has a therapeutic activity different from peptide or the invention, a chemotherapeutic agent, an antibody or protein for tissue specific targeting. Peptides of the invention include fusion peptides. For example, fusion peptides may comprise peptides of the invention linked, for example, to antibodies that target the peptides to diseased tissue, for example, tumor tissue or the retina.
[94] The peptides may be fused with the constant domain of immunoglobulins (IgA, IgE, IgG, IgM), or portions thereof (CH1 , CH2, CH3, or any combination thereof), resulting in chimeric polypeptides. These fusion polypeptides or proteins can facilitate purification and may show an increased half-life in vivo. Such fusion proteins may be more efficient in binding and neutralizing other molecules than monomeric polypeptides or fragments thereof alone.
[95] Fusion proteins of the invention also include the peptides fused with albumin, for example recombinant human serum albumin or fragments or variants thereof (see, e.g., US Patent No. 5876969, EP Patent 0413622 and US Patent No. 5766883).
[96] Fusions can be made either at the amino terminus or at the carboxy terminus of the polypeptide. The fusion proteins may be direct with no linker or adapter molecule or indirect using a linker or adapter molecule. A linker or adapter molecule may be one or more amino acid residues, typically up to about 20 to about 50 amino acid residues. A linker or adapter molecule may also be designed with a cleavage site for a protease to allow for the separation of the fused moieties. For example, the peptide may be fused to one or more domains of an Fc region of human IgG to increase the half-life of the peptide or the addition of a Fab variable domain to shorten the half-life of the peptide.
[97] A peptide of the invention can be modified with or covalently coupled to one or more of a variety of hydrophilic polymers to increase solubility and circulation half-life of the peptide. Suitable nonproteinaceous hydrophilic polymers for coupling to a peptide include, but are not limited to, polyalkylethers as exemplified by polyethylene glycol and polypropylene glycol, polylactic acid, polyglycolic acid, polyoxyalkenes, polyvinylalcohol, polyvinylpyrrolidone, cellulose and cellulose derivatives, dextran, and dextran derivatives, polyvinyl pyrrolidones, glycopeptides, and polyamino acids.
[98] Generally, such hydrophilic polymers have an average molecular weight ranging from about 500 to about 100,000 daltons, from about 2,000 to about 40,000 daltons, or from about 5,000 to about 20,000 daltons. The peptide can be derivatized with or coupled to such polymers using any of the methods known in the art.
[99] In some embodiments, the peptide is linked to a coupling partner. In some embodiments, the coupling partner is chosen from an effector molecule, a label, a drug, a toxin, a carrier, and a transport molecule.
[100] In some embodiments, the polypeptide is modified by the addition of cysteine or biotin.
[101] In some embodiments, the peptide is modified by a moiety to facilitate crosslinking. In some embodiments, the moiety is chosen from benzophenone, maleimide, and activated esters.
[102] The peptides may be in the form of multimers. Thus, multimers of 2, 3 or more individual RK1 , RK, or other peptide of the invention monomeric units, or two or more other peptides of the invention, or their combination, are within the scope of the invention.
[103] In one embodiment, such multimers may be used to prepare a monomeric peptide by preparing a multimeric peptide that includes the monomeric unit, and a cleavable site (i.e., an enzymatically cleavable site), and then cleaving the multimer to yield a desired monomer. The multimers can be homomers or heteromers.
[104] In one embodiment, the use of multimers can increase the binding affinity for an integrin. In some embodiments, the integrin is a1 b1 and/or anb3 integrin.
[105] Homodimeric and monomeric peptides described herein may be used for any purposes for which native homodimeric disintegrins may be employed.
[106] The peptides described herein are intended, at least in some embodiments, to be administered to a human or other mammal to treat or prevent a disorder associated with tumor cell migration, tumor cell proliferation, cell adhesion, and angiogenesis. Peptides are typically administered parenterally, e.g., by intravenous, subcutaneous, or intramuscular injection, or via the intranasal cavity, and may be readily metabolized by plasma proteases. In some embodiments the peptide may be delivered in microcapsules of poly(DL-lactide-co-glycolide) - controlled release over 30 days.
[107] Various prodrugs have been developed that enable parenteral and oral administration of therapeutic peptides. In certain embodiments, a prodrug is produced by modifying a pharmaceutically active compound such that the active compound will be regenerated upon in vivo administration. The prodrug can be designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug. By virtue of knowledge of pharmacodynamic processes and drug metabolism in vivo, those of skill in this art, once a pharmaceutically active compound is known, can design prodrugs of the compound.
[108] Peptides or polypeptides can be conjugated to various moieties, such as polymeric moieties, to modify the physiochemical properties of the peptide drugs, for example, to increase resistance to acidic and enzymatic degradation and to enhance penetration of such drugs across mucosal membranes.
[109] In alternate embodiments, the peptides can be provided in a suitable capsule or tablet with an enteric coating, so that the peptide is not released in the stomach. Alternatively, or additionally, the peptide can be provided as a prodrug, such as the prodrugs described above. In one embodiment, the peptides are present in drug delivery devices as prodrugs.
[1 10] The activity of the peptides of the invention can be measured by methods that are routine in that art. In some embodiments, the activity is measured according to the methods described in the EXAMPLES.
[1 1 1] In addition, cell detachment and adhesion may be detected by any suitable method, which may include, but are not limited to, cell cytometry (e.g. trypan blue), fluorescent based cell detection assays (e.g. Calcein AM (InVitrogen, Inc.), and Mitotracker Red (InVitrogen, Inc.), luminescent based detection assays (e.g. Cell-Titer glo (Promega, Inc.) and spectrophotometry based detection assays (e.g. crystal violet, MTS/MTT assays such as Promega CellTiter 96.RTM. AQueous Non-Radioactive Cell Proliferation Assay, Promega, Inc. and Chemicon Cell Adhesion Assays).
[1 12] Also, cell migration or invasion may be detected by any suitable method, which may include, but are not limited to, the scratch wound assay, cell invasion assays using fluorescent detection of cell invasion (e.g. activin, serum) (BD BioCoat matrigel invasion chambers, Fisher Scientific; EMD, Calbiochem; Chemicon International) with or without the use of a chemotaxic agent.
[1 13] Cell viability may be detected by any suitable method, which may include, but are not limited to, cell cytometry (e.g. trypan blue), fluorescent based cell detection assays (e.g. calcein AM (InVitrogen, Inc.), and Mitotracker Red (InVitrogen, Inc.)), luminescent based detection assays (e.g. Cell-Titer glo (Promega, Inc.)) and spectrophotometry based detection assays (e.g. crystal violet, MTS/MTT assays such as Promega CellTiter 96.RTM. AQueous Non- Radioactive Cell Proliferation Assay, Promega, Inc.).
[1 14] The effects of the peptides of the invention on cell proliferation may be assessed by any suitable method known in the art, including the method exemplified in the EXAMPLES section. Numerous methods of assessing cell proliferation are known in the art, including, for example, DNA synthesis cell proliferation assays (BrdU, IdU and CldU, EdU), metabolic proliferation assays (MTT, XTT, WST-1), detection of proliferation markers (PCNA, Ki67, MCM- 2), measuring ATP. The detection methods for these assays can be ICC, IHC, FACS, ELISA, WB, microscopy, among others.
[1 15] Animal models to determine antitumor efficacy of a compound are generally carried out in mice. Either murine tumor cells are inoculated subcutaneously into the hind flank of mice from the same species (syngeneic models) or human tumor cells are inoculated subcutaneously into the hind flank of severe combined immune deficient (SCID) mice or other immune deficient mouse (nude mice) (xenograft models). Advances in mouse genetics have generated a number of mouse models for the study of various human diseases including cancer. The MMHCC (Mouse models of Human Cancer Consortium) web page (emice.nci.nih.gov), sponsored by the National Cancer Institute, provides disease-site-specific compendium of known cancer models, and has links to the searchable Cancer Models Database (cancermodels.nci.nih.gov), as well as the NCI-MMHCC mouse repository. Mouse repositories can also be found at: The Jackson Laboratory, Charles River Laboratories, Taconic, Harlan, Mutant Mouse Regional Resource Centers (MMRRC) National Network and at the European Mouse Mutant Archive. Such models may be used for in vivo testing of the peptides of the invention, as well as for determining a therapeutically effective dose.
Production of molecules with novel activities based on RK and RK1
[1 16] The generation of molecules, with high affinity and specificity for biological targets, is a central goal in chemistry, biology and pharmaceutical sciences. In particular, binding ligands is important for the creation of drugs that can intervene in the biological processes. RK and RK1 bear many excellent properties that can serve as an alternative protein scaffold for biomedical application. RK and RK1 are bioactive mini-proteins from scorpion venoms that have the unique topological feature of cyclic backbone . Because of this structure, they are ultra-stable, present low immunogenicity (important to reduce unexpected side effects and damage of healthy tissue) and have advantages in passing tissue barrier. Thus, they attract interest as a peptide- based templates for drug design applications or diagnostic agents. This allows them to be used as pharmaceutical templates onto which bioactive peptide sequences can be grafted. Production of native or modified peptide
[1 17] RK and RK1 present many other benefits, such as cost reduction during development/production (chemical and genetical engineering) due to their limited amino-acid composition.
[1 18] Different methods for generating these molecules can be used: Site-direct mutagenesis: different approaches- including minimal residue substitution, e.g., one or more amino acid additions, amino acid substitutions, amino acid deletions and combinatorial chemistry- can be employed to engineer molecules with new functions. Molecular docking models are also established to investigate the interactions between receptors and ligands. Functional epitope exchange Grafting epitopes with known function is a straightforward stratagem for a protein to gain new function.
[1 19] The peptides of the present invention can be synthesized de novo using conventional solid phase synthesis methods. In such methods, the peptide chain is prepared by a series of coupling reactions in which the constituent amino acids are added to the growing peptide chain in the desired sequence. The use of various N-protecting groups, e.g., the carbobenzyloxy group or the t-butyloxycarbonyl group; various coupling reagents e.g., dicyclohexylcarbodiimide or carbonyldimidazole; various active esters, e.g., esters of N- hydroxyphthalimide or N-hydroxy-succinimide; and the various cleavage reagents, e.g., trifluoroactetic acid (TFA), HCI in dioxane, boron tris-(trifluoracetate) and cyanogen bromide; and reaction in solution with isolation and purification of intermediates are within the skill in the art. A preferred peptide synthesis method follows conventional Merrifield solid phase procedures well known to those skilled in the art.
[120]
III. COMPOSITIONS
[121] The invention also provides compositions comprising one or more of the peptides of the invention. In some embodiments, the compositions comprise a pharmaceutically acceptable carrier, diluent, or excipient.
[122] The term "pharmaceutically acceptable carrier" refers to a carrier for administration of a therapeutic agent. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., intravenous, oral topical, aerosol, suppository, parenteral or spinal injection. Such carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. For drugs administered orally, pharmaceutically acceptable carriers include, but are not limited to pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives. Suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents. Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.
[123] Excipients or other additives included in a composition have different purposes depending, for example on the nature of the drug, and the mode of administration. Examples of generally used excipients include, without limitation: stabilizing agents, solubilizing agents and surfactants, buffers, antioxidants and preservatives, tonicity agents, bulking agents, lubricating agents, emulsifiers, suspending or viscosity agents, inert diluents, fillers, disintegrating agents, binding agents, wetting agents, lubricating agents, antibacterials, chelating agents, sweetners, perfuming agents, flavouring agents, coloring agents, administration aids, and combinations thereof.
[124] The peptides of the invention can be formulated for sustained release or comprise a sustained release carrier. In some embodiments, the sustained release carrier comprises a semipermeable polymer matrix. In some embodiments, the semipermeable polymer matrix comprises one or more of polylactides copolymers of L-glutamic acid and gamma ethyl-L- glutamate, poly (2-hydroxyethyl-methacrylate), or ethylene vinyl acetate.
[125] The peptides of the invention, or pharmaceutically acceptable salts thereof, can be formulated for oral, sublingual, intranasal, intraocular, rectal, transdermal, mucosal, topical or parenteral administration for the therapeutic or prophylactic treatment of a variety of disorders, as described below. Parenteral modes of administration include without limitation, intradermal, subcutaneous (s.c., s.q., sub-Q, Hypo), intramuscular (i.m.), intravenous (i.v.), intraperitoneal (i.p.), intra-arterial, intramedulary, intracardiac, intra-articular (joint), intrasynovial (joint fluid area), intracerabral or intracranial, intraspinal, intracisternal, and intrathecal (spinal fluids). Any known device useful for parenteral injection or infusion of drug formulations can be used to effect such administration. For oral and/or parental administration, compounds of the present invention can be mixed with conventional pharmaceutical carriers and excipients and used in the form of solutions, emulsions, tablets, capsules, soft gels, elixirs, suspensions, syrups, wafers and the like.
[126] Parenteral administration, in particular intravenous injection, is most commonly used for administering peptides, and the composition may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water. [127] For example, in intravenous (IV) use, a sterile composition of the peptide and optionally one or more additives, including solubilizers or surfactants, can be dissolved or suspended in any of the commonly used intravenous fluids and administered by infusion. Intravenous fluids include, without limitation, physiological saline, phosphate buffered saline, 5% glucose or Ringer's solution.
[128] In another example, in intramuscular preparations, a sterile formulation of the peptide of the present invention or suitable soluble salts or prodrugs forming the peptide, can be dissolved and administered in a pharmaceutical diluent such as Water-for-lnjection (WFI), physiological saline or 5% glucose. A suitable insoluble form of the compound may be prepared and administered as a suspension in an aqueous base or a pharmaceutically acceptable oil base, e.g. an ester of a long chain fatty acid such as ethyl oleate.
[129] Sterile injectable solutions are prepared by incorporating the peptides of the invention in the required amount in the appropriate solvent with various of the other ingredients enumerated below, as required. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and/or the other ingredients. In the case of sterile powders for the preparation of sterile injectable solutions, suspensions or emulsion, the preferred methods of preparation are vacuum-drying or freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered liquid medium thereof. The liquid medium should be suitably buffered if necessary and the liquid diluent first rendered isotonic prior to injection with sufficient saline or glucose. The composition must be stable under the conditions of manufacture and storage, and preserved against the contaminating action of microorganisms, such as bacteria and fungi. It will be appreciated that endotoxin contamination should be kept minimally at a safe level, for example, less that 0.5 ng/mg protein. Suitable pharmaceutically acceptable carriers for parenteral composition include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as polyethylene glycol (PEG). Aqueous injection suspensions may contain compounds which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, dextran, or the like. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl cleats or triglycerides, or liposomes.
[130] For oral use, solid formulations such as tablets and capsules are particularly useful. Sustained released or enterically coated preparations may also be devised. For pediatric and geriatric applications, suspension, syrups and chewable tablets are especially suitable. For oral administration, the pharmaceutical compositions are in the form of, for example, tablets, capsules, suspensions or liquid syrups or elixirs, wafers and the like. For general oral administration, excipient or additives include, but are not limited to inert diluents, fillers, disintegrating agents, binding agents, wetting agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives.
[131] The oral pharmaceutical composition is preferably made in the form of a unit dosage containing a therapeutically-effective amount of the active ingredient. Examples of such dosage units are tablets and capsules. For therapeutic purposes, the tablets and capsules which can contain, in addition to the active ingredient, conventional carriers such as: inert diluents (e.g., sodium and calcium carbonate, sodium and calcium phosphate, and lactose), binding agents (e.g., acacia gum, starch, gelatin, sucrose, polyvinylpyrrolidone (Providone), sorbitol, or tragacanth methylcellulose, sodium carboxymethylcellulose, hydroxypropyl methylcellulose, and ethylcellulose), fillers (e.g., calcium phosphate, glycine, lactose, maize-starch, sorbitol, or sucrose), lubricants or lubricating agents (e.g., magnesium stearate or other metallic stearates, stearic acid, polyethylene glycol, waxes, oils, silica and colloical silica, silicon fluid or talc), disintegrants or disintegrating agents (e.g., potato starch, corn starch and alginic acid), flavouring, coloring agents, or acceptable wetting agents. Carriers may also include coating excipients such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.
[132] Oral liquid preparations, generally in the form of aqueous or oily solutions, suspensions, emulsions, syrups or elixirs, may contain conventional additives such as suspending agents, emulsifying agents, non-aqueous agents, preservatives, coloring agents and flavoring agents. Examples of additives for liquid preparations include acacia, almond oil, ethyl alcohol, fractionated coconut oil, gelatin, glucose syrup, glycerin, hydrogenated edible fats, lecithin, methyl cellulose, methyl or propyl para-hydroxybenzoate, propylene glycol, sorbitol, or sorbic acid.
[133] For both liquid and solid oral preparations, flavoring agents such as peppermint, oil of wintergreen, cherry, grape, fruit flavoring or the like can also be used. It may also be desirable to add a coloring agent to make the dosage form more aesthetic in appearance or to help identify the product. For topical use the compounds of present invention can also be prepared in suitable forms to be applied to the skin, or mucus membranes of the nose and throat, and can take the form of creams, ointments, liquid sprays or inhalants, lozenges, or throat paints. Such topical formulations further can include chemical compounds such as dimethylsulfoxide (DMSO) to facilitate surface penetration of the active ingredient. For application to the eyes or ears, the compounds of the present invention can be presented in liquid or semi-liquid form formulated in hydrophobic or hydrophilic bases as ointments, creams, lotions, paints or powders. For rectal administration the compounds of the present invention can be administered in the form of suppositories admixed with conventional carriers such as cocoa butter, wax or other glyceride.
[134] The peptides of the invention may be formulated into a composition in a free acid or base, neutral or salt form. Pharmaceutically acceptable salts are salts that substantially retain the biological activity of the free acid or base. These include the acid addition salts, e.g., those formed with the free amino groups of a proteinaceous composition, or which are formed with inorganic acids such as for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric or mandelic acid. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as for example, sodium, potassium, ammonium, calcium or ferric hydroxides; or such organic bases as isopropylamine, trimethylamine, histidine or procaine. Pharmaceutical salts tend to be more soluble in aqueous and other protic solvents than are the corresponding free base forms.
[135] The compositions comprising a peptide of the present invention can contain from about 0.1 % to about 99.9%, about 1 % to about 98%, about 5% to about 95%, about 10% to about 80% or about 15% to about 60% by weight of the active peptide.
IV. METHODS OF USE
[136] In one embodiment, the invention provides peptide RK1 (a 14 mer), which is the first very short peptide from Buthus occitanus tunetanus that inhibits tumor cell migration, proliferation, and angiogenesis.
[137] In one embodiment, the peptides may be used to treat diseases or disorders mediated by tumor cell migration, primarily cancer. In one embodiment, the peptides may be used to treat diseases or disorders mediated by tumor cell proliferation, also known as neoplastic disorders. In one embodiment, the peptides may be used to treat diseases or disorders mediated by angiogenesis. Diseases associated with these aspects of tumor development including tumor cell spreading from the original site, colonisation of new tumor sites, and neovascularization can also be treated with the peptides of the invention.
[138] In one embodiment, the peptides of the invention are used as anti-cancer agents. The term“anti-cancer agent” includes an agent that can inhibit cancer cell growth/proliferation, migration, and/or adhesion. In one embodiment, the peptides of the invention can be used to inhibit angiogenesis, or a combination thereof. Accordingly, the peptides of the invention are capable of acting both on cancer and on non-cancer cells (e.g., endothelial cells, fibroblasts, stromal cells).
[139] As used herein, the terms "neoplasm", "neoplastic disorder", "neoplasia" "cancer," "tumor" and "proliferative disorder" refer to cells having the capacity for autonomous growth/proliferation, i.e. , an abnormal state or condition characterized by rapidly proliferating cell growth which generally forms a distinct mass that show partial or total lack of structural organization and functional coordination with normal tissue. Cancer is meant to include all types of cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness. The terms are meant to encompass hematopoietic cancers (e.g. lymphomas or leukemias) as well as solid cancers (e.g. sarcomas or carcinomas), including all types of pre-cancerous and cancerous growths, or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness. Hematopoietic cancers are malignant tumors affecting hematopoietic structures (structures pertaining to the formation of blood cells) and components of the immune system, including leukemias (related to leukocytes (white blood cells) and their precursors in the blood and bone marrow) arising from myeloid, lymphoid or erythroid lineages, and lymphomas (relates to lymphocytes). Solid cancers include sarcomas, which are malignant cancers that originate from connective tissues such as muscle, cartilage, blood vessels, fibrous tissue, fat or bone. Solid cancers also include carcinomas, which are malignant cancers arising from epithelial structures (including external epithelia (e.g., skin and linings of the gastrointestinal tract, lungs, and cervix), and internal epithelia that line various glands (e.g., breast, pancreas, thyroid). Examples of cancers that are particularly susceptible to treatment by the methods of the invention include leukemia, and hepatocellular cancers, sarcoma, vascular endothelial cancers, breast cancers, central nervous system cancers (e.g. astrocytoma, gliosarcoma, neuroblastoma, oligodendroglioma and glioblastoma), prostate cancers, lung and bronchus cancers, larynx cancers, esophagus cancers, colon cancers, colorectal cancers, gastro- intestinal cancers, melanomas, ovarian and endometrial cancer, renal and bladder cancer, liver cancer, endocrine cancer (e.g. thyroid), and pancreatic cancer.
[140] Other examples of cancers include but are not limited to apudoma, choristoma, branchioma, malignant carcinoid syndrome, carcinoid heart disease, carcinoma (e.g., Walker, basal cell, basosquamous, Brown-Pearce, ductal, Ehrlich tumor, non-small cell lung, oat cell, papillary, bronchiolar, bronchogenic, squamous cell, and transitional cell), histiocytic disorders, leukemia (e.g., B cell, mixed cell, null cell, T cell, T-cell chronic, HTLV-ll-associated, lymphocytic acute, lymphocytic chronic, mast cell, and myeloid), histiocytosis malignant, Hodgkin disease, immunoproliferative small, non-Hodgkin lymphoma, plasmacytoma, reticuloendotheliosis, melanoma, chondroblastoma, chondroma, chondrosarcoma, fibroma, fibrosarcoma, giant cell tumors, histiocytoma, lipoma, liposarcoma, mesothelioma, myxoma, myxosarcoma, osteoma, osteosarcoma, Ewing sarcoma, synovioma, adenofibroma, adenolymphoma, carcinosarcoma, chordoma, craniopharyngioma, dysgerminoma, hamartoma, mesenchymoma, mesonephroma, myosarcoma, ameloblastoma, cementoma, odontoma, teratoma, thymoma, trophoblastic tumor, adeno-carcinoma, adenoma, cholangioma, cholesteatoma, cylindroma, cystadenocarcinoma, cystadenoma, granulosa cell tumor, gynandroblastoma, hepatoma, hidradenoma, islet cell tumor, Leydig cell tumor, papilloma, Sertoli cell tumor, theca cell tumor, leiomyoma, leiomyosarcoma, myoblastoma, myosarcoma, rhabdomyoma, rhabdomyosarcoma, ependymoma, ganglioneuroma, glioma, medulloblastoma, meningioma, neurilemmoma, neuroblastoma, neuroepithelioma, neurofibroma, neuroma, paraganglioma, paraganglioma nonchromaffin, angiokeratoma, angiolymphoid hyperplasia with eosinophilia, angioma sclerosing, angiomatosis, glomangioma, hemangioendothelioma, hemangioma, hemangiopericytoma, hemangiosarcoma, lymphangioma, lymphangiomyoma, lymphangiosarcoma, pinealoma, carcinosarcoma, chondrosarcoma, cystosarcoma, phyllodes, fibrosarcoma, hemangiosarcoma, leimyosarcoma, leukosarcoma, liposarcoma, lymphangiosarcoma, myosarcoma, myxosarcoma, ovarian carcinoma, rhabdomyosarcoma, sarcoma (e.g., Ewing, experimental, Kaposi, and mast cell), neurofibromatosis, and cervical dysplasia, and other conditions in which cells have become immortalized or transformed.
[141] The peptides of the invention can be used to modulate angiogenesis. Diseases or disorders that are mediated by angiogenesis (or associated with angiogenesis) include tumors, ocular disorders, dermatological disorders, and malignant or metastatic conditions, inflammatory diseases, osteoporosis and other conditions mediated by accelerated bone resorption, restenosis, inappropriate platelet activation, recruitment, or aggregation, thrombosis, or a condition requiring tissue repair or wound healing. [142] Among the ocular disorders that can be treated according to the present invention are eye diseases characterized by ocular neovascularization including, but not limited to, diabetic retinopathy (a major complication of diabetes), retinopathy of prematurity (this devastating eye condition, that frequently leads to chronic vision problems and carries a high risk of blindness, is a severe complication during the care of premature infants), neovascular glaucoma, retinoblastoma, retrolental fibroplasia, rubeosis, uveitis, macular degeneration, and corneal graft neovascularization. Other eye inflammatory diseases, ocular tumors, and diseases associated with choroidal or iris neovascularization can also be treated according to the present invention.
[143] Alternatively, the disorder associated with or mediated by angiogenesis is arteriosclerosis, arthritis, psoriasis or endometriosis.
[144] The peptides of the present invention can also be used to treat inflammatory diseases including, but not limited to, arthritis, rheumatism, inflammatory bowel disease, and psoriasis.
[145] Among the cancers that can be treated with the peptides of the invention are seminomas, melanomas, teratomas, gliomas, colon cancer, rectal cancer, kidney cancer, breast cancer, prostate cancer, cancer of the uterus, ovarian cancer, endometrial cancer, cancer of the esophagus, blood cancer, liver cancer, pancreatic cancer, skin cancer, brain cancer and lung cancer, lymphomas, astrocytoma, neuroblastoma, glioblastoma, mesothelioma.
[146] In one embodiment, the peptides of the invention, including peptide antagonists of the ionic channels subtype and/or target selectivity, can be used to treat epilepsy, memory, learning, neuropsychiatric, neurological, neuromuscular, and immunological disorders, schizophrenia, bipolar disorder, sleep apnea, neurodegeneration, smooth muscle disorders. In other embodiments, the peptides of the invention can be used as antibacterial peptides, antifungal peptides antimalarial peptides antiviral peptides, bradykinin-potentiating peptides, autoimmunity targeting potassium channels blockers, immuno-modulators, and analgesic peptides .
[147] Other diseases and conditions that can be treated according to the present invention include benign tumors and preneoplastic conditions, myocardial angiogenesis, hemophilic joints, scleroderma, vascular adhesions, asthma and allergy, eczema and dermatitis, graft versus host disease, sepsis, adult respiratory distress syndrome, telangiectasia, and wound granulation.
[148] In one embodiment, the invention provides peptide RK (a 17 mer), which is the first scorpion peptide with dual disintegrin activity on a1 b1 and anb3 integrins. In one embodiment, the peptide of the invention may act by interacting with a1 b1 and/or anb3 integrins. Disintegrins can be used to inhibit biological processes such as platelet aggregation (can be used as antithrombotic agents for use in thrombolytic therapy by enhancing and sustaining arterial thrombolysis in conjunction with recombinant tissue plasminogen activator), cell growth, adhesion, metastasis, and neovascularization.
[149] The peptides of the invention can be used for the treatment and prevention of anb3 integrins associated diseases in a mammal, which include osteoporosis, bone tumor or cancer growth, angiogenesis-related tumor growth and metastasis, tumor metastasis in bone, malignancy-induced hypercalcemia, angiogenesis-related eye diseases, Paget's disease, rheumatic arthritis, ovariectomy-induced physiological change, inflammation, coagulation diseases, and osteoarthritis. The angiogenesis-related eye diseases include age-related macular degeneration, diabetic retinopathy, corneal neovascularizing diseases, ischaemia-induced neovascularizing retinopathy, high myopia, and retinopathy of prematurity. Disintegrings block adhesive functions and act as platelet aggregation inhibitors.
[150] The peptides of the invention can also be use in the diagnosis of cardiovascular diseases and as therapeutic agents in arterial thrombosis.
[151] In addition, homodimeric, heterodimeric, and monomeric disintegrins/peptides of the disclosure may be used to modulate the adhesion, motility, and invasiveness of integrin- expressing tumor cells. When formulated as a pharmaceutically acceptable composition, such proteins can be used to treat patients by inhibiting or disrupting disease processes associated with a ligand binding to a1 b1 and anb3 integrins.
[152] Homodimeric, heterodimeric, and monomeric peptide disintegrins described herein (e.g. RK) may be used in methods to decrease the motility of an cd b1 and/or anb3 integrins expressing cell, the method comprising cross-linking at least two a1 b1 and anb3 integrins on the integrin expressing cells thereby inhibiting the motility of said cells. Such crosslinking is believed to disrupt FAK signaling and activates tyrosine phosphorylation of FAK and CAS. Moreover, the crosslinking is believed to induce an alteration in cell morphology, including changes of cytoskeletal or focal adhesion structures. In a preferred embodiment, the a1 b1 and anb3 integrins integrin expressing cell is a tumor cell.
[153] Homodimeric, heterodimeric, and monomeric disintegrins described herein (e.g. RK) may be used to inhibit the adhesion of integrin expressing cells to vitronectin by exposing the cells to the homodimeric and monomeric disintegrin. The homodimeric and monomeric disintegrin is believed to inhibits adhesion by binding to an integrin, in particular a1 b1 and anb3 integrins.
[154] Homodimeric, heterodimeric, and monomeric disintegrins described herein (e.g. RK) may be formulated as compositions for the treatment of thrombotic diseases in mammals, alone or in conjunction with one or more thrombolytic agents. In particular, such compositions have utility in treating or preventing arterial, venous and microvascular thrombosis and thromboembolism. Such compositions also have utility in treating stroke, transient ischemic attacks, arteriosclerosis, atherosclerosis, pulmonary embolism, aneurisms and angina. In particular, such compositions have utility in preventing or treating myocardial infarctions.
[155] Homodimeric, heterodimeric, and monomeric disintegrins described herein (e.g. RK) may be used to inhibit metastasis in melanoma, carcinoma and sarcoma patients. In particular embodiments. Homodimeric, heterodimeric, and monomeric disintegrins may be used to prevent metastasis in breast cancer patients and other cancers.
[156] Homodimeric, heterodimeric, and monomeric disintegrins described herein (e.g. RK) may be used to treat osteoporosis. Compositions and methods for treatment of osteoporosis employing an amount of a homodimeric and monomeric disintegrin effective to inhibit bone resorption by osteoclasts may be used.
[157] Homodimeric, heterodimeric, and monomeric disintegrins described herein (e.g. RK) may be used to promote wound healing. Homodimeric, heterodimeric, and monomeric disintegrins may inhibit cell-cell and cell-extracellular matrix interactions (including interaction with fibronectin), thus promoting wound repair, including keloid formation. Compositions containing homodimeric, heterodimeric, and monomeric disintegrins may be used to prevent adhesion formation when administered to a patient in need of such treatment.
[158] In one embodiment, the peptides of the invention are not able to alter cell adhesion through a2b1 , anb6, anb5, a5b1 and a6b4 integrins, but significantly reduce the adhesive activity of a1 b1 and anb3 integrins to extracellular matrix proteins. In one embodiment, the extracellular matrix protein comprises collagen. In other embodiments, the extracellular matrix protein comprises vitronectin, fibronectin, fibrinogen, osteopoietin, and/or bone sialoprotein. In one embodiment, the activity is reduced by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 90%, or 100%.
[159] The present invention also relates to RK and RK1 , as well as peptide analogs of RK or RK1 , as peptides with novel functional properties such as greater ionic channels antagonist activity and/or target selectivity. Molecules that recognize certain targets, specifically with high affinity to different receptors, are useful for many clinical (e.g., diagnostic and/or therapeutic) and biotechnological applications.
[160] In one embodiment, the peptides of the invention can be used to inhibit the proliferation and/or migration of a cancer cell, or cancer cell line. As used herein, the term "cancer cell line" or "cancer cells" may be used interchangeably and refers to cells isolated from a tumor, metastasis or abnormal growth derived from an animal or human. These cells typically, but not always, grow rapidly in culture when supplemented with appropriate growth factors, often fetal animal serum. This term also refers to normal cells transformed into cells that display typical features of cancer cells, i.e. they divide in cell culture under trophic support and/or form tumors when administered to animals.
Cancer cell lines may include, but are not limited to, human prostate cancer cells (e.g., PC-3, LNCaP, DU-145), human mammary epithelial cells (e.g. MCF-7, MCF-10A, MDA-MB-438, MDA- 231 , MDA-468, T47D, SkBr3), human neuronal cells (e.g., M17, SHSY5Y, H4, U87), human acute myeloid leukemia cells (e.g., THP-1), human bone cancer cells (e.g., Saos-2 cells), human melanoma cells (e.g., 721), human glioblastoma cells (e.g., A172), human head and neck carcinoma cells (e.g., A253), human skin epithelial cells (e.g., A431), human lung carcinoma epithelial cells (e.g., A- 549), human peripheral blood mononuclear cell lymphoma (e.g., BCP-1), human pancreatic adenocarcinoma cells (e.g., BxPC3), human squamous cell carcinoma (e.g., Cal-27), human CML acute phase T cell leukemia cells (e.g., CML T1), human CML blast crisis Ph+ CML cells e.g., (EM2), human CML blast crisis Ph+CML cells (e.g., EM3), human metastatic lymph node melanoma cells e.g., (FM3), human lung cancer cells (e.g., H1299), human hybridoma cells (e.g., HB54), human fibroblasts (e.g., HCA2), human kidney embryonic epithelial cell (e.g., HEK-293), human cervical cancer epithelial cell (e.g., HeLa), human myeloblast blood cells (e.g., HL-60), human mammary epithelial cells (e.g., HMEC), human colon epithelial adenocarcinoma cells (e.g., HT-29), human umbilical cord vein endothelial cells (e.g., HUVEC), human T-cell-leukemia white blood cells (e.g., Jurkat), human lymphoblastoid EBV immortalized B cells (e.g., JY cells), human lymphoblastoid CML blast crisis cells (e.g., K562 cells), human lymphoblastoid erythroleukemia cells (e.g., Ku812), human lymphoblastoid CML cells (e.g., KCL22), human lymphoblastoid AML cells (e.g., KG1), human lymphoblastoid CML cells (e.g., KY01), human melanoma cells (e.g., Ma-Mel 1 , 2, 3 through 48), human WBC myeloid metaplasic AML cells (e.g., MONO-MAC 6), human T cell leukemia (e.g., Peer), human osteosarcoma cells (e.g., Saos-2), human T cell leukemia/B cell line hybridoma (e.g., T2), human colorectal carcinoma/lung metastasis epithelium cells (e.g., T84), human colorectal adenocarcinoma cells (e.g., HCT-15, HT-29), human monocyte AML cells (e.g., THP1), human glioblastoma- astrocytoma epithelial cells (e.g., U373), human glioblastoma-astrocytoma epithelial-like cells (e.g., U87), human leukemic monocytic lymphoma cells (e.g., U937), human lymphoblastoid cells (e.g., WT-49), human B-cell EBV transformed cells (e.g., YAR), human breast adenocarcinoma cells (e.g., NC1/ADR-RES, MDA-MB-231), human CNS glioblastoma cells (e.g., SF-268), human ovary adenocarcinoma cells (e.g., SK-OV-3), human lung carcinoma cells (e.g., NCIH460), human lung adenocarcinoma cells (e.g., A549), human liver carcinoma cells (e.g., Hep3B), human uterine sarcoma-drug sensitive cells (e.g., MES-SA), human uterine sarcoma--drug resistant cells (e.g., MES-SA/DX5), human skin primary melanoma cells (e.g., WM39), ape- kidney fibroblast cells (e.g., COS-7), African green monkey kidney epithelial cells (e.g., Vero cells), murine brain/cerebral cortex endothelial cells (e.g., bEnd.3), murine embryonic mesenchymal cells (e.g., C3H-10T1/2), murine T cell leukemia ECACC cells (e.g., EL4), murine embryonic fibroblasts (e.g., NIH-3T3), murine embryonic calvarial cells (e.g., MC3T3), murine hepatoma epithelial cell (e.g., Hepal cic7), murine adenocarcinoma cells (e.g., MC-38), murine epithelial cells (e.g., MTD-1A), murine endothelial cells (e.g., MyEnd), murine renal carcinoma cells (e.g., RenCa), murine melanoma cells (e.g., X63), murine lymphoma cells (e.g., YAC-1), murine T cell tumor cells (e.g., RMA/RMAS), murine breast adenocarcinoma cells (e.g., 4T1), murine mammary normal epithelial cells (e.g., NmuMG), rat glioblastoma cells (e.g., 9L), rat neuroblastoma cells (e.g., B35), canine mammary tumor cells (e.g., CMT), canine osteosarcoma cells (e.g., D17), canine histiocytosismonocyte/macrophages (e.g., DH82), rat pheochromocytoma cells (e.g., PC-12), rat pituitary tumor (e.g., GH3), canine kidney epithelial cells (e.g., MDCK II), murine B lymphoma B cells (e.g., lymphocyte A20), murine bone marrow stromal cells (e.g., ALC), murine melanoma cells (e.g., B16), murine colorectal carcinoma cells (e.g., CT26), baby hamster kidney fibroblasts (e.g., BHK-21), Asian tiger mosquito larval tissue (e.g., C6/36), insect-ovary cells (e.g., Sf-9), Chinese hamster ovary cells (e.g., CHO), onyvax prostate cancer cells (e.g., OPCN, OPCT), tobacco cells (e.g., BY-2), zebrafish cells (e.g., ZF4 and AB9), Madin-Darby Canine Kidney (e.g., MDCK) epithelial cells, Xenopus kidney epithelial cells (e.g., A6).
[161] The peptides of the invention can be administered alone or in combination with other drugs (e.g., as an adjuvant) or therapeutic agents. Such additional therapeutic agent may comprise any active ingredients suitable for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. For example, numerous chemotherapeutics, cytotoxic and/or cytostatic agents, especially antineoplastic drugs, immunomodulatory agents are available for combination with the peptides of the invention. In a particular embodiment, the additional therapeutic agent is an anti-cancer agent, for example a microtubule disruptor, an antimetabolite, a topoisomerase inhibitor, a DNA intercalator, an alkylating agent, a hormonal therapy, a kinase inhibitor, a receptor antagonist, an activator of tumor cell apoptosis, or an antiangiogenic agent.
[162] As used herein, adjunctive or combined administration (co-administration) includes simultaneous administration of the compounds in the same or different dosage form, or separate administration of the compounds (e.g., sequential administration). For example, the peptides of the invention can be simultaneously administered with a kinase inhibitor, wherein both the compound and the kinase inhibitor are formulated together. Alternatively, the compound can be administered in combination with kinase inhibitor, wherein both the compound and the kinase inhibitor are formulated for separate administration and are administered concurrently or sequentially. For example, the kinase inhibitor can be administered first followed by (e.g., immediately followed by) the administration of the compound of the disclosure, or vice versa. Such concurrent or sequential administration beneficially results in both the compound of the disclosure and kinase inhibitor being simultaneously present in treated patients.
[163] The cytotoxic and/or cytostatic agents, and the anti-angiogenesis agents, may be any agents known to inhibit angiogenesis, the growth and/or replication of and/or kill cells, and in particular the growth/replication of cancer and/or tumor cells. Numerous agents having cytotoxic and/or cytostatic properties are known in the literature. Non-limiting examples of classes of cytotoxic and/or cytostatic agents include, by way of example and not limitation, radionuclides, alkylating agents, DNA cross-linking agents, DNA intercalating agents (e.g., groove binding agents such as minor groove binders), cell cycle modulators, apoptosis regulators, kinase inhibitors, protein synthesis inhibitors, mitochondria inhibitors, nuclear export inhibitors, topoisomerase I inhibitors, topoisomerase II inhibitors, RNA/DNA antimetabolites and antimitotic agents.
[164] Specific non-limiting examples of agents within certain of these various classes are provided below.
[165] Alkylating Agents: asaley (L-Leucine, N-[N-acetyl-4-[bis-(2-chloroethyl)amino]-
DL-phenylalanyl]-, ethylester); AZQ (1 ,4-cyclohexadiene-1 ,4-dicarbamic acid, 2, 5-bis(1- aziridinyl)-3,6-dioxo-, diethyl ester); BCNU (N,N'-Bis(2-chloroethyl)-N-nitrosourea); busulfan (1 ,4- butanediol dimethanesulfonate); (carboxyphthalato)platinum; CBDCA (cis-(1 ,1- cyclobutanedicarboxylato)diammineplatinum(ll))); CCNU (N-(2-chloroethyl)-N'-cyclohexyl-N- nitrosourea); CHIP (iproplatin; NSC 256927); chlorambucil; chlorozotocin (2-[[[(2-chloroethyl) nitrosoamino]carbonyl]amino]-2-deoxy-D-glucopyranose); c/s-platinum (cisplatin); clomesone; cyanomorpholinodoxorubicin; cyclodisone; dianhydrogalactitol (5,6-diepoxydulcitol); fluorodopan ((5-[(2-chloroethyl)-(2-fluoroethyl)amino]-6-methyl-uracil); hepsulfam; hycanthone; indolinobenzodiazepine dimer DGN462; melphalan; methyl CCNU ((1-(2-chloroethyl)-3-(trans-4- methylcyclohexane)-1 -nitrosourea); mitomycin C; mitozolamide; nitrogen mustard ((bis(2- chloroethyl) methylamine hydrochloride); PCNU ((1-(2-chloroethyl)-3-(2,6-dioxo-3-piperidyl)-1- nitrosourea)); piperazine alkylator ((1-(2-chloroethyl)-4-(3-chloropropyl)-piperazine dihydrochloride)); piperazinedione; pipobroman (N,N'-bis(3-bromopropionyl) piperazine); porfiromycin (N-methylmitomycin C); spirohydantoin mustard; teroxirone (triglycidylisocyanurate); tetraplatin; thio-tepa (N,N’,N”-tri-1 ,2-ethanediylthio phosphoramide); triethylenemelamine; uracil nitrogen mustard (desmethyldopan); Yoshi-864 ((bis(3-mesyloxy propyl)amine hydrochloride).
[166] DNA Alkylating-like Agents: Cisplatin; Carboplatin; Nedaplatin; Oxaliplatin; Satraplatin; Triplatin tetranitrate; Procarbazine; altretamine; dacarbazine; mitozolomide; temozolomide.
[167] Alkylating Antineoplastic Agents: Carboquone; Carmustine; Chlornaphazine; Chlorozotocin; Duocarmycin; Evofosfamide; Fotemustine; Glufosfamide; Lomustine; Mannosulfan; Nimustine; Phenanthriplatin; Pipobroman; Ranimustine; Semustine; Streptozotocin; ThioTEPA; Treosulfan; Triaziquone; Triethylenemelamine; Triplatin tetranitrate.
[168] DNA replication and repair inhibitors: Altretamine; Bleomycin; Dacarbazine; Dactinomycin; Mitobronitol; Mitomycin; Pingyangmycin; Plicamycin; Procarbazine; Temozolomide; ABT-888 (veliparib); olaparib; KU-59436; AZD-2281 ; AG-014699; BSI-201 ; BGP- 15; INO-1001 ; ONO-2231.
[169] Cell Cycle Modulators: Paclitaxel; Nab-Paclitaxel; Docetaxel; Vincristine; Vinblastine; ABT-348; AZD-1 152; MLN-8054; VX-680; Aurora A-specific kinase inhibitors; Aurora B-specific kinase inhibitors and pan-Aurora kinase inhibitors; AZD-5438; BMI-1040; BMS-032; BMS-387; CVT-2584; flavopyridol; GPC-286199; MCS-5A; PD0332991 ; PHA-690509; seliciclib (CYC-202, R-roscovitine); ZK-304709; AZD4877, ARRY-520; GSK923295A.
[170] Apoptosis Regulators: AT-101 ((-)gossypol); G3139 or oblimersen (Bcl-2- targeting antisense oligonucleotide); I PI-194; I PI-565; N-(4-(4-((4'-chloro(1 , T-biphenyl)-2- yl)methyl)piperazin-1-ylbenzoyl)-4-(((1 R)-3-(dimethylamino)-1-
((phenylsulfanyl)methyl)propyl)amino)-3-nitrobenzenesulfonamide); N-(4-(4-((2-(4- chlorophenyl)-5,5-dimethyl-1-cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoyl)-4-(((1 R)-3-
(morpholin-4-yl)-1-((phenylsulfanyl)methyl)propyl)amino)-3-
((trifluoromethyl)sulfonyl)benzenesulfonamide; GX-070 (Obatoclax®; 1 H-lndole, 2-(2-((3,5- dimethyl-1 H-pyrrol-2-yl)methylene)-3-methoxy-2H-pyrrol-5-yl)-)); HGS1029; GDC-0145; GDC- 0152; LCL-161 ; LBW-242; venetoclax; agents that target TRAIL or death receptors (e.g., DR4 and DR5) such as ETR2-ST01 , GDC0145, HGS-1029, LBY-135, PRO-1762; drugs that target caspases, caspase-regulators, BCL-2 family members, death domain proteins, TNF family members, Toll family members, and/or NF-kappa-B proteins.
[171] Angiogenesis Inhibitors: ABT-869; AEE-788; axitinib (AG-13736); AZD-2171 ; CP-547,632; IM-862; pegaptamib; sorafenib; BAY43-9006; pazopanib (GW-786034); vatalanib (PTK-787, ZK-222584); sunitinib; SU-11248; VEGF trap; vandetanib; ABT-165; ZD-6474; DLL4 inhibitors.
[172] Proteasome Inhibitors: Bortezomib; Carfilzomib; Epoxomicin; Ixazomib; Salinosporamide A.
[173] Kinase Inhibitors: Afatinib; Axitinib; Bosutinib; Crizotinib; Dasatinib; Erlotinib; Fostamatinib; Gefitinib; Ibrutinib; Imatinib; Lapatinib; Lenvatinib; Mubritinib; Nilotinib; Pazopanib; Pegaptanib; Sorafenib; Sunitinib; SU6656; Vandetanib; Vemurafenib; CEP-701 (lesaurtinib); XL019; INCB018424 (ruxolitinib); ARRY-142886 (selemetinib); ARRY-438162 (binimetinib); PD- 325901 ; PD-98059; AP-23573; CCI-779; everolimus; RAD-001 ; rapamycin; temsirolimus; ATP- competitive TORC1/TORC2 inhibitors including PI-103, PP242, PP30, Torin 1 ; LY294002; XL- 147; CAL-120; ONC-21 ; AEZS-127; ETP-45658; PX-866; GDC-0941 ; BGT226; BEZ235; XL765.
[174] Protein Synthesis Inhibitors: Streptomycin; Dihydrostreptomycin; Neomycin; Framycetin; Paromomycin; Ribostamycin; Kanamycin; Amikacin; Arbekacin; Bekanamycin; Dibekacin; Tobramycin; Spectinomycin; Hygromycin B; Paromomycin; Gentamicin; Netilmicin; Sisomicin; lsepamicin;Verdamicin; Astromicin; Tetracycline; Doxycycline; Chlortetracycline; Clomocycline; Demeclocycline; Lymecycline; Meclocycline; Metacycline; Minocycline; Oxytetracycline; Penimepicycline; Rolitetracycline; Tetracycline; Glycylcyclines;Tigecycline; Oxazolidinone; Eperezolid; Linezolid; Posizolid; Radezolid; Ranbezolid; Sutezolid; Tedizolid; Peptidyl transferase inhibitors; Chloramphenicol; Azidamfenicol; Thiamphenicol; Florfenicol; Pleuromutilins; Retapamulin; Tiamulin; Valnemulin; Azithromycin; Clarithromycin; Dirithromycin; Erythromycin; Flurithromycin; Josamycin; Midecamycin; Miocamycin; Oleandomycin; Rokitamycin; Roxithromycin; Spiramycin; Troleandomycin; Tylosin; Ketolides; Telithromycin; Cethromycin; Solithromycin; Clindamycin; Lincomycin; Pirlimycin; Streptogramins; Pristinamycin; Quinupristin/dalfopristin; Virginiamycin.
[175] Histone deacetylase inhibitors.: Vorinostat; Romidepsin; Chidamide; Panobinostat; Valproic acid; Belinostat; Mocetinostat; Abexinostat; Entinostat; SB939 (pracinostat); Resminostat; Givinostat; Quisinostat; thioureidobutyronitrile (Kevetrin™); CUDC- 10; CHR-2845 (tefinostat); CHR-3996; 4SC-202; CG200745; ACY-1215 (rocilinostat); ME-344; sulforaphane.
[176] Topoisomerase I Inhibitors: camptothecin; various camptothecin derivatives and analogs (for example, NSC 100880, NSC 603071 , NSC 107124, NSC 643833, NSC 629971 , NSC 295500, NSC 249910, NSC 606985, NSC 74028, NSC 176323, NSC 295501 , NSC 606172, NSC 606173, NSC 610458, NSC 618939, NSC 610457, NSC 610459, NSC 606499, NSC 610456, NSC 364830, and NSC 606497); morpholinisoxorubicin; SN-38. [177] Topoisomerase II Inihibitors: doxorubicin; amonafide (benzisoquinolinedione); m-AMSA (4'-(9-acridinylamino)-3'-methoxymethanesulfonanilide); anthrapyrazole derivative ((NSC 355644); etoposide (VP-16); pyrazoloacridine ((pyrazolo[3,4,5-kl]acridine-2(6H)- propanamine, 9-methoxy-N, N-dimethyl-5-nitro-, monomethanesulfonate); bisantrene hydrochloride; daunorubicin; deoxydoxorubicin; mitoxantrone; menogaril; N,N-dibenzyl daunomycin; oxanthrazole; rubidazone; teniposide.
[178] DNA Intercalating Agents: anthramycin; chicamycin A; tomaymycin; DC-81 ; sibiromycin; pyrrolobenzodiazepine derivative; SGD-1882 ((S)-2-(4-aminophenyl)-7-methoxy-8- (3-(((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-5,1 1 a-dihydro-1 H-benzo[e]pyrrolo[1 ,2- a][1 ,4]diazepin-8-yl)oxy)propoxy)-1 H-benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepin-5(11 aH)-one); SG2000 (SJG-136; (11 aS, 1 1 a'S)-8,8'-(propane-1 ,3-diylbis(oxy))bis(7-methoxy-2-methylene-2,3-dihydro- 1 H-benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepin-5(11 aH)-one)).
[179] RNA/DNA Antimetabolites: L-alanosine; 5-azacytidine; 5-fluorouracil; acivicin; aminopterin derivative N-[2-chloro-5-[[(2, 4-diamino-5-methyl-6- quinazolinyl)methyl]amino]benzoyl] L-aspartic acid (NSC 132483); aminopterin derivative N-[4- [[(2, 4-diamino-5-ethyl-6-quinazolinyl)methyl]amino]benzoyl] L-aspartic acid; aminopterin derivative N-[2-chloro-4-[[(2, 4-diamino-6-pteridinyl)methyl] amino]benzoyl] L-aspartic acid monohydrate; antifolate PT523 ((Na-(4-amino-4-deoxypteroyl)-NY-hemiphthaloyl-L-ornithine)); Baker's soluble antifol (NSC 139105); dichlorallyl lawsone ((2-(3, 3-dichloroallyl)-3-hydroxy-1 ,4- naphthoquinone); brequinar; ftorafur ((pro-drug; 5-fluoro-1-(tetrahydro-2-furyl)-uracil); 5,6- dihydro-5-azacytidine; methotrexate; methotrexate derivative (N-[[4-[[(2, 4-diamino-6- pteridinyl)methyl]methylamino]-1-naphthalenyl]carbonyl ] L-glutamic acid); PALA ((N- (phosphonoacetyl)-L-aspartate); pyrazofurin; trimetrexate.
[180] DNA Antimetabolites: 3-HP; 2'-deoxy-5-fluorouridine; 5-HP; a-TGDR (a-2’- deoxy-6-thioguanosine); aphidicolin glycinate; ara C (cytosine arabinoside); 5-aza-2'- deoxycytidine; b-TGDR (P-2’-deoxy-6-thioguanosine); cyclocytidine; guanazole; hydroxyurea; inosine glycodialdehyde; macbecin II; pyrazoloimidazole; thioguanine; thiopurine.
[181] Mitochondria Inhibitors: pancratistatin; phenpanstatin; rhodamine-123; edelfosine; d-alpha-tocopherol succinate; compound 11□; aspirin; ellipticine; berberine; cerulenin; GX015-070 (Obatoclax®; 1 H-lndole, 2-(2-((3,5-dimethyl-1 H-pyrrol-2-yl)methylene)-3- methoxy-2H-pyrrol-5-yl)-); celastrol (tripterine); metformin; Brilliant green; ME-344.
[182] Antimitotic Agents: allocolchicine; auristatins, such as MMAE (monomethyl auristatin E) and MMAF (monomethyl auristatin F); halichondrin B; cemadotin; colchicine; cholchicine derivative (N-benzoyl-deacetyl benzamide); dolastatin-10; dolastatin-15; maytansine; maytansinoids, such as DM1 (A/2'-deacetyl-/\/2'-(3-mercapto-1-oxopropyl)-maytansine); rhozoxin; paclitaxel; paclitaxel derivative ((2'-N-[3-(dimethylamino)propyl]glutaramate paclitaxel); docetaxel; thiocolchicine; trityl cysteine; vinblastine sulfate; vincristine sulfate.
[183] Nuclear Export Inhibitors: callystatin A; delactonmycin; KPT-185 (propan-2-yl (Z)-3-[3-[3-methoxy-5-(trifluoromethyl)phenyl]-1 ,2,4-triazol-1-yl]prop-2-enoate); kazusamycin A; leptolstatin; leptofuranin A; leptomycin B; ratjadone; Verdinexor ((Z)-3-[3-[3,5- bis(trifluoromethyl)phenyl]-1 ,2,4-triazol-1-yl]-N'-pyridin-2-ylprop-2-enehydrazide).
[184] Hormonal Therapies: anastrozole; exemestane; arzoxifene; bicalutamide; cetrorelix; degarelix; deslorelin; trilostane; dexamethasone; flutamide; raloxifene; fadrozole; toremifene; fulvestrant; letrozole; formestane; glucocorticoids; doxercalciferol; sevelamer carbonate; lasofoxifene; leuprolide acetate; megesterol; mifepristone; nilutamide; tamoxifen citrate; abarelix; prednisone; finasteride; rilostane; buserelin; luteinizing hormone releasing hormone (LHRH); Histrelin; trilostane or modrastane; fosrelin; goserelin.
[185] In one embodiment, an antiangiogenic agent, a cytotoxic agent, a cytostatic agent comprises at least one agent chosen from endostatin, angiostatin, VEGF inhibitors, cytotoxic agents, alkaloids, antimetabolites, cancer growth inhibitors, gene therapy therapeutics, cancer vaccines, interferons, monoclonal antibodies, radiotherapy, hormonal therapy, and other supportive therapy.
[186] In one embodiment, the chemotherapy drugs are chosen from amsacrine, bleomycin, busulfan, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, crisantaspase, cycolophosphamide, cytarabine, dacarbazine, dactinomycine, daunorubicin, docetaxel, doxorubicin, epirubicin, etoposide, fludarabine, fluorouracil, gemcitabine, gliadel implants, hydroxycarbamide, idarubicin, ifosfamide, irinotecan, leucovorin, liposomal doxorubicin, liposomal daunorubicin, lomustine, melphalan, mercaptopurine, mesna, methotrexate, mitomycin, mitoxantrone, oxaliplatin, paclitaxel, pemetrexed, pentostatin, procarbazine, raltitrexed, streptozocin, teg afur- uracil, temozolomide, teniposide, thiotepa, tioguanine, topotecan, treosulfan, vinblastine, vincristine, vindesine, and vinorelbine.
[187] When the peptides are used in the treatment of an infectious disease, the peptides can be administered in combination with at least one anti-bacterial agent or at least one anti-viral agent. In this respect, the anti-bacterial agent can be any suitable antibiotic known in the art. The anti-viral agent can be any vaccine of any suitable type that specifically targets a particular virus (e.g., live-attenuated vaccines, subunit vaccines, recombinant vector vaccines, and small molecule anti-viral therapies (e.g., viral replication inhibitors and nucleoside analogs). [188] In one embodiment, the peptides of the disclosure can be used to inhibit cancer/tumor cell proliferation, migration, and/or adhesion in vivo.
[189] In one embodiment, the peptides of the disclosure are used to inhibit tumor cell proliferation. The utility of the peptides of the present invention in inhibiting tumor cell proliferation in vivo can be illustrated, for example, by their activity in vitro in the in vitro tumor cell proliferation assays described below or known in the art. The link between activity in tumor cell proliferation assays in vitro and anti-tumor activity in the clinical setting has been very well established in the art. For example, the therapeutic utility of taxol, taxotere, and topoisomerase inhibitors were first demonstrated with the use of in vitro tumor proliferation assays. In addition, in vivo xenograft tumor models simulate biological activity observed in humans by grafting relevant and well characterized human primary tumors or tumor cell lines into immune-deficient nude mice. Studies on treatment of tumor xenograft mice with anti-cancer reagents have provided valuable information regarding in vivo efficacy of the tested reagents.
[190] In one embodiment, the peptides of the invention are used in a method of inhibiting tumor cell proliferation by contacting the tumor cell with an effective amount of the peptide. In one embodiment, the tumor cells are glioblastoma cells. In another embodiment, the tumor cells are melanoma cells. In another embodiment, the tumor cells are cells of the tumors listed elsewhere in the specification. In one embodiment, the inhibition is in vitro. In another embodiment, the inhibition is in vivo. The peptide can be used alone or in combination with other drugs or therapeutic agents, many of which are exemplified elsewhere in the specification.
[191] In one embodiment, the peptides of the invention are used in a method of inhibiting tumor cell migration by contacting the cancer cells with an effective amount of the peptide. In one embodiment, the tumor cells are glioblastoma cells. In another embodiment, the tumor cells are melanoma cells. In another embodiment, the tumor cells are cells of the tumors listed elsewhere in the specification. In one embodiment, the inhibition is in vitro. In another embodiment, the inhibition is in vivo. The peptide can be used alone or in combination with other drugs or therapeutic agents, many of which are exemplified elsewhere in the specification.
[192] In one embodiment, the peptides of the invention are used in a method of inhibiting tumor cell adhesion by contacting the tumor cell with an effective amount of the peptide. In one embodiment, the tumor cells are glioblastoma cells. In another embodiment, the tumor cells are melanoma cells. In another embodiment, the tumor cells are cells of the tumors listed elsewhere in the specification. In one embodiment, the inhibition is in vitro. In another embodiment, the inhibition is in vivo. The peptide can be used alone or in combination with other drugs or therapeutic agents, many of which are exemplified elsewhere in the specification. [193] In one embodiment, the peptides of the invention can be used to inhibit non cancer cell growth/proliferation, migration, and/or adhesion. In one embodiment, the cells are endothelial cells. In one embodiment, the cells are
[194] The dose of the peptide administered to achieve an effective amount may vary depending upon the disorder being treated or the type of tumor cell to be inhibited. In alternate embodiments, a dosage to be achieved in vivo would be equivalent to an in vitro level of greater than 10-12 M, or 10 11 M, or 10 10 M, or 10 9 M, or 108 M, or 107 M, or 106 M, or 105 M. Thus, a dosage to be achieved in vivo may be equivalent to an in vitro level of 10 12 M to 105 M, or 10 11 M to 106 M, or 10 10 M to 107 M, or 109 M to 107 M or ranges therein. In alternate embodiments, the dosage used may be equivalent to an in vitro level of about 1-10000 ngml 1 , or about 10-5000 ngml 1, or about 100-1000 ngml 1.
[195] In some embodiments, the dosage to be achieved in vivo may be equivalent to an in vitro level of 1 mM, 2 pM, 3 pM, 4 pM, 5 pM, 6 pM, 7 pM, 1 pM, 8 pM, 9 pM, 10 pM, 20 pM, 30 pM, 40 pM, 50 pM, 70 pM, 80 pM, 90 pM, or 100 pM. The equivalence between in vitro and in vivo levels is calculated or estimated by methods known to one of ordinary skill in the art. In some embodiments, these are dosages appropriate for inhibiting cell (tumor cell, endothelial cell, fibroblast, or any other tissue cell) proliferation. In other embodiments, these are dosages appropriate for inhibiting cell migration. In other embodiments, these are dosages appropriate for inhibiting angiogenesis. In some embodiments, the inhibition is about 10%. In other embodiments, the inhibition is about 20%. In other embodiments, the inhibition is about 30%. In other embodiments, the inhibition is about 40%. In other embodiments, the inhibition is about 50%. In other embodiments, the inhibition is about 60%. In other embodiments, the inhibition is about 70%. In other embodiments, the inhibition is about 80%. In other embodiments, the inhibition is about 90%. In other embodiments, the inhibition is about 100%.
[196] In some embodiments, the dosage achieved is a blood/plasma level of at least 1 pM, 2 pM, 3 pM, 4 pM, 5 pM, 6 pM, 7 pM, 1 pM, 8 pM, 9 pM, 10 pM, 20 pM, 30 pM, 40 pM, 50 pM, 70 pM, 80 pM, 90 pM, or 100 pM. In some embodiments, the blood level is 2-5 pM (e.g., from 2 to 5), 5-10 pM, 10-100 pM, 100-1000 pM, 50-60 pM, 60-70 pM, 70-80 pM, 80-90 pM, 90-95 pM, 95-99 pM, 99-100 pM. In some embodiments, the blood level is 1-2 pM, 2-3 pM, 3-4 pM, 4-5 pM, 5-6 pM, 6-7 pM, 7-8 pM, 8-9 pM, 9-10 pM, 10-20 pM, and all the other ranges between the values set in this paragraph.
[197] In one embodiment, this level or plasma Tmax is achieved after 1 minute, no more than 2 minutes, no more than 3 minutes, no more than 4 minutes, no more than 5 minutes, no more than 8 minutes, no more than 10 minutes, no more than 12 minutes, no more than 15 minutes, no more than 20 minutes, no more than 30 minutes, no more than 35 minutes, no more than 40 minutes, no more than 45 minutes, no more than 50 minutes, no more than 55 minutes, is between 2 minutes and 30 minutes, or is no more than 60 minutes after one administration. In one embodiment, this level or Tmax is achieved after 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 1 1 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 22 hours, 23 hours, or 24 hours. In one embodiment, this level or other desired level or plasma Tmax is achieved within 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 22 days, 23 days, or 24 days.
[198] One exemplary dosage of the peptide of the invention would be in the range from about 0.005 mg/kg to about 10 mg/kg. In other non-limiting examples, a dose may also comprise from about 1 microgram/kg body weight, about 5 microgram/kg body weight, about 10 microgram/kg body weight, about 50 microgram/kg body weight, about 100 microgram/kg body weight, about 200 microgram/kg body weight, about 350 microgram/kg body weight, about 500 microgram/kg body weight, about 1 milligram/kg body weight, about 5 milligram/kg body weight, about 10 milligram/kg body weight, about 50 milligram/kg body weight, about 100 milligram/kg body weight, about 200 milligram/kg body weight, about 350 milligram/kg body weight, about 500 milligram/kg body weight, to about 1000 mg/kg body weight or more per administration, and any range derivable therein. In non-limiting examples of a derivable range from the numbers listed herein, a range of about 5 mg/kg body weight to about 100 mg/kg body weight, about 5 microgram/kg body weight to about 500 milligram/kg body weight, etc., can be administered, based on the numbers described above. Thus, one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 5.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient.
[199] Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the peptide molecule). An initial higher loading dose, followed by one or more lower doses may be administered. In some embodiments, the dosages may be administered daily, every other day, continuously over one day or more. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
[200] In certain embodiments, the dosage may comprise from about 0.00001 to 500 mg/kg/day, or from about 0.0001 to 300 mg/kg/day, or from about 0.003 to 100 mg/kg/day, or from about 0.03 to 30 mg/kg/day, or from about 0.1 mg/kg/day to 10 mg/kg/day, or from about 0.3 mg/kg/day to 3 mg/kg/day. [201] In another embodiment, the dosage is 20 mg per peptide per day. In another embodiment, the dosage is 10 mg/peptide/day. In another embodiment, the dosage is 30 mg/peptide/day. In another embodiment, the dosage is 40 mg/peptide/day. In another embodiment, the dosage is 60 mg/peptide/day. In another embodiment, the dosage is 80 mg/peptide/day. In another embodiment, the dosage is 100 mg/peptide/day. In another embodiment, the dosage is 150 mg/peptide/day. In another embodiment, the dosage is 200 mg/peptide/day. In another embodiment, the dosage is 300 mg/peptide/day. In another embodiment, the dosage is 400 mg/peptide/day. In another embodiment, the dosage is 600 mg/peptide/day. In another embodiment, the dosage is 800 mg/peptide/day. In another embodiment, the dosage is 1000 mg/peptide/day.
[202] Other dosage ranges are contemplated by this invention. In one embodiment, the dosage is 20 pg per peptide per day. In another embodiment, the dosage is 10 pg/peptide/day. In another embodiment, the dosage is 30 pg/peptide/day. In another embodiment, the dosage is 40 pg/peptide/day. In another embodiment, the dosage is 60 pg/peptide/day. In another embodiment, the dosage is 80 pg/peptide/day. In another embodiment, the dosage is 100 pg/peptide/day. In another embodiment, the dosage is 150 pg/peptide/day. In another embodiment, the dosage is 200 pg/peptide/day. In another embodiment, the dosage is 300 pg/peptide/day. In another embodiment, the dosage is 400 pg/peptide/day. In another embodiment, the dosage is 600 pg/peptide/day. In another embodiment, the dosage is 800 pg/peptide/day. In another embodiment, the dosage is 1000 pg/peptide/day. In another embodiment, the dosage is 1500 pg/peptide/day. In another embodiment, the dosage is 2000 pg/peptide/day.
[203] In another embodiment, the dosage is 10 pg/peptide/dose. In another embodiment, the dosage is 30 pg/peptide/dose. In another embodiment, the dosage is 40 pg/peptide/dose. In another embodiment, the dosage is 60 pg/peptide/dose. In another embodiment, the dosage is 80 pg/peptide/dose. In another embodiment, the dosage is 100 pg/peptide/dose. In another embodiment, the dosage is 150 pg/peptide/dose. In another embodiment, the dosage is 200 pg/peptide/dose. In another embodiment, the dosage is 300 pg/peptide/dose. In another embodiment, the dosage is 400 pg/peptide/dose. In another embodiment, the dosage is 600 pg/peptide/dose. In another embodiment, the dosage is 800 pg/peptide/dose. In another embodiment, the dosage is 1000 pg/peptide/dose. In another embodiment, the dosage is 1500 pg/peptide/dose. In another embodiment, the dosage is 2000 pg/peptide/dose.
In another embodiment, the dosage is 10-20 pg/peptide/dose. In another embodiment, the dosage is 20-30 pg/peptide/dose. In another embodiment, the dosage is 20-40 pg/peptide/dose. In another embodiment, the dosage is 30-60 pg/peptide/dose. In another embodiment, the dosage is 40-80 pg/peptide/dose. In another embodiment, the dosage is 50-100 pg/peptide/dose. In another embodiment, the dosage is 50-150 pg/peptide/dose. In another embodiment, the dosage is 100-200 pg/peptide/dose. In another embodiment, the dosage is 200-300 pg/peptide/dose. In another embodiment, the dosage is 300-400 pg/peptide/dose. In another embodiment, the dosage is 400-600 pg/peptide/dose. In another embodiment, the dosage is 500- 800 pg/peptide/dose. In another embodiment, the dosage is 800-1000 pg/peptide/dose. In another embodiment, the dosage is 1000-1500 pg/peptide/dose. In another embodiment, the dosage is 1500-2000 pg/peptide/dose.
[204] In another embodiment, the dosage is 10 mg/peptide/dose. In another embodiment, the dosage is 30 mg/peptide/dose. In another embodiment, the dosage is 40 mg/peptide/dose. In another embodiment, the dosage is 60 mg/peptide/dose. In another embodiment, the dosage is 80 mg/peptide/dose. In another embodiment, the dosage is 100 mg/peptide/dose. In another embodiment, the dosage is 150 mg/peptide/dose. In another embodiment, the dosage is 200 mg/peptide/dose. In another embodiment, the dosage is 300 mg/peptide/dose. In another embodiment, the dosage is 400 mg/peptide/dose. In another embodiment, the dosage is 600 mg/peptide/dose. In another embodiment, the dosage is 800 mg/peptide/dose. In another embodiment, the dosage is 1000 mg/peptide/dose.
[205] In another embodiment, the dosage is 10-20 mg/peptide/dose. In another embodiment, the dosage is 20-30 mg/peptide/dose. In another embodiment, the dosage is 20-40 mg/peptide/dose. In another embodiment, the dosage is 30-60 mg/peptide/dose. In another embodiment, the dosage is 40-80 mg/peptide/dose. In another embodiment, the dosage is 50- 100 mg/peptide/dose. In another embodiment, the dosage is 50-150 mg/peptide/dose. In another embodiment, the dosage is 100-200 mg/peptide/dose. In another embodiment, the dosage is 200- 300 mg/peptide/dose. In another embodiment, the dosage is 300-400 mg/peptide/dose. In another embodiment, the dosage is 400-600 mg/peptide/dose. In another embodiment, the dosage is 500-800 mg/peptide/dose. In another embodiment, the dosage is 800-1000 mg/peptide/dose.
[206] Initial dosages can also be estimated from in vivo data, e.g., animal models, using techniques that are well known in the art. One having ordinary skill in the art could readily optimize administration to humans based on animal data.
[207] The dosage amount and interval may be adjusted individually to provide plasma levels of the peptides that are sufficient to maintain therapeutic effect. Usual patient dosages for administration by injection range from about 0.1 to 50 mg/kg/day, typically from about 0.5 to 1 mg/kg/day. Therapeutically effective plasma levels may be achieved by administering multiple doses each day. Levels in plasma may be measured, for example, by HPLC.
[208] In one embodiment, the plasma concentration of the peptides is about 0.05 to 0.9 ng/ml after about 15 minutes after application, or about 0.1 to 1.4 ng/ml after about 30 minutes, or about 0.1 to 1.6 ng/ml after about 1 hour, or about 0.1 to 1.4 ng/ml after about 1.5 hours, or about 0.1 to 1.3 ng/ml after about 2 hours, or less than about 0.7 ng/ml after 5 hours, or less than about 0.2 ng/ml after 10 hours.
[209] In one embodiment, the plasma concentration of the peptides is about 1-20 mM after about 15 minutes after application, or about 1-20 mM after about 30 minutes, or about 1-20 pM after about 1 hour, or about 1-20 pM after about 1.5 hours, or about 1-20 pM after about 2 hours, or less than about 1-20 pM after 5 hours, or less than about 1-20 pM after 10 hours.
[210] Depending on the means of administration, the dose may be administered all at once, such as with an oral formulation in a capsule, or slowly over a period of time, such as with an intravenous administration. For slower means of administration, the administering period can be a matter of minutes, such as about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120 or more minutes, or a period of hours, such as about 0.5, 1 , 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5 or more hours. The administration of the dose may be interrupted, such as where the dose is administered via intravenous infusion and the dose is divided into two or more infusion bags. Under such circumstances, the administration of the dose may be interrupted while the infusion bags are changed.
[21 1] In one embodiment, because they bind targets in tumors (e.g., a1 b1 and anb3 integrins) the peptides of the invention can also be used for tumor visualization by single-photon emission computed tomography (SPECT), which is based on g-rays, and the peptides of the invention are here linked to radionuclides such as 99mTc, 177Lu, 1231 and 1 1 11n. On the other hand, the positron-emitting radioisotopes 68Ga, 1241 or 89Zr are used for positron emission tomography (PET) purposes. Prior to radiolabeling, the peptides of the invention can be conjugated with bifunctional chelating agents which possess a metal binding moiety for sequestration of the metallic radionuclide and are generally DPTA (acyclic)-, DOTA (macrocyclic), or NOTA-based. Moreover, the chelating agents are equipped with a chemically reactive functional group for attachment to the peptides of the invention, which can occur in several ways well known to one of ordinary skill in the art. To avoid ionizing radiation, the peptides of the invention can also be conjugated to near-infrared fluorophores such as IRDye800CW to perform optical tumor imaging, a technique that is cost-effective, flexible, sensitive and fast. Additional suitable labels include an affinity label (e.g., biotin, avidin), a spin label, an enzyme, a fluorescent group, or a chemiluminescent group. When labels are not employed, complex formation (e.g., between the peptide and a target) can be determined by surface plasmon resonance, ELISA, FACS, or other suitable methods.
[212] In another embodiment of the invention, an article of manufacture containing materials that comprise the peptides of the invention and are useful for the treatment, prevention and/or diagnosis of the disorders described above is provided. The article of manufacture comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is a peptide of the invention. The label or package insert indicates that the composition is used for treating the condition of choice. Moreover, the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises peptide of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent. The article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition. Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
[213] EXAMPLES OF OTHER USES FOR THE DISCLOSED INVENTION
[214] The molecular docking model between peptide RK and integrin described in this disclosure is also an embodiment of the invention. In one embodiment, the molecular docking model between peptide RK and integrins is used to design or select/screen for organic drugs or other peptides that can be used in any of the other methods described herein (e.g., methods of treatment). EXAMPLES
The RK 14mer
[215] Scorpion venoms is a rich soup of peptide compounds, known for their biological properties. Many of them were found to act on different cancer hallmarks [21] Few of the scorpion peptides are able to act on cell proliferation, migration and angiogenesis. Margatoxin, Iberiotoxin and Charybdotoxin were reported to inhibit cell proliferation while TM601 , a synthetic analogue of Chlorotoxin is capable to inhibit angiogenesis [22] These scorpion peptides have the common feature of presenting multiple disulfide bridges (at least 3) and a number of composing residues up to 30 amino acids [23] In our work however, the inventors describe a new short peptide, RK1 from Buthus occitanus tunetanus with 1467.67 KDa consisting of 14 amino acids. Apparently, the peptide does not belong to any of the characterized families. It is also particular in containig one disulfide bridge formed between the C3 and the C12 amino acid.
[216] Toxins and peptides derived from scorpion venoms generally have direct effects on the ion channels (K+, Na+, Ca2+...) [24] The few peptides that have an anti-tumor effect are generally cytotoxic. For example, two peptides named neopladine 1 and neopladine 2, isolated from Tityus discrepans scorpion venom, were reported to be effective in inducing apoptosis and necrosis of SKBR3 breast cancer cells [25] Before testing the biological effect of RK1 , the inventors assessed its toxicity on Tumor cells U87 and IGR39. After 48 h treatment, RK1 has no cytotoxic effect up to a concentration of 8 mM. Therefore for the further tests, the inventors operated under cytotoxic concentrations to study the specific effect of RK1.
[217] Cell proliferation is a key step in tumor development. Indeed, the anarchic proliferation of cells makes them more solid and resistant tumours [26] For this, the inventors have evaluated the anti-proliferative potential of RK1 which is demonstrated on both cells lines U87 and IGR39. The effect is mostly sustained at constant levels for the assay period of four days. This might, in fact, show the stability of RK1 in the in vitro conditions. Moreover, since the the anti-proliferative effect is similar at both concentrations of 2 and 4 pM, it is suggested that the activity is denoted under saturation levels.
[218] Our results show that RK1 completely inhibit the proliferation of U87 cells from the first day at 2 pM. It also inhibits more than 90% of the proliferation of IGR39 melanoma cells from at 2 pM.
[219] Until today, there has been no description of a scorpion venom peptide wich completely inhibits cell proliferation. Only two venom extracts, Bmk from Chinese venom of Buthus matensii Karsch which inhibits the proliferation of MCF-7 breast cancer cells and SMMC7721 of hepatoma cells with rates that do not exceed 40%. We found also the extract of Odontobuthus doriae venom that inhibits DNA synthesis while inducing apoptosis 21 Two 36 amino acids recombinant peptides derived from chlortoxines termed CA4 and CTX-23 inhibit 40% of glioma cell proliferation in dose dependent manner at 6 mM dose [28] Our results showed that RK1 is more efficient while it completely inhibit U87 cell proliferation at 2 pM concentration.
[220] Cell migration is a physiological process essential to life, in oncology it intervenes in the formation of new metastases [29] Our results show that RK1 inhibits migration of melanoma cells IGR39 by more than 40 % at 2 pM and 80 % at 4 pM dose. Moreover, it inhibits also glioma cell migration by 22 % and 40 % at 2pM and 4 pM concentrations, respectively. In comparison to RK1 , CA4 and CTX-23 inhibited glioma cell migration by 60% at 2 pM treatment while Chlorotoxin has been reported to significantly reduce glioma cell migration dose-dependently at 5 pM concentration [30] A recombinant fusion protein SUMO-AGAP Peptide from the Venom of Mesobuthus martensii Karsch which combined a small ubiquitin related modifier to AGAP was proven to have antitumor activity [31] Further study showed that SUMO-AGAP inhibited cell proliferation and migration of SHG-44 human malignant glioma cells up to 50 % at 30 pM treatment concentration [32] From a certain size (2-3 mm), tumor causes the establishment of new vessels to feed its growth and ensure its survival, this fundamental step corresponds to the "angiogenic switch" described by Folkman [33] Using CAM model, RK1 was shown to inhibit ex- vivo vascular growth by 50 % at 4 pM dose and by 60 % at 8 pM. Similar results were observed with Chlorotoxin, CA4 and CTX-23. Theses peptides inhibited (at 10 pM) 41 %, 44% and 46% respectively after Chlorotoxin, CA4 and CTX-23 application [28] Hemilipin2, the small sub unit from secreted heterodimeric phospholipase A2 (sPLA2) from Hemiscorpius lepturus scorpion venom showed an important anti-angiogenic activity since it inhibits more than 37 % of ex vivo angiogenesis at 1 pM [34]
[221] On all of our results, the inventors show that RK1 has an anti-tumor effect by inhibiting the proliferation and migration of gliomatous and melanomatous cells on the other hand, RK1 strongly inhibits neoangiogenesis. Our peptide has no toxcity, in vitro and in vivo, which can decrease the harmful toxic effects of chemotherapy and radiotherapy. Small size of RK1 hence the possibility of generating less immunogenicity and its capacity to form a disulfide bond which might increase its stability in physiological conditions. RK1 is a very short peptide (14 aa) that has significant anti-tumor activity and can be classified as one of the few anti-cancer scorpion venom peptides that do not have cytotoxic activity.
[222] In our perspectives, the inventors plan to better investigate this anti-tumor effect and study the involved signaling pathways in order to better understand its mechanism of action and to define its molecular targets by which it exert the described effects. RK1 PEPTIDE-EXAMPLE 1
Materials and Methods
[223] Animals
[224] The Buthus occitanus tunetanus scorpions from Beni Khedach (Tunisia) were electrically stimulated on their post-abdomen by the veterinarian service of the Pasteur Institute of T unisia to release venom in liquid state. The pooled venom is kept frozen at -20 ° C in its crude form (lyophilized) until its use. All reagents were purchased from Sigma-Aldrich ® Chemical company, except indicated otherwise.
[225] Cell culture
[226] U87 (Glioblastoma) and the human cell lines derived from melanoma (IGR39) were graciously provided by Jose Luis (Laboratory of Cell Biology, Faculty of Pharmacy, Marseille). Dulbecco's modified Eagle's medium (DMEM), fetal calf serum (FCS), and modified Eagle's medium (MEM) were purchased from Lonza (Walkersville, MD). Cancer cell line U87 was first cultured in MEM medium supplemented with 10% FCS containing 100 lU/ml penicillin. IGR39 (Melanoma) were routinely cultured in DMEM containing 10% FCS. All cell lines were maintained at 37°C in 5% C02.
[227] Purification of RK1
[228] The purification of the soluble components was initially performed by gel filtration on Sephadex G-50 column chromatography (grade superfine, Pharmacia Fine Chemicals, Uppsala, Sweden). The lyophilized venom was dissolved in 1 M ammonium acetate buffer (pH 8.5), and applied directly to the column. Different fractions were eluted and tested for their toxicity on mice as described below. Only fraction (BotG50) showing a toxic activity was then applied onto semi preparative reversed-phase HPLC C8 column (10 mm x 250mm, 5 pm, Beckman Fullerton) equilibrated in 0.1 % trifluoroacetic acid in water, at a flow rate of 1 ml/min [15] HPLC purification of the non-retained fraction was performed using an analytical C18 reversed-phase HPLC column (4.6mm x 250 mm, 5 microns Beckman). Detection was monitored at 214 nm.
[229] Mass spectrometry
[230] The peptide was analyzed by MALDI-TOF mass spectrometer (Perspective Biosystems, Inc., Framingham, MA). The sample was dissolved in CH3CN/H20 (30/70) with 0.3% trifluoroacetic acid to obtain a concentration of 1-10 pmol. pL-1.The matrix was prepared as follows: alpha-cyanohydroxycinnamic acid was dissolved in 50% CH3CN in 0.3% trifluoroacetic acid/H20 to obtain a saturated solution. Peptide solution was placed on the sample plate, and 0.5 pL of the matrix solution was added. This mixture was allowed to dry. Mass spectra were recorded in linear mode, calibrated with suitable standards and were analyzed by GRAMS/386 software.
[231] Amino acid sequence determination and peptide synthesis
[232] Reduction and alkylation of peptides and sequence determination of native and S-alkylated peptide were performed as described in [16] The RK1 peptide was synthesized by Genosphere Biotechnologies (Paris, France). Automated synthesizer by the Fmoc method on solid phase (Merrifield, R. B. 1969. Solid-phase peptide synthesis. Adv. Enzymol. Relat. Areas Mol. Biol.32:221.6) Analytical RP-HPLC and MS confirmed the purity (> 95%) and molecular mass of the synthesized peptide. The peptide stock was diluted in distilled water and kept at -20°C, and quantified by a Nano DropND-1000 V3.5.2 Spectrophotometer at 280 nm. The reduced peptide was solubilized in 0.2 M Tris/HCI, pH 8, at 5 mM and stirred under air to allow folding (48 h, 25 °C). 2.6. Cell viability test and in vivo toxicity test
[233] Cell viability and in vivo toxicity
[234] Cell viability was assessed by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide) assay[17] MTT solution was added to the culture medium 4 h before the end of treatment. Cells were incubated with RK1 for 72 h, then fixed with 1 % glutaraldehyde, stained with a solution of 0.1 % crystal violet, and lysed with 1 % SDS. Absorbance was then measured at 560 nm. A control was used in the same conditions but without RK1. The in vivo toxicity of RK1 was tested on 20±2 g C57/BL6 male mice by intracerebro-ventricular (I.C.V) injection of 0.1 % (w/v) BSA solutions containing increasing amounts of the peptides in a volume of 5 pl_. Six mice were used for each dose; two control mice were injected with only 0.1 % BSA in water. All procedures met with the approval of the Institutional Research Board of the Pasteur Institute of Tunis. I.C.V administration was performed under ether anesthesia [18]
[235] Cell proliferation assay
[236] U87 and IGR39 cells were seeded in a microtiter plate (5000 cells/well) and incubated overnight at 37°C in a humid atmosphere of 5% C02 in air. The medium is then updated in the presence of different concentration of RK1 (2 and 4 mM). After 4 days of incubation, the wells are washed with PBS and the cells were fixed with 1 % glutaraldehyde every day, stained with 0.1 % crystal violet and quantified by absorbency at 560 nm.
[237] Cell migration
[238] Cell migration was measured using wound-healing assay as previously described [19] In a sterile environment use a 20 mI_ pipette to to press firmly against the top of the tissue culture plate and swiftly make a scratch on the cell surface when the cells were cultured to full confluence. Flowingly, the cells were washed with PBS to remove debris and incubated in serum- free DMEM (IGR39) or MEM (U87) for 24 h. The cell migration index was measured at indicated time points (0 and 24 h) using Image J software (NIH, Bethesda, MD, USA).
[239] Chicken chorioallantoic membrane assay CAM
[240] Chick embryos from 3-day-old eggs were opened and placed in double Petri- dishes with water for humidity. After five days at 37°C, filter paper disks (diameter 6 mm) soaked in buffer (0.9% NaCI) as control, RK1 (2, 4 and 8 mM) were applied on the Chicken chorioallantoic membrane (CAM). After 48 h, spontaneous angiogenesis was observed and photographed with a digital camera at 10* magnification. Data analysis
[241] All values are expressed as mean ± standard deviation (SD). The statistical significance of differential findings between experimental and control groups was determined by Student's t-test using GraphPad Prism 4 software. P < 0.05 was considered statistically significant and is indicated with asterisks over the value (*:p < 0.05; **:p < 0.01 ; ***p < 0.001).
RK1 PEPTIDE-EXAMPLE 2
Purification of the RK1 Peptide
[242] The crude venom of Buthus occitanus tunetanus was separated by gel filtration on a Sephadex G- 50 column chromatography as previously described [20] The toxic fraction (BotG50 ) obtained from this separation was purified by high performance liquid chromatography (HPLC) using a semi preparative C8 column. The fraction eluting at 19-22 min (Fig. 1A) was further purified (using an analytical C18 reversed-phase HPLC column, the component eluted at 37.5 min (Fig. 1 B) was homogeneous, as indicated by mass spectrometry analysis and amino acid sequencing. It is designated RK1. An analytical HPLC run of RK1 showed a single symmetric peak (Fig. 1 C). Elution was performed using a linear gradient and monitored by measuring the absorbance (214 nm) with a Beckman 166 detector.
RK1 PEPTIDE-EXAMPLE 3
Sequence determination of RK1 and peptide synthesis
[243] The automatic Edman degradation of 25pmol of native RK1 was performed with a reproducible yield of 95% during 14 cycles. After these steps, no PTH (Phenylthiohydantoin) amino acid was detected. 50 pmol of S-alkylated-protein was used to confirm without ambiguity the determined sequence, and to identify cystein positions. RK1 is a short peptide composed of 14 amino acid residues containing two cystein residues (Fig. 1 E).
[244] The experimental molecular mass of native RK1 (1467.67 Da) obtained by MALDI-Tof mass spectrometry (Fig. 1 D), is nearly identical with the average theoretical molecular mass calculated for the peptide with Oxidize cysteines form (1467.03 Da). Due to its very low concentration in venom (0.01 % of the proteins), RK1 was chemically synthesized. The BLAST and PSI-BLAST search did not return any significant homology hit. This is thus a new short peptide, RK1 from Buthus occitanus tunetanus with 1467.67 Da consisting of 14 amino acids. Apparently, the peptide does not belong to any of the characterized families. It is also particular in containing one disulfide bridge formed between the C3 and the C12 amino acid.
RK1 PEPTIDE-EXAMPLE 4
Biological activity
[245] Before testing the biological effect of RK1 , the inventors assessed the peptide’s toxicity on Tumor cells U87 and IGR39. After 48 h treatment, RK1 has no cytotoxic effect up to a concentration of 8 mM. Therefore, for the further tests, the inventors operated under cytotoxic concentrations to study the specific effect of RK1 Evaluation of the toxicity of RK1 and RK1 s revealed that both peptides do not exhibit any neurotoxicity up to a 2pg/20g mice or 100 pg/kg body weight as determined by intracerebroventricular injection. RK1s was tested on the viability of glioblastoma (U87) and melanoma (IGR-39) cell lines using the MTT assay. At 100 pM concentration, RK1 s does not affect the viability of the two cell lines after 24h, 48h or 72h incubation time (Fig. 2A).
RK1 PEPTIDE-EXAMPLE 5
Effect of RK1 on tumour cell proliferation and migration
[246] Here, the inventors investigated the anti-proliferative effect of RK1 on both U87 and IGR39 cells. Obtained results showed that our peptide is able to completely reduce IGR39 cell proliferation at 2 pM dose and more than 90% for U87 cell proliferation after 4 days’ incubation (Fig. 2B).
[247] Our results show that RK1 completely inhibit the proliferation of U87 cells from the first day at 2 pM. It also inhibits more than 90% of the proliferation of IGR39 melanoma cells at 2 pM. The effect is mostly sustained at constant levels for the assay period of four days. This might, in fact, show the stability of RK1 in the in vitro conditions. Moreover, since the the anti proliferative effect is similar at both concentrations of 2 and 4 pM, it is suggested that the activity is denoted under saturation levels.
[248] These results are unexpected. Until today, there has been no description of a scorpion venom peptide which completely inhibits cell proliferation.
[249] The ability of RK1 to inhibit migration of U87 and IGR39 cells was tested using wound-healing assay. First, the peptide was tested at 2 and 4 pM on both used cell lines. Results obtained show that RK1 inhibit differently the migration of these cells. RK1 has a large effect since it inhibits more than 40 % of U87 cell migration (Fig. 3B) and 80 % of IGR39 migration at 4 pM concentration (Fig. 3A). [250] Our results show that RK1 inhibits migration of melanoma cells IGR39 by more than 40 % at 2 mM and 80 % at 4 mM dose. Moreover, it inhibits also glioma cell migration by 22 % and 40 % at 2mM and 4 mM concentrations, respectively
RK1 PEPTIDE-EXAMPLE 5
Chicken chorioallantoic membrane assay (CAM)
[251] To further characterize pharmacological properties of RK1 , the inventors performed in vivo angiogenesis using chick chorioallantoic membrane (CAM) assays. The spontaneous angiogenesis in CAM was observed after 48 h. As illustrated in Fig. 4A, RK1 induced a marked reduction in the number of new capillaries and branching vessels in the CAM, without affecting pre-existing vessels. Furthermore, a decrease in the vascular density could be observed. Quantification shows that the total vessel length was reduced by 25%, 48% and 77% for 2 mM, 4 mM and 8mM concentration, respectively, compared with the untreated conditions (Fig. 4B).
[252] Using CAM model, RK1 was shown to inhibit ex-vivo vascular growth by 50 % at 4 mM dose and by 60 % at 8 mM.
[253] The results show that RK1 has an anti-tumor effect by inhibiting the proliferation and migration of gliomatous and melanomatous cells. On the other hand, RK1 strongly inhibits neoangiogenesis, which means that the peptide is capable of acting on endothelial cells. Our peptide has no toxicity, in vitro and in vivo, which can decrease the harmful toxic effects of chemotherapy and radiotherapy. Small size of RK1 hence the possibility of generating less immunogenicity and its capacity to form a disulfide bond which might increase its stability in physiological conditions. RK1 is a very short peptide (14 aa) that has significant anti-tumor activity and can be classified as one of the few anti-cancer scorpion venom peptides that do not have cytotoxic activity.
[254] In summary, RK1 is the first very short peptide from Buthus occitanus tunetanus that, unexpectedly, inhibits three key aspects of cancer development, namely tumor cell migration, proliferation, and angiogenesis.
The RK 17mer
[255] A novel peptide, named RK was also isolated and characterized from the Buthus occitanus tunetanus scorpion venom targeting the cell adhesion activity by acting on integrins. To our knowledge, this is the first disintegrin-like peptide purified from a scorpion venom. Interestingly, RK peptide is unique among the variety of known peptides purified from scorpion venoms in terms of its structure. It is a small peptide of 17 amino acids (IDCGTVMIPSECDPKSS; SEQ ID NO:1) containing a single disulfide bond. RK may represent the first member of a new group of scorpion peptides. [256] There are only 23 scorpion peptides published so far whose primary structure comprises lesser than 20 residues with different pharmacological activities [30] They all possess variable pharmacological properties. Their sequence covers a range of length anywhere between 4 and 17 amino acids. Among them, only PIMT and Pit toxins share the same sequence length than RK [31] It was therefore not trivial to assign RK to any of the pre-existing families because the peptide has no obvious structural or sequence homology with these other peptides.
[257] Interestingly RK did not show any toxicity a dose of 100 pg per kg mice body weight. It is also non-cytotoxic until 100 pM concentration. RK causes the detachment of three cell lines on different ECM proteins. The ineffectiveness of the RK on the non-specific substratum poly-L-lysine coated plates suggests that the inhibition effect of tumoral cells adhesion, by RK peptide, is integrin dependent. High levels of anb3, a5b1 and a3b1 are expressed in U87 [32], while IGR-39 highly expresses a2b1 and anb5 integrins and modestly expresses anb3 [33]
[258] The disintegrin activity of RK on U87 and IGR-39 would be linked to an effect on anb3 integrin. This could explain why a more important effect is exhibited by the peptide on U87 compared to IGR39. The lack of a significant activity on Fibronectin coated plates on IGR39 seems to be unlikely at first glance as the inventors expected for the inhibition effect of RK to be similar on both Fibrinogen and Fibronectin like the inventors did observe in U87. However, these results might be explained by the fact that IGR39 expresses high levels of anb5 which can bind to Fibronectin though it is more specific to vitronectin [34] and for such reason, the cell can still adhere to the extracellular matrix even in the presence of RK. A snake venom anti-adhesion effect of IGR39 was previously reported for lebectin, a C-type lectin. In contrast to RK, lebectin is only active on fibronectin and fibrinogen matrix types but not on collagen I and IV [35]
[259] RK peptide disintegrin activity on U87 was evaluated at IC50 values of 10 mM and 10.33 pM receptively for Fibrinogen and Fibronectin. The assay on PC12 cell line shows an anti adhesion activity on collagen IV coated plates, while no significant effect is noted for the rest of the ECM proteins. PC12 expresses a1 b1 and a3b1 integrins, both are able to bind laminin in vitro, but only a1 b1 can interact with type I and type IV collagen [36] Therefore, in addition to anb3 the inventors suggest in our study that RK acts on a1 b1 integrin.
[260] As a comparison, the anti-adhesion to fibrinogen and fibronectin of PIVL (7691 Da), a snake venom disintegrin targeting anb3, on U87-cells was evaluated to similar IC50 of about 250 nM and 300 nM respectively [37] The similar IC50 values might suggest also that RK inhibits the activity of a common target receptor for both Fibrinogen and Fibronectin which the inventors suggest to be the anb3 integrin. This is supported by the previous calculation of the association rate constants of fibrinogen and fibronectin to anb3 integrin which are also very similar, evaluated at 2.49 + 0.8 x 104 M-1 s-1 and 2.63 +- 0.05 x 104 M-1 s-1 , respectively [38] The antibody assay, confirms that both integrins are potential targets for the scorpion peptide.
[261] To our knowledge, RK is the first biomolecule demonstrating the dual activity on a1 b1 , which binds to collagen IV, and anb3 which binds to all ECM macromolecules presenting the RGD motif (vitronectin, fibronectin, fibrinogen, osteopentin, and bone sialoprotein) [39] We though, that such property might be caused by the presence of one or two functional motifs described for the snake venom disintegrin. RK sequence contains two potential disintegrin motifs: ECD and KSS. Indeed, the ECD motif was previously described in Acurhagin-C to be responsible for the inhibition of the anb3 mediated endothelial cell adhesion to extracellular matrix [40]
Putative
Sequence
Disintegrin adhesion Ligand Cell lines IC50 Ref
Length Integrin
motif
collagen
Obtustatin 41 KTS aΐbΐ a1K562 2 nM [49]
IV
collagen 0.08
Viperistatin 41 KTS aΐbΐ lK562 [50]
IV nM
PC12 Rat
collagen
Lebestatin 41 KTS ΐbΐ pheochromocyto 0.2 nM [43]
IV
ma
Ii, D2 PC12 Rat
collagen 4.84
RK 17 M7 and aΐbΐ pheochromocyto
IV mM
Kl5 ma
collagen 0.15
IV Mouse 0.34
Acurhagin-C 421 ECD nb3 [40] gelatin B melanoma cells 0.65 and Fn (B16-F10) mM
nb3,
Melanoma 100
Leberagin-C 205 ECD a5b1, Vn, Fn [42] tumour cells nM
Figure imgf000062_0001
Mouse
collagen
Altemagin-C 196 ECD a2b1 fibroblast 2.2mM [41]
I
(NIH-3T3)
f8 U87 10 mM
RK 17 ECD anb3 Fn Glioblastoma 10.33
mM
[262] Table 1. Effect of different disintegrins with either KTS or ECD motif on cell adhesion activity compared to RK peptide.
[263] Legend: Fg, fibrinogen; Vn, vitronectin; Fn, fibronectin; a1 K562: K562 cells transfected with specific ai subunits of integrins. Ref, references.
[264] Furthermore, the peptidomimetic of 12 amino acids, issued from the parent ECD loop carrier in Acurhagin-C purified from Formosan Agkistrodon acutus venom, showed similar disintegrin activity to RGD motif containing peptide [41] ECD motif was also described in Alternagin-C, and Leberagin-C [42] On the other hand, the KSS segment in RK could be is similar to KTS motif which is thought to be responsible for the a1 b1 disintegrin activities for Obtustatin, Viperistatin and Lebestatin [43] The importance of KTS motif was also corroborated in short linear peptide constructions [44] To investigate the expected interaction of RK with a1 b1 and anb3 integrins, the inventors conducted computational studies. The molecular dynamics simulation of RK highlights the conformational flexibility of the peptide which allows it to adopt multiple stable states.
[265] The free energy landscape shows a dense populated global minimum with a large bottom. Such characteristic allows a great adaptability toward the binding site of a1 b1 integrin (Fig. 11). The latter has the property to undergo a structural rearrangement upon the binding of collagen IV notably for the C loop as concluded by the crystal and NMR solution of the integrin [45-46-47-48] The binding to a1 b1 could then be ensured favorably enough even if the peptide structure shifts significantly from the global minimum conformation which corresponds to a low drifts in the energy of the system. This property also, imply that the RK peptide must preserve a minimum specific set of contacts with high structural stability, in order to interact with the integrin.
[266] The RMSF profile suggests that the central segment of the peptide, limited by the positions of the two cystein residues, are stable during the molecular dynamics simulation. Particularly, the segment V6, M7 and I8 of the peptide might be critical to ensure the minimal contacts required for the interaction with a1 b1 regarding also the results from the docking (Fig. 11). The local conformation of the segment corresponding to a turn-like structure that remains stable during the 300 ns of molecular dynamics simulation. It allows to the M7 side chain to preserve a high level of solvent exposure but also to minimize the structural constraints with other atoms of the peptide.
[267] These properties are essential for the interaction with the small hydrophobic pocket on the surface of the integrin. The low flexibility in the 12-15 segment is also essential to stabilize the structure of K15 residue. Such stability might allow the formation of the salt bridge with E285 amino acid of Dial . The position of the S16 and S17 residues at the C-terminal end of the sequence causes a high instability. In addition, based on the protein-peptide docking study, none of the final retained solutions shows any critical implication of the KSS segment in the interaction with the integrin. Therefore, the inventors suggest that the interaction of RK peptide with a1 b1 integrin does not implicate the KSS segment.
[268] Our results are in accordance with those obtained with the snake venom disintegrin when mutagenesis study demonstrates that the substitution of KTS with AAA sequence in obtustatin, do not allow the total loose of its activity, compared to the wild type obtustatin [49], although this motif has been earlier proposed as the functional amino acid triplet for its activity [50] These findings, suggest that the interaction mechanism of the snake venom disintegrin with a1 b1 might not rely only on the KTS triplet and that other critical residues might be involved in the formation of the complex. In the absence, of any interaction model of a1 b1 snake venom disintegrin, the mechanism is still to be investigated.
[269] The incubation of U87 with RGD peptide coated plates in the presence and the absence of RK suggests that both peptides share the same binding site on anb3. Consequently, the inventors investigated the involvement of the ECD segment of RK in the interaction with the integrin. The rational behind this choice is the similarity with the same motif described in other snake disintegrin inhibiting the same receptor [51-39-42] Our results showed that RK was able to sample a set of structures among the molecular dynamics ensemble for which the ECD segment has very similar geometric property compared to RGD in the co-crystal structure [52-53] (Fig. 12A). In addition, the conformations with the lowest clash score correspond also to ideal geometries of interactions with the anb3 integrin (Fig. 12B). This in fact suggests that RK is able to engage the ECD segment in the interaction with RGD binding site without been hindered by the other residues of the same peptide. Therefore the inventors suggest that, ECD might be the functional segment which controls the interaction with anb3 integrin (Fig. 12C). However, the conformations with very similar RGD geometry for the defined reactions coordinates in our computational analysis are not densely represented among the total ensemble sampled in the molecular dynamics simulation. This in fact, can explain the low inhibition activity of RK toward anb3 compared to a1 b1.
[270] In conclusion, this study was the first to describe scorpion peptide with dual disintegrin activity on a1 b1 and anb3 integrins. This double effect of RK is due to the presence of contiguous motifs ECD and KSS. It is to highlight that selective blockade of a1 b1 and anb3 integrins is a desirable goal for the therapy of a number of pathological conditions including essentially cancer and tumor angiogenesis.
RK PEPTIDE 17 mer-EXAMPLE 6
Purification of the peptide
[271] RK was substantially purified from the Buthus occitanus tunetanus scorpion venom by a first stage of gel filtration on a Sephadex G-50 column chromatography as previously described [28] The toxic fraction (BotG50) obtained from this separation was purified by high performance liquid chromatography (HPLC) using a semi preparative C8 column. The fraction eluting at 19-22 min (Fig. 5A), was further purified (using an analytical C18 reversed-phase HPLC column. The component eluted at 33.5 min (Fig. 5B) was homogeneous, as indicated by mass spectrometry analysis and amino acid sequencing. It is designated RK. An analytical HPLC run of RK showed a single symmetric peak (Fig. 5C).
RK PEPTIDE 17 mer-EXAMPLE 8
Sequence determination of RK and peptide synthesis (RKs)
[272] The automatic Edman degradation of 25 pmol of native RK was performed with a reproducible yield of 95% during 17 cycles. After these steps, no PTH (Phenylthiohydantoin) amino acid was detected. 50 pmol of S-alkylated-protein was used to confirm without ambiguity the determined sequence, and to identify cystein positions. RK is a short peptide composed of 17 amino acid residues containing two cystein residues (Fig. 5D).
[273] The experimental molecular mass of native RK (1780.03 Da) obtained by MALDI- Tof mas spectrometry (supplementary data 1), is nearly identical with the average theoretical molecular mass calculated for the peptide with Oxidize cysteines form (1780.021 Da).
[274] This shows that the two cysteine residues at position 3 and 12 are involved in intramolecular disulfide bonds. Due to its very low concentration in venom (0.01 % of the proteins), RK was chemically synthesized. After renaturation, the synthetic RK (RKs) was eluted with natural RK on analytical C18 reversed-phase HPLC. Mass spectrometry RKs gave an experimental value of 1780.05 Da, which is in agreement with the natural RK molecular mass. RK PEPTIDE 17 mer-EXAMPLE 8
In vivo toxicity and tumor cells viability
[275] Evaluation of the toxicity of RK and RKs revealed that both peptides do not exhibit any toxicity up to a 2 pg/20g mice or 100 pg/kg body weight as determined by intracerebro- ventricular injection. RKs was tested on the viability of glioblastoma (U87), melanoma (IGR-39) and Rat pheochromocytoma (PC12) cell lines using the MTT assay. At 100 mM concentration, RKs does not affect the viability of all three cell lines including after 24h, 48h or 72h incubation time (Fig 6).
RK PEPTIDE 17 mer-EXAMPLE 8
RKs inhibits cell adhesion
[276] In order to investigate the effects of RKs, we performed cell adhesion assays by using a large array of purified ECM proteins (collagen IV (Coll IV), fibronectin (Fn), fibronogen (Fg), laminin-1 (Lam)) and the non-specific substratum Poly-L-Lysine (PLL). RKs blocked notably the adhesion of human glioblastoma cells U87 (Fig. 7A) and melanoma cells IGR39 (Fig. 7B) to fibrinogen and fibronectin, while no effect could be observed on collagen IV or laminin. RKs was able to inhibit adhesion of Rat pheochromocytoma cells (PC12) only to type IV collagen (Fig. 7C).
[277] Moreover, no inhibition could be observed on the integrin-independent substratum, poly-L-lysine, suggesting that the effect of RKs may involve the integrin family as adhesion receptors. As shown in Fig. 8C, RKs has the highest activity on PC12 cell adhesion to collagen IV. Inhibition of adhesion occurs in a dose-dependent manner with an IC5o value of 4.84 pM. RKs blocked notably the adhesion of human glioblastoma cells U87 cells to fibrinogen and fibronectin with IC5o values of 10 pM and 10.33 pM respectively (Fig. 8A). However the inhibitory effect of RKs on IGR39 cells on fibrinogen and fibronectin does not exceed 40% (Fig. 8B).
RK PEPTIDE 17 mer-EXAMPLE 9
RKs activity is selective to a1 b1 and anb3 integrins
[278] To identify the possible targeted integrins, we checked the RKs effect on various cell/ECM protein pairs involving in each case a unique integrin: cd b1 (PC12/type I collagen), a2b1 (HT1080/type I collagen), a5b1 (K562/fibronectin), anb5 (HT29-D4/vitronectin), anb6 (HT29- D4/fibronectin), a6b4 (HT29-D4/laminin) and anb3 (HT29-D4 transfected by 3 subunit/fibrinogen). As illustrated in Fig. 9A, RKs was not able to alter cell adhesion through a2b1 , anb6, anb5, a5b1 and a6b4 integrins, but significantly reduced the adhesive function of a1 b1 and anb3 integrins.
[279] We tested the effect of function-blocking antibodies against integrins cd b1 , anb3 and a5b1 on U87 cell adhesion to RKs used as a substrate. As shown in Fig. 9B, at the concentration of 5 pM of RKs, only antibodies against a1 b1 (60%), and anb3 (20%) abolished U87 cells attachment. Interestingly, the synthetic RGD peptide (1 mM) was able to inhibit more than 80% of human glioblastoma cell adhesion on RKs (Fig. 9B). These results suggest that the interaction between RKs and cells might involve an RGD-like motif.
RK PEPTIDE 17 mer-EXAMPLE 10
Computational study
[280] To understand the structural behavior of the RK peptide, the inventors constructed a tridimensionnel structure model using PEP-FOLD server which applies a de novo approach. In fact, the inventors failed to detect any significant homologous peptides with RK using BLAST2 and PSI-BLAST algorithms. PEP-FOLD server returned five different structures for RK. We selected the one which presents a disulfide bond closure between residues C3-C12. The Ca (The alpha carbon) distance between each of these amino acids is of 5.9 A (Angstrom) which still in the cutoff range of disulfide bond formation (4.4 to 6.8 A). The structure of the RK peptide consists of 3 sub-segments. The first one corresponds to the dipeptide 11 D2.
[281] The second one is the segment flanked by the two cystein residues (C3GTVMIPSEC12) which harbors a one-turn a helix consisting of residues 9-12. The third sub- segment is the C-terminal end corresponding to the DPKSS (aa 1 1-16 of SEQ ID NO: 1) sequence (Fig. 10A). Except for the two cystein residues, all the amino acids of RK are solvent exposed. The Accessible surface area calculated for each residue using GETAREA shows a minimum of exposure for E11 (53%) and a maximum (100%) for T5, P8, S16 and S17.
RK PEPTIDE 17 mer-EXAMPLE 11
Molecular dynamics of the RK peptide
[282] To gain more insight of the conformational properties of RK, the inventors run a molecular dynamics simulation of 300 ns (nanosecond). The RMSD (Root Mean Square Deviation) of the peptide Ca atoms shows broadly three phases on the plot. The first phase is very short consisting of an equilibration ending at the 8th nanosecond of the simulation. The second plateau phase lasts between the 8th and the 52nd nanosecond where the RMSD fluctuate approximately around an average value of 3.8 A relative to the starting structure. Until the end of this phase, the peptide preserves the overall conformation of the initial structure, mainly the one turn a helix. The following phase lasts for the rest of the trajectory where the RMSD values increased significantly to reach an average of 6.8 A but the values can reach more than 7 A. In fact, The RMSD at this phase is not as stable as its predecessor. We also noticed that RK peptide loses the overall initial conformation at this phase, in which the segment between the two cysteins shows a disordered structure (Fig. 10B). [283] To characterize the flexibility of RK, the inventors calculated the RMSF values per Ca atom. The C-terminal end of the peptide consisting in Si6 and S17 residues is the most flexible segment of RK. These amino acids show an RMSF values of 4.8 and 7.0 A, respectively (Fig. 10B). Residues 12-15 are the least flexible with RMSF (Root Mean Square Fluctuation) values ranging between 2.7 and 4.8 A. The RMSF values for the residues of central segment, 4- 12, seem to be more uniform which could be the result of a collective behavior of these amino acids. In addition to the previous analysis, the inventors established the energy landscape of the peptide based on the radius of gyration and the RMSD values as reaction coordinates. The Fig. 10C shows that the peptide describes mainly two low energy wells. The first one is the largest in which the conformation with the lowest energy preserves the one turn a helix. The well is highly populated and corresponds to a global minima. The second well is not as much populated as the first well in which the conformations present low energy values and corresponds to local minima. The two wells are separated by two other metastable local minima. The well representing the global minima of the free energy landscape of RK structure served to construct the docking ensemble of the ligand.
RK PEPTIDE 17 mer-EXAMPLE 10
Peptide-protein docking
[284] The clustering of the RK structure models leads to an ensemble of 13 structures. The receptor structure used in this study is the domain I of a1 integrin (Dial). It consists of an inserted segment near the N-terminus found only in a1 , a2 and a12 integrin sequences. For instance, in contrast to av s the MIDAS (metal-ion-dependent adhesion site) is harbored in Dial for the a1 integrin by which it interacts with the extracellular collagen molecules. There are eleven structures in NMR ensemble of Dial (PDB code: 2M32). To these, the inventors added three other conformations resulting from the output of ENCoM server after submitting the crystal unbound structure of Dial (PDB code: 1 PT6). As a result, a total number of 14 receptor conformers were processed in this study. Consequently, the inventors affected a number of 182 (13 for the ligand vs. 14 for the receptor) docking and a total of 364000 complexes were evaluated using the ZRANK scoring function.
[285] We end up with 14 conformers. At the final stage of the docking study the inventors run molecular dynamics simulations for nine complexes of 10 ns. Only three of them show a stable RMSD time evolution. The free energy of interaction, estimated by PRODIGY web server (Xue et al. , 2016)[29], are of -7.0, -7.3 and -8.4 kcal/mol. The docking solution with the lowest binding energy shows also a very stable profile of RMSD time evolution. In fact, the complex Ca RMSD fluctuates around 2.5 A (Fig. 11 A) starting from the second ns to the end of the simulation.
[286] We propose this docking solution as a model of interaction between the RK peptide and (Dial). The interaction interface between the peptide and the receptor covers the largest part of the binding site of the collagen. The l2 and D2 residues interact with the C Loop of Dial . M7 of RK seems to play an important role in the interaction. Its side chain occupies a small cavity situated away from the collagen interaction site. The cavity results from the 3D arrangement of two segments: 181-187 and 243-247 of the integrin (Notice that the amino acids numbering is based on the Uniprot accession P56199). W188 residues forms the interior of the cavity interacting with the methyl group of M7. In addition, the residue K15 of RK is able to establish a stable salt bridge with E285 of Dial (Fig. 11 B), with an average distance of 2.7 A during the 10 ns of molecular dynamics. K15 also establishes a stable hydrogen bond with D13 with an occupancy of 57% during the 10 ns simulation time (Fig. 11 C). It seems that this bond, reduces the rotameric flexibility and restraints K15 side chain to a close distance to E285 side chain which might contribute in a more stabilization of the salt bridge formation between the two amino acids. In such case, K15 has the role of a bridging amino acid between residues D13 of RK and E285 of Dial There is no extensive network of hydrogen bonds established between the peptide and the integrin. In fact, the only interaction of this type which is reasonably stable (20% occupancy of the total snapshots from the trajectory) is made between C3 of RK and G3I8 residue from the C loop segment of Dial .
INCORPORATION BY REFERENCE
[287] Various publications, articles and patents are cited or described in the background and throughout the specification; each of these references is herein incorporated by reference in its entirety. Discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is for the purpose of providing context for the invention. Such discussion is not an admission that any or all of these matters form part of the prior art with respect to any inventions disclosed or claimed.
REFERENCES
1. A.K. Weaver, X. Liu, H. Sontheimer, Role for calcium-activated potassium channels (BK) in growth control of human malignant glioma cells, J. Neurosci. Res. 78 (2004) 224-34.
2. A. Schwab, J. Reinhardt, S.W. Schneider, B. Gassner, B. Schuricht, K+ channel-dependent migration of fibroblasts and human melanoma cells, Cell. Physiol. Biochem. 9 (1999) 126- 32, domain receptor 1 (DDR1), as a potential biomarker for serous ovarian cancer.
International Journal of Molecular Sciences. 2011 , 12, 971-982. doi:
10.3390/ijms12020971. uo, W.; Giancotti, F.G. Integrin signalling during tumour progression. Nat Rev Mol. Cell Biol.
2004, 5, 816-826.
epparda, B.U. In vivo functions of integrins: lessons from null mutations in mice Dean.
Matrix Biol. 2000, 19, 203-209Guo, W.; Giancotti, F.G. Integrin signalling during tumour progression. Nat Rev Mol.
iancotti, F.G. Integrin signaling: specificity and control of cell survival and cell cycle progression. Curr Opin Cell Biol. 1997, 9, 691-700.
ddington, R.C.; Ginsberg M.H. Integrin activation takes shape. J Cell Biol. 2002, 158, 833-
839.
ahmberg, C.G.; Fagerholm, S.C.; Nurmi, S.M.; Chavakis, T.; Marchesan, S.; Gronholm, M.
Regulation of integrin activity and signalling. Biochimica et Biophysica Acta. 2009, 6, 431- 444. doi: 10.1016/j.bbagen.2009.03.007.
an der Flier, A.; Sonnenberg, A. Function and interactions of integrins. Cell Tissue
Research. 2001 , 3, 285-298.
ynes, R.O. Integrins: bidirectional, allosteric signaling machines. Cell. 2000, 1 10, 673-687. Hynes, R.O. Integrins: versatility, modulation, and signaling in cell adhesion. Cell. 1992, 69, 1 1-25.
Juarez, P.; Comas, I.; Gonzalez-Candelas, F.; Calvete, J.J. Evolution of snake venom disintegrins by positive Darwinian selection. Mol Biol Evol. 2008, 25, 2391-407. doi:
10.1093/molbev/msn179.
McLane, M.A.; Marcinkiewicz, C.; Vijay-Kumar, S.; Wierzbicka-Patynowski, I.;
Niewiarowski, S. Viper venom disintegrins and related molecules. Proc. Soc. Exp. Biol.
Med. 1998, 219, 109-1 19.
Springer, T.A.; Wang, J-h. The three-dimensional structure of integrins and their ligands, and conformational regulation of cell adhesion. Adv Protein Chem. 2004, 68, 29-63.
Lee, J.O.; Bankston, L.A.; Arnaout, M.A.; Liddington, R.C. Two conformations of the integrin A-domain (l-domain): A pathway for activation. Structure. 1995, 3, 1333-1340.
Heino, J. The collagen receptor integrins have distinct ligand recognition and signaling functions. Matrix Biol. 2000, 19, 319-23.
Shi, M.; Pedchenko, V.; Greer, B.H.; Van Horn, W.D.; Santoro, S.A.; Sanders, C.R.;
Hudson, B.G.; Eichman, B.F.; Zent, R., Pozzi, A. Enhancing integrin cd inserted (I) domain affinity to ligand potentiates integrin aΐ bΐ-mediated down-regulation of collagen synthesis.
J Biol Chem. 2012, 287, 35139-35152. doi: 10.1074/jbc.M1 12.358648.
enner, C.; Sacca, B.; Moroder, L. Synthetic heterotrimeric collagen peptides as mimics of cell adhesion sites of the basement membrane. Biopolymers. 2004, 76, 34-47.
rruda Macedo, J.K.; Fox, J.W.; de Souza Castro, M. Disintegrins from snake venoms and their applications in cancer research and therapy. Curr Protein Pept Sci. 2015, 16, 532-48. arcinkiewicz, C. Functional characteristic of snake venom disintegrins: Potential therapeutic implication. Current Pharmaceutical Design. 2005, 1 1 , 815-27.
alvete, J.J.; Marcinkiewicz, C.; Monleon, D.; Esteve, V.; Celda, B.; Juarez, P.; Sanz, L. Snake venom disintegrins: evolution of structure and function. Toxicon. 2005, 45, 1063- 1074.
ebin, J.A.; Maggio, J.E.; Strichartz, G.R. Purification and characterization of chlorotoxin, a chloride channel ligand from the venom of the scorpion. Am J Physiol. 1993, 264, 361-9. ardevet, L.; Rani, D.; Aziz, T.A.; Bazin, I.; Sabatier, J.M.; Fadl, M.; Brambilla, E.; De Waard, M. Chlorotoxin : a helpful natural scorpion peptide to diagnose glioma and fight tumor invasion. Toxins (Basel). 2015, 7, 1079-101. doi: 10.3390/toxins7041079.
e, Q.Y.; He, Q.Z.; Deng, X.C.; Yao, L.; Meng, E.; Liu, Z.H.; Liang, S.P. ATDB: a unidatabase platform for animal toxins. Nucleic Acids Res. 2008, 36(Database issue), D293- D297. doi: 10.1093/nar/gkm832
arvey, A.L. Toxins and drug discovery. Toxicon. 2014, 92, 193-200.
doi: 10.1016/j.toxicon.2014.10.020.
ita, C.; Roumestand, C.; Toma, F.; Menez, A. Scorpion toxins as natural scaffolds for protein engineering. Biochemistry. 1995, 92, 6404-6408.
eng, X.C.; Corzo, G.; Harin, R. Scorpion venom peptides without disulfide bridges. IUBMB Life. 2005, 57, 13-21.
EIFessi-Magouri, R.; Peigneur, S.; Houcemeddine, O.; Srairi-Abid, N.; El ayeb, M.; Jan, T.; Riadh, K. Characterization of Kbot21 Reveals Novel Side Chain Interactions of Scorpion Toxins Inhibiting Voltage Gated Potassium Channels. PLoS One. 2015, 10(9), e013761 1. doi: 10.1371/journal.pone.013761 1. eCollection 2015.
ue, L.C.; Rodrigues, J.P.; Kastritis, P.L.; Bonvin, A.M.; Vangone, A. PRODIGY: a web server for predicting the binding affinity of protein-protein complexes. Bioinformatics. 2016, 32, 3676-3678. doi:10.1093/bioinformatics/btw514
lexey, I.K.; Nikolay, A.K.; Anton, O.C.; Eugene, V.G.; Alexander, A.V. Kalium: a database of potassium channel toxins from scorpion venom. Database (Oxford). 2016, baw056. chola, J.B.; Lwande, W.; Thiong’o, T.; Rogo, L.; Herrmann, R.; Schepers, E.; Bagine, R.; Mungai, P.; Ndiege, I.O. Identification of insect-selective and mammal-selective toxins from Parabuthus leiosoma venom. Toxicon. 2007, 50, 449-56. doi:
10.1016/j.toxicon.2007.04.020
ingras, M.C.; Roussel, E.; Bruner, J.M.; Branch, C.D.; Moser, R.P. Comparison of cell adhesion molecule expression between glioblastoma multiforme and autologous normal brain tissue. J Neuroimmunol. 1995, 57, 143 53.
Siret, C.; Terciolo, C.; Dobric, A.; Habib, M.C.; germain, S.; Bonnier, R.; Lombardo, D.; Rigot, V.; Andre, F. Interplay between cadherins and a2b1 integrin differentially regulates melanoma cell invasion. Br J Cancer. 2015, 1 13, 1445-53.
Eva-maria, F.; Gustav-Paul, W.; Ute, K.B.; Bodo, H.; Simon, L.G.; Jan-Dirk, R.
Immunohistochemical analysis of integrins anb3, anb5 and a5b1 , and their ligands, fibrinogen, fibronectin, osteopontin and vitronectin, in frozen sections of human oral head and neck squamous cell carcinomas. Exp Ther Med. 201 1 , 2, 9-19.
Sarray, S.; Berthet, V.; Calvete, J.J.; Secchi, J.; Marvaldi, J.; El-Ayeb, M.; Marrakchi, N.; Luis, J. Lebectin, a novel C-type lectin from Macrovipera lebetina venom, inhibits integrin- mediated adhesion, migration and invasion of human tumour cells. Lab. Invest. 2004, 84, 573-581.
Tomaselli, K.J.; Hall, D.E.; Flier, L.A.; Gehlsen, K.R.; Turner, D.C.; Carbonetto, S.;
Reichardt, L.F. A neuronal cell line (PC12) expresses two beta 1 -class integrins-alpha 1 beta 1 and alpha 3 beta 1 -that recognize different neurite outgrowth-promoting domains in laminin. Neuron. 1990, 5, 651-62.
Morjen, M.; Kallech-Ziri, O.; Bazaa, A.; Othman, H.; Mabrouk, K.; Zouari-Kessentini, R.; Sanz, L.; Calvete, J.J.; Srairi-Abid, N.; El Ayeb, M.; Luis, J.; Marrakchi, N. PIVL, a new serine protease inhibitor from Macrovipera lebetina transmediterranea venom, impairs motility of human glioblastoma cells. Matrix Biol. 2013, 32, 52-62. doi:
10.1016/j.matbio.2012.11.015.
Smith, J.W., Piotrowicz, R.S., Mathis, D. A mechanism for divalent cation regulation of beta 3-integrins. J Biol Chem. 1994, 269, 960-7.
Arnaout, M.A.; Mahalingam, B.; Xiong, J.P. Integrin structure, allostery, and bidirectional signaling. Annu Rev Cell Dev Biol. 2005, 21 , 381-410.
Wang, W.J. Acurhagin-C, an ECD disintegrin, inhibits integrin alphavbeta3-mediated human endothelial cell functions by inducing apoptosis via caspase-3 activation. Br J Pharmacol. 2010, 160, 1338-51.
Selistre-de-Araujo, H.S.; Pontes, C.L.; Montenegro, C.F.; Martin, A.C. Snake venom disintegrins and cell migration. Toxins (Basel). 2010, 2, 2606-21. Limam, I.; Bazaa, A.; Srairi-Abid, N.; Taboubi, S.; Jebali, J.; Zouari-Kessentini, R.; Kallech- Ziri, O.; Mejdoub, H.; Hammami, A.; El Ayeb, M.; Luis, J.; Marrakchi, N. Leberagin-C, A disintegrin-like/cysteine-rich protein from Macrovipera lebetina transmediterranea venom, inhibits alphavbeta3 integrin-mediated cell adhesion. Matrix Biol. 2010, 29, 1 17-126.
Olfa, K.Z.; Jose, L; Salma, D.; Amine, B.; Najet, S.A.; Nicolas, A.; Maxime, L.; Raoudha, Z.; Kamel, M.; Jacques, M., Jean-Marc, S.; Mohamed el, A.; Naziha, M. Lebestatin, a disintegrin from Macrovipera venom, inhibits integrin-mediated cell adhesion, migration and angiogenesis. Lab. Invest. 2005, 85, 1507-1516. doi: 10.1038/labinvest.3700350
Kisiel, D.G.; Calvete, J.J.; Katzhendler, J.; Fertala, A.; Lazarovici, P.; Marcinkiewicz, C. Structural determinants of the selectivity of KTS-disintegrins for the alphal betal integrin. FEBS Lett. 2004, 577, 478-82. doi: 10.1016/j.febslet.2004.10.050
Nolte, M.; Pepinsky, R.B.; Venyaminov, S.Y.; Koteliansky, V.; Gotwals, P.J.; Karpusas, M. Crystal structure of the a1 b1 integrin l-domain: insights into integrin l-domain function. FEBS Lett. 1999, 452, 379-385.
Rich, R.L.; Deivanayagam, C.C.; Owens, R.T.; Carson, M.; Hook, A.; Moore, D.;
Symersky, J.; Yang, V.W.; Narayana, S.V.; Hook, M. Trench- shaped binding sites promote multiple classes of interactions between collagen and the adherence receptors, a1 b1 integrin and Staphylococcus aureus cna MSCRAMM. J Biol Chem. 1999, 274, 24906- 24913.
Salminen, T.A.; Nymalm, Y.; Kankare, J.; Kapyla, J.; Heino, J.; Johnson, M.S. Production, crystallization and preliminary X-ray analysis of the human integrin al l domain. Acta Crystallogr. D Biol. Crystallogr. 1999, 55, 1365-1367.
Nymalm, Y.; Puranen, J.S.; Nyholm, T.K.; Kapyla, J.; Kidron, H.; Pentikainen, O. T.;
Airenne, T.T.; Heino, J., Slotte, J.P.; Johnson, M.S.; Salminen, T.A. Jararhagin-derived RKKH peptidesinduce structural changes in al l domain of human integrin a1 b1. J. Biol. Chem. 2004, 279, 7962-7970.
Brown, M.C.; Eble, J.A.; Calvete, J.J.; Marcinkiewicz. Structural requirements of KTS- disintegrins for inhibition of alpha(1)beta(1) integrin. Biochem J. 2009, 417, 95-101. doi:
10.1042/BJ20081403.
Moreno-Murciano, P.; Daniel Monleon, D.; Calvete, J.J.; Celda, B.; Marcinkiewicz, C. Amino acid sequence and homology modeling of obtustatin, a novel non-RGD-containing short disintegrin isolated from the venom of Vipera lebetina obtusa. Protein Sci. 2003, 12, 366-371. doi: 10.1 1 10./ps.0230203
Shih, C.H.; Chiang, T.B.; Wang, W.J. Inhibition of integrins an/ad-dependent functions in melanoma cells by an ECD-disintegrin acurhagin-C. Matrix Biol. 2013, 32, 152-159.
Xiong, J.P.; Stehle, T.; Diefenbach, B.; Zhang, R.; Dunker, R.; Scott, D.L.; Joachimiak, A.; Goodman, S.L.; Arnaout, M.A. Crystal structure of the extracellular segment of integrin anb3. Science (NY). 2001 , 294, 339-345.
Xiong, J.P.; Stehle, T.; Zhang, R.; Joachimiak, A.; Freeh, M.; Goodman, S.L.; Arnaout, M.A. Crystal structure of the extracellular segment of integrin a\/b3 in complex with an Arg- Gly-Asp ligand. Science (NY). 2002, 296, 151-155.
Martin, M.F.; Rochat, H. Purification of thirteen toxins active on mice from the venom of the North African scorpion Buthus occitanus tunetanus. Toxicon. 1984, 22, 279-92.
Mahjoubi-Boubaker, B.; Crest, M.; Ben Khalifa, R.; El Ayeb, M.; Kharrat, R. Kbotl , a three disulfide bridges toxin from Buthus occitanus tunetanus venom highly active on both SK and Kv channels. Peptides. 2004, 25, 637-45.

Claims

CLAIMS We claim:
1. An isolated peptide selected from the group consisting of peptides whose amino acid sequence comprises or consists of:
IDCSKVNLTAECSS (peptide RK1 ; SEQ ID NO:2);
IDCGTVMIPSECDPKS (peptide RK; SEQ ID NO:3);
a sequence has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 91 %, at least about 92%, at least about 93%, at least about 94%, at last about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity to that of RK1 ; and
a sequence has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 91 %, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity to that of RK.
2. The peptide according to claim 1 , comprising at least one substitution modification, insertion, or deletion modification relative to IDCSKVNLTAECSS (SEQ ID NO:2) and I DCGTVM I PSECDPKSS (SEQ ID NO: 1).
3. The peptide according to any one of claims 1-2, wherein the sequence comprises:
a.) the segment V6, M7, and l8 of the RK peptide (aa 6-8 of SEQ ID NO: 1), and/or
Sn ECDPKSis (aa 1 1-16 of SEQ ID NO: 1), and/or Ei2CDPKi5 (aa 12-15 of SEQ ID NO:1), and/or D14PK16 (aa 14-16 of SEQ ID NO: 1), of the RK peptide and, optionally, still binds a1 b1 integrin; or
b.) DCSK (aa 2-5 of SEQ ID NO:1), DCSKXXXXXECS (SEQ ID NO:4), and/or
DCSKXXXXXECDS (SEQ ID NO:5) of the RK1 peptide.
4. The peptide according to any one of claims 1-3, wherein the sequence comprises residues E12CDPK15 (aa 12-15 of SEQ ID NO: 1) of the RK peptide.
5. The peptide according to any one of claims 1-4, wherein the peptide is no more than 20 amino acids long.
6. The peptide according to any one of claims 1-5, wherein the peptide is 17 amino acids long.
7. The peptide according to any one of claims 1-6, wherein the peptide is 14 amino acids long.
8. The peptide according to any one of claims 1-7, wherein the amino acid sequence is IDCGTVMIPSECDP (aa 1-14 of SEQ ID NO: 1).
9. The peptide according to any one of claims 1-8, wherein the amino acid sequence is IDCSKVNLTAECSS (SEQ ID NO:2) or IDCGTVMIPSECDPKSS (RK; SEQ ID NO:1).
10. The peptide according to any one of claims 1-9, wherein the peptide is isolated from Buthus occitanus tunetanus scorpion venom.
11. The peptide according to any one of claims 1-10, wherein the peptide is produced synthetically.
12. The peptide according to any one of claims 1-11 , wherein the peptide comprises a disulfide bond.
13. The peptide according to any one of claims 1-12, comprising an N-terminal and/or C-terminal modification.
14. The peptide according to of any one of claims 1-13, wherein the peptide is amidated at its C-terminal end.
15. The peptide according to any one of claims 1-14, wherein the peptide is conjugated to a half-life extending moiety.
16. The peptide according to any one of claims 1-15, wherein the peptide is linked to a polymer.
17. The peptide according to any one of claims 1-16, wherein the polymer is chosen from polyalkylethers (e.g. polyethylene glycol and polypropylene glycol), polylactic acid, polyglycolic acid, polyoxyalkenes, polyvinylalcohol, polyvinylpyrrolidone, cellulose and cellulose derivatives, dextran, and dextran derivatives, polyvinyl pyrrolidones, glycopeptides, and polyamino acids.
18. The peptide according to any one of claims 1-17, wherein the peptide is linked to a coupling partner.
19. The peptide according to any one of claims 1-18, wherein the coupling partner is chosen from an effector molecule, a label, a drug, a toxin, a carrier, and a transport molecule.
20. The peptide according to any one of claims 1-19, wherein the peptide is modified by the addition of cysteine or biotin.
21. The peptide according to any one of claims 1-20, wherein the peptide is modified by a moiety to facilitate crosslinking.
22. The peptide according to any one of claims 1-21 , wherein the moiety is chosen from benzophenone, maleimide, and activated esters.
23. The peptide according to any one of claims 1-22, wherein the peptide is not able to alter cell adhesion through a2b1 , anbq, anb5, a5b1 and a6b4 integrins, but significantly reduces the adhesive activity of a1 b1 and anb3 integrins to extracellular matrix proteins.
24. The peptide according to any one of claims 1-23, wherein the extracellular matrix protein comprises collagen.
25. The peptide according to any one of claims 1-24, wherein the extracellular matrix protein comprises vitronectin, fibronectin, fibrinogen, osteopoietin, and/or bone sialoprotein.
26. The peptide according to any one of claims 1-25, wherein the peptide competes with a synthetic RGD peptide for binding to aibi and/or anb3 integrins
27. A composition comprising the peptide according to any one of claims 1-26 and a pharmaceutically acceptable carrier, diluent, or excipient.
28. The composition of claim 27, wherein the composition is a sustained release formulation or is in a sustained release carrier.
29. The composition according to any one of claims 27-28, wherein the composition is formulated to be administered in microspheres, liposomes, or one of other microparticulate delivery systems.
30. The composition according to any one of claims 27-29, wherein the sustained release carrier comprises a semipermeable polymer matrix.
31. The composition according to any one of claim 27-30, wherein the
semipermeable polymer matrix comprises one or more of polylactides copolymers of L-glutamic acid and gamma ethyl-L-glutamate, poly (2-hydroxyethyl-methacrylate), or ethylene vinyl acetate.
32. A method of treating cancer in a subject in need thereof, comprising
administering to the subject a therapeutically effective amount of a peptide according to any one of claims 1-26 or a composition according to any one of claims 27-31.
33. The method of claim 32, wherein the peptide is administered at a dosage ranging from 0.00003 mg/kg/day to 30 mg/kg/day.
34. The method according to any one of claims 32-33 or 45-60, wherein the peptide is administered at a dosage ranging from 0.003 mg/kg/ day to 3 mg/kg/day.
35. The method according to any one of claims 32-34 or 45-60, wherein the peptide is administered at a dosage ranging from 0.03 mg/kg/day to 0.3 mg/kg/day.
36. The method according to any one of claims 32-35 or 45-60, wherein the peptide is administered at a dosage ranging from 0.1 to 50 mg/kg/day, typically from about 0.5 to 1 mg/kg/day.
37. The method according to any one of claims 32-25 or 45-60, wherein the dosage of the peptide to be achieved in vivo may be equivalent to an in vitro level of 1 mM, 2 mM, 3 pM,
4 pM, 5 pM, 6 pM, 7 pM, 1 pM, 8 pM, 9 pM, 10 pM, 20 pM, 30 pM, 40 pM, 50 pM, 70 pM, 80 pM, 90 pM, or 100 pM.
38. The method according to any one of claims 32-25 or 45-60, wherein the dosage of the peptide to be achieved in vivo may be a blood/plasma level of at least 1 pM, 2 pM, 3 pM,
4 pM, 5 pM, 6 pM, 7 pM, 1 pM, 8 pM, 9 pM, 10 pM, 20 pM, 30 pM, 40 pM, 50 pM, 70 pM, 80 pM, 90 pM, or 100 pM.
39. The method according to any one of claims 32-38 or 45-60, wherein the peptide is administered intravenously continuously for more than one day.
40. The method according to any one of claims 32 39 or 45-60, wherein the composition is administered orally, parenterally, topically, by inhalation, intranasally, or rectally.
41. The method according to any one of claims 32-40 or 45-60, wherein the composition is administered intravenously, oral, sublingual, intranasal, intraocular, rectal, transdermal, mucosal, topical or parenteral.
42. The method according to any one of claims 32-41 or 45-60, wherein the composition comprises at least one additive chosen from a pharmaceutically acceptable excipients, carriers, preservatives, buffers, stabilizers, antioxidants, or other additives.
43. The method according to any one of claim 32-42 or 45-60, wherein the composition is in a form chosen from a tablet, capsule, powder, or liquid.
44. The method according to any one of claim 32-43 or 45-60, wherein the liquid comprises at least one additive chosen from liquid carriers, petroleum, animal oils, vegetable oils, mineral oils, synthetic oils, physiological saline solutions, saccharide solutions, and glycols.
45. The method according to any one of claim 32-44 or 45-60, wherein the cancer is selected from glioblastoma, melanoma, leukemia, and hepatocellular cancers, sarcoma, vascular endothelial cancers, breast cancers, central nervous system cancers (e.g.
astrocytoma, gliosarcoma, neuroblastoma, oligodendroglioma and glioblastoma), prostate cancers, lung and bronchus cancers, larynx cancers, esophagus cancers, colon cancers, colorectal cancers, gastro-intestinal cancers, melanomas, ovarian and endometrial cancer, renal and bladder cancer, liver cancer, endocrine cancer (e.g. thyroid), and pancreatic cancer.
46. A method of inhibiting angiogenesis in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a peptide according to any one of claims 1-26 or a composition according to any one of claims 27-31.
47. A method for treatment of an integrin-associated disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a peptide according to any one of claims 1-26 or a composition according to any one of claims 27-31.
48. The method of claim 45 wherein the integrin-associated disease is osteoporosis, bone tumor or cancer growth, angiogenesis-related tumor growth and metastasis, tumor metastasis in bone, malignancy-induced hypercalcemia, angiogenesis-related eye diseases, Paget's disease, rheumatic arthritis, ovariectomy-induced physiological change, inflammation, coagulation diseases, or osteoarthritis.
49. The method according to any one of claims 32-48, wherein the method further comprises administering to the subject a therapeutically effective amount of one or more other therapeutic agents.
50. The method according to claim 49, wherein said therapeutic agent is selected from the group consisting of an antiangiogenic agent, a cytotoxic agent, a cytostatic agent, and an immunomodulatory agent.
51. The method according to any one of claim 32-50, wherein the composition is administered in combination with radiotherapy.
52. The method according to claim 50, wherein the at least one an antiangiogenic agent, a cytotoxic agent, a cytostatic agent comprises at least one agent chosen from endostatin, angiostatin, VEGF inhibitors, cytotoxic agents, alkaloids, antimetabolites, cancer growth inhibitors, gene therapy therapeutics, cancer vaccines, interferons, monoclonal antibodies, radiotherapy, hormonal therapy, and other supportive therapy.
53. The method according to any one of claims 32-52, wherein the therapeutic agent is selected from Alkylating Agents, DNA Alkylating-like Agents, Alkylating Antineoplastic Agents, DNA replication and repair inhibitors, Cell Cycle Modulators, Apoptosis Regulators,
Angiogenesis Inhibitors, Proteasome Inhibitors, Kinase Inhibitors, Protein Synthesis Inhibitors, Histone deacetylase inhibitors, Topoisomerase I Inhibitors, Topoisomerase II Inihibitors, DNA Intercalating Agents, RNA/DNA Antimetabolites, DNA Antimetabolites, Mitochondria Inhibitors, Antimitotic Agents, Nuclear Export Inhibitors, and Hormonal Therapies.
54. The method according to any one of claims 32-53, wherein therapeutically effective means that the administration of the composition to the subject results in any demonstrated clinical benefit compared with no therapy (when appropriate) or to a known standard of care.
55. The method according to any one of claim 54, wherein clinical benefit is assessed based on objective response rate (ORR) (determined using RECIST version 1.1), duration of response (DOR), progression-free survival (PFS), and/or overall survival (OS).
56. A method of decreasing binding of aibi and/or anb3 integrins expressing cells to extracellular molecules, comprising contacting the cells with according to any one of claims 1-26 or a composition according to any one of claims 27-31.
57. A method of screening for inhibition of tumor cell growth, comprising
administering a peptide according to any one of claims 1-26 or a composition according to any one of claims 27-31 to a subject, and determining whether the peptide reduces tumor cell growth.
58. A method of inhibiting tumor cell proliferation, comprising contacting the tumor cell with an effective amount of a peptide according to any one of claims 1-26 or a composition according to any one of claims 27-31.
59. A method of inhibiting tumor cell migration, comprising contacting the tumor cell with an effective amount of a peptide according to any one of claims 1-26 or a composition according to any one of claims 27-31.
60. The method according to any one of claims 55-59, wherein the tumor cells are glioblastoma cells or melanoma cells.
61. A method of designing or screening for a disintegrin peptide or other integrin inhibitor, wherein the method comprises using the model described in the figures as a basis for the design or screening.
62. A peptide according to any one of claims 1-26 as a medicament.
63. A composition according to any one of claim 27-31 as a medicament.
64. A peptide according to any one of claims 1-26 for the treatment of cancer, inhibiting tumor cell migration, inhibiting tumor cell proliferation, decreasing binding of aibi and/or anb3 integrins expressing cells to extracellular molecules, treatment of an integrin- associated disease, and/or inhibiting angiogenesis.
65. A composition according to any one of claim 27-31 for the treatment of cancer, inhibiting tumor cell migration, inhibiting tumor cell proliferation, decreasing binding of aibi and/or anb3 integrins expressing cells to extracellular molecules, treatment of an integrin- associated disease, and/or inhibiting angiogenesis.
66. The use of a peptide according to any one of claims 1-26 or a composition according to anyone of claims 27-31 for the preparation of a drug for the treatment of cancer, inhibiting tumor cell migration, inhibiting tumor cell proliferation, decreasing binding of aibi and/or anb3 integrins expressing cells to extracellular molecules, treatment of an integrin- associated disease, and/or disorders mediated by/associated with angiogenesis.
67. The use of a peptide according to any one of claims 1-26 or a composition according to anyone of claims 27-31 for the preparation of a drug for the treatment of epilepsy, memory, learning, neuropsychiatric, neurological, neuromuscular, and immunological disorders, schizophrenia, bipolar disorder, sleep apnea, neurodegeneration, smooth muscle disorders, bacterial diseases, fungal diseases, malaria, viral diseases, Immuno-modulator-responsive disorders or pain.
68. The peptide according to any one of claims 1-26, or a composition according to anyone of claims 27-31 for use as a medicament.
69. The peptide according to any one of claims 1-26, or a composition according to anyone of claims 27-31 for use as a medicinal product intended for the treatment of cancer, inhibiting tumor cell migration, inhibiting tumor cell proliferation, decreasing binding of aibi and/or anb3 integrins expressing cells to extracellular molecules, treatment of an integrin- associated disease, disorders mediated by/associated with angiogenesis, epilepsy, memory, learning, neuropsychiatric, neurological, neuromuscular, and immunological disorders, schizophrenia, bipolar disorder, sleep apnea, neurodegeneration, smooth muscle disorders, bacterial diseases, fungal diseases, malaria, viral diseases, Immuno-modulator-responsive disorders, or pain.
70. A kit comprising a peptide according to any one of claims 1-26 or a composition according to any one of claims 27-31.
PCT/IB2019/052358 2018-03-28 2019-03-22 Anti-cancer and disintegrin scorpion venoms WO2019186344A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862649151P 2018-03-28 2018-03-28
US62/649,151 2018-03-28

Publications (1)

Publication Number Publication Date
WO2019186344A1 true WO2019186344A1 (en) 2019-10-03

Family

ID=66448593

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2019/052358 WO2019186344A1 (en) 2018-03-28 2019-03-22 Anti-cancer and disintegrin scorpion venoms

Country Status (1)

Country Link
WO (1) WO2019186344A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113384682A (en) * 2021-05-31 2021-09-14 南方医科大学 Application of scorpion venom polypeptide Smp43 in preparation of antitumor drugs

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0413622A1 (en) 1989-08-03 1991-02-20 Rhone-Poulenc Sante Albumin derivatives with therapeutic functions
US5766883A (en) 1989-04-29 1998-06-16 Delta Biotechnology Limited Polypeptides
US5876969A (en) 1992-01-31 1999-03-02 Fleer; Reinhard Fusion polypeptides comprising human serum albumin, nucleic acids encoding same, and recombinant expression thereof
US20070092926A1 (en) * 2005-10-14 2007-04-26 Alterman Michail A Analysis of protein isoforms using unique tryptic peptides by mass spectrometry and immunochemistry

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5766883A (en) 1989-04-29 1998-06-16 Delta Biotechnology Limited Polypeptides
EP0413622A1 (en) 1989-08-03 1991-02-20 Rhone-Poulenc Sante Albumin derivatives with therapeutic functions
US5876969A (en) 1992-01-31 1999-03-02 Fleer; Reinhard Fusion polypeptides comprising human serum albumin, nucleic acids encoding same, and recombinant expression thereof
US20070092926A1 (en) * 2005-10-14 2007-04-26 Alterman Michail A Analysis of protein isoforms using unique tryptic peptides by mass spectrometry and immunochemistry

Non-Patent Citations (67)

* Cited by examiner, † Cited by third party
Title
A. SCHWAB; J. REINHARDT; S.W. SCHNEIDER; B. GASSNER; B. SCHURICHT: "K+ channel-dependent migration of fibroblasts and human melanoma cells", CELL. PHYSIOL. BIOCHEM., vol. 9, 1999, pages 126 - 32
A.K. WEAVER; X. LIU; H. SONTHEIMER: "Role for calcium-activated potassium channels (BK) in growth control of human malignant glioma cells", J. NEUROSCI. RES., vol. 78, 2004, pages 224 - 34
ALEXEY, I.K.; NIKOLAY, A.K.; ANTON, O.C.; EUGENE, V.G.; ALEXANDER, A.V.: "Kalium: a database of potassium channel toxins from scorpion venom", DATABASE (OXFORD, 2016, pages baw056
ALTSCHUL ET AL., METHODS IN ENZYMOLOGY, vol. 266, 1996, pages 460 - 480
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1991, pages 3389 - 3402
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402
ARNAOUT, M.A.; MAHALINGAM, B.; XIONG, J.P.: "Integrin structure, allostery, and bidirectional signaling", ANNU REV CELL DEV BIOL, vol. 21, 2005, pages 381 - 410
ARRUDA MACEDO, J.K.; FOX, J.W.; DE SOUZA CASTRO, M.: "Disintegrins from snake venoms and their applications in cancer research and therapy", CURR PROTEIN PEPT SCI, vol. 16, 2015, pages 532 - 48
BENNASR HMED ET AL: "Scorpion Peptides: Potential Use for New Drug Development", JOURNAL OF TOXICOLOGY, vol. 2013, 2013, US, pages 1 - 15, XP055593703, ISSN: 1687-8191, DOI: 10.1155/2013/958797 *
BROWN, M.C.; EBLE, J.A.; CALVETE, J.J.: "Marcinkiewicz. Structural requirements of KTS-disintegrins for inhibition of alpha(1)beta(1) integrin", BIOCHEM J., vol. 417, 2009, pages 95 - 101
CALVETE, J.J.; MARCINKIEWICZ, C.; MONLEON, D.; ESTEVE, V.; CELDA, B.; JUAREZ, P.; SANZ, L.: "Snake venom disintegrins: evolution of structure and function", TOXICON, vol. 45, 2005, pages 1063 - 1074, XP004938215, DOI: doi:10.1016/j.toxicon.2005.02.024
DARDEVET, L.; RANI, D.; AZIZ, T.A.; BAZIN, I.; SABATIER, J.M.; FADL, M.; BRAMBILLA, E.; DE WAARD, M.: "Chlorotoxin : a helpful natural scorpion peptide to diagnose glioma and fight tumor invasion", TOXINS (BASEL, vol. 7, 2015, pages 1079 - 101, XP055411747, DOI: doi:10.3390/toxins7041079
DATABASE CA [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 2003, LI, SHUN-YI ET AL.: "Isolation and sequencing of five variants of phospholipases A2 from venom of snake Trimeresurus stejnegeri", XP002791749, Database accession no. 139:97143 *
DATABASE UniProtKB [online] 1 March 2001 (2001-03-01), "Trimeresurus stejnegeri", XP002792192, retrieved from UniProt Database accession no. P82896 *
DEBIN, J.A.; MAGGIO, J.E.; STRICHARTZ, G.R.: "Purification and characterization of chlorotoxin, a chloride channel ligand from the venom of the scorpion", AM J PHYSIOL., vol. 264, 1993, pages 361 - 9
E.A. RIVAS-MERCADO ET AL: "Disintegrins obtained from snake venom and their pharmacological potential", MEDICINA UNIVERSITARIA, vol. 19, no. 74, 15 July 2017 (2017-07-15), pages 32 - 37, XP055593705, ISSN: 1665-5796, DOI: 10.1016/j.rmu.2017.02.004 *
EIFESSI-MAGOURI, R.; PEIGNEUR, S.; HOUCEMEDDINE, O.; SRAIRI-ABID, N.; EL AYEB, M.; JAN, T.; RIADH, K.: "Characterization of Kbot21 Reveals Novel Side Chain Interactions of Scorpion Toxins Inhibiting Voltage Gated Potassium Channels", PLOS ONE, vol. 10, no. 9, 2015, pages e0137611
EVA-MARIA, F.; GUSTAV-PAUL, W.; UTE, K.B.; BODO, H.; SIMON, L.G.; JAN-DIRK, R.: "Immunohistochemical analysis of integrins av(33, av(35 and α5β1, and their ligands, fibrinogen, fibronectin, osteopontin and vitronectin, in frozen sections of human oral head and neck squamous cell carcinomas", EXP THER MED, vol. 2, 2011, pages 9 - 19
GAHMBERG, C.G.; FAGERHOLM, S.C.; NURMI, S.M.; CHAVAKIS, T.; MARCHESAN, S.; GRONHOLM, M.: "Regulation of integrin activity and signalling", BIOCHIMICA ET BIOPHYSICA ACTA, vol. 6, 2009, pages 431 - 444, XP026169761, DOI: doi:10.1016/j.bbagen.2009.03.007
GIANCOTTI, F.G.: "Integrin signaling: specificity and control of cell survival and cell cycle progression", CURR OPIN CELL BIOL, vol. 9, 1997, pages 691 - 700, XP001180693, DOI: doi:10.1016/S0955-0674(97)80123-8
GINGRAS, M.C.; ROUSSEL, E.; BRUNER, J.M.; BRANCH, C.D.; MOSER, R.P.: "Comparison of cell adhesion molecule expression between glioblastoma multiforme and autologous normal brain tissue", J NEUROIMMUNOL., vol. 57, 1995, pages 143 53
GUO, W.; GIANCOTTI, F.G.: "Integrin signalling during tumour progression", NAT REV MOL
GUO, W.; GIANCOTTI, F.G.: "Integrin signalling during tumour progression", NAT REV MOL. CELL BIOL., vol. 5, 2004, pages 816 - 826
HARVEY, A.L.: "Toxins and drug discovery", TOXICON, vol. 92, 2014, pages 193 - 200
HE, Q.Y.; HE, Q.Z.; DENG, X.C.; YAO, L.; MENG, E.; LIU, Z.H.; LIANG, S.P.: "ATDB: a uni-database platform for animal toxins", NUCLEIC ACIDS RES., vol. 36, 2008, pages D293 - D297
HEINO, J.: "The collagen receptor integrins have distinct ligand recognition and signaling functions", MATRIX BIOL., vol. 19, 2000, pages 319 - 23
HYNES, R.O.: "Integrins: bidirectional, allosteric signaling machines", CELL, vol. 110, 2000, pages 673 - 687
HYNES, R.O.: "Integrins: versatility, modulation, and signaling in cell adhesion", CELL, vol. 69, 1992, pages 11 - 25, XP023884392, DOI: doi:10.1016/0092-8674(92)90115-S
INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 12, 2011, pages 971 - 982
JUAREZ, P.; COMAS, I.; GONZALEZ-CANDELAS, F.; CALVETE, J.J.: "Evolution of snake venom disintegrins by positive Darwinian selection", MOL BIOL EVOL., vol. 25, 2008, pages 2391 - 407, XP002739341, DOI: doi:10.1093/MOLBEV/MSN179
KARLIN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 87, 1990, pages 2264 - 2268
KARLIN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 5873 - 5877
KHAMESSI OUSSEMA ET AL: "RK1, the first very short peptide fromButhus occitanus tunetanusinhibits tumor cell migration, proliferation and angiogenesis", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, ELSEVIER, AMSTERDAM, NL, vol. 499, no. 1, 31 January 2018 (2018-01-31), pages 1 - 7, XP085365983, ISSN: 0006-291X, DOI: 10.1016/J.BBRC.2018.01.133 *
KISIEL, D.G.; CALVETE, J.J.; KATZHENDLER, J.; FERTALA, A.; LAZAROVICI, P.; MARCINKIEWICZ, C.: "Structural determinants of the selectivity of KTS-disintegrins for the alpha1 beta1 integrin", FEBS LETT, vol. 577, 2004, pages 478 - 82
LEE, J.O.; BANKSTON, L.A.; ARNAOUT, M.A.; LIDDINGTON, R.C.: "Two conformations of the integrin A-domain (I-domain): A pathway for activation", STRUCTURE, vol. 3, 1995, pages 1333 - 1340, XP004587940, DOI: doi:10.1016/S0969-2126(01)00271-4
LIDDINGTON, R.C.; GINSBERG M.H.: "Integrin activation takes shape", J CELL BIOL., vol. 158, 2002, pages 833 - 839
LIMAM, I.; BAZAA, A.; SRAIRI-ABID, N.; TABOUBI, S.; JEBALI, J.; ZOUARI-KESSENTINI, R.; KALLECH-ZIRI, O.; MEJDOUB, H.; HAMMAMI, A.;: "Leberagin-C, A disintegrin-like/cysteine-rich protein from Macrovipera lebetina transmediterranea venom, inhibits alphavbeta3 integrin-mediated cell adhesion", MATRIX BIOL, vol. 29, 2010, pages 117 - 126, XP026911424, DOI: doi:10.1016/j.matbio.2009.09.009
MAHJOUBI-BOUBAKER, B.; CREST, M.; BEN KHALIFA, R.; EL AYEB, M.; KHARRAT, R.: "Kbot1, a three disulfide bridges toxin from Buthus occitanus tunetanus venom highly active on both SK and Kv channels", PEPTIDES, vol. 25, 2004, pages 637 - 45
MARCINKIEWICZ, C.: "Functional characteristic of snake venom disintegrins: Potential therapeutic implication", CURRENT PHARMACEUTICAL DESIGN, vol. 11, 2005, pages 815 - 27, XP002585548
MARTIN, M.F.; ROCHAT, H.: "Purification of thirteen toxins active on mice from the venom of the North African scorpion Buthus occitanus tunetanus", TOXICON, vol. 22, 1984, pages 279 - 92
MCLANE, M.A.; MARCINKIEWICZ, C.; VIJAY-KUMAR, S.; WIERZBICKA-PATYNOWSKI, I.; NIEWIAROWSKI, S.: "Viper venom disintegrins and related molecules", PROC. SOC. EXP. BIOL. MED., vol. 219, 1998, pages 109 - 119
MERRIFIELD, R. B.: "Solid-phase peptide synthesis", ADV. ENZYMOL. RELAT. AREAS MOL. BIOL., vol. 32, 1969, XP002957003
MORENO-MURCIANO, P.; DANIEL MONLEON, D.; CALVETE, J.J.; CELDA, B.; MARCINKIEWICZ, C.: "Amino acid sequence and homology modeling of obtustatin, a novel non-RGD-containing short disintegrin isolated from the venom of Vipera lebetina obtusa", PROTEIN SCI., vol. 12, 2003, pages 366 - 371
MORJEN, M.; KALLECH-ZIRI, O.; BAZAA, A.; OTHMAN, H.; MABROUK, K.; ZOUARI-KESSENTINI, R.; SANZ, L.; CALVETE, J.J.; SRAIRI-ABID, N.;: "PIVL, a new serine protease inhibitor from Macrovipera lebetina transmediterranea venom, impairs motility of human glioblastoma cells", MATRIX BIOL, vol. 32, 2013, pages 52 - 62
NOLTE, M.; PEPINSKY, R.B.; VENYAMINOV, S.Y.; KOTELIANSKY, V.; GOTWALS, P.J.; KARPUSAS, M.: "Crystal structure of the α1β1 integrin -domain: insights into integrin -domain function", FEBS LETT, vol. 452, 1999, pages 379 - 385, XP004259786, DOI: doi:10.1016/S0014-5793(99)00666-3
NYMALM, Y.; PURANEN, J.S.; NYHOLM, T.K.; KAPYLA, J.; KIDRON, H.; PENTIKAINEN, O. T.; AIRENNE, T.T.; HEINO, J.; SLOTTE, J.P.; JOHNS: "Jararhagin-derived RKKH peptidesinduce structural changes in a11 domain of human integrin α1β1", J. BIOL. CHEM., vol. 279, 2004, pages 7962 - 7970, XP002411305, DOI: doi:10.1074/jbc.M312912200
OCHOLA, J.B.; LWANDE, W.; THIONG'O, T.; ROGO, L.; HERRMANN, R.; SCHEPERS, E.; BAGINE, R.; MUNGAI, P.; NDIEGE, I.O.: "Identification of insect-selective and mammal-selective toxins from Parabuthus leiosoma venom", TOXICON, vol. 50, 2007, pages 449 - 56, XP022234298, DOI: doi:10.1016/j.toxicon.2007.04.020
OLFA, K.Z.; JOSE, L.; SALMA, D.; AMINE, B.; NAJET, S.A.; NICOLAS, A.; MAXIME, L.; RAOUDHA, Z.; KAMEL, M.; JACQUES, M.: "Lebestatin, a disintegrin from Macrovipera venom, inhibits integrin-mediated cell adhesion, migration and angiogenesis", LAB. INVEST., vol. 85, 2005, pages 1507 - 1516
RENNER, C.; SACCA, B.; MORODER, L.: "Synthetic heterotrimeric collagen peptides as mimics of cell adhesion sites of the basement membrane", BIOPOLYMERS, vol. 76, 2004, pages 34 - 47
RICH, R.L.; DEIVANAYAGAM, C.C.; OWENS, R.T.; CARSON, M.; HOOK, A.; MOORE, D.; SYMERSKY, J.; YANG, V.W.; NARAYANA, S.V.; HOOK, M.: "Trench- shaped binding sites promote multiple classes of interactions between collagen and the adherence receptors, a1(31 integrin and Staphylococcus aureus cna MSCRAMM", J BIOL CHEM., vol. 274, 1999, pages 24906 - 24913, XP002317865, DOI: doi:10.1074/jbc.274.35.24906
SALMINEN, T.A.; NYMALM, Y.; KANKARE, J.; KAPYLA, J.; HEINO, J.; JOHNSON, M.S.: "Production, crystallization and preliminary X-ray analysis of the human integrin a11 domain", ACTA CRYSTALLOGR. D BIOL. CRYSTALLOGR., vol. 55, 1999, pages 1365 - 1367
SARRAY, S.; BERTHET, V.; CALVETE, J.J.; SECCHI, J.; MARVALDI, J.; EI-AYEB, M.; MARRAKCHI, N.; LUIS, J.: "Lebectin, a novel C-type lectin from Macrovipera lebetina venom, inhibits integrin-mediated adhesion, migration and invasion of human tumour cells", LAB. INVEST., vol. 84, 2004, pages 573 - 581
SELISTRE-DE-ARAUJO, H.S.; PONTES, C.L.; MONTENEGRO, C.F.; MARTIN, A.C.: "Snake venom disintegrins and cell migration", TOXINS (BASEL, vol. 2, 2010, pages 2606 - 21
SHEPPARDA, B.U.: "In vivo functions of integrins: lessons from null mutations in mice Dean", MATRIX BIOL., vol. 19, 2000, pages 203 - 209
SHI, M.; PEDCHENKO, V.; GREER, B.H.; VAN HORN, W.D.; SANTORO, S.A.; SANDERS, C.R.; HUDSON, B.G.; EICHMAN, B.F.; ZENT, R.; POZZI, A: "Enhancing integrin a1 inserted (I) domain affinity to ligand potentiates integrin a1131-mediated down-regulation of collagen synthesis", J BIOL CHEM., vol. 287, 2012, pages 35139 - 35152
SHIH, C.H.; CHIANG, T.B.; WANG, W.J.: "Inhibition of integrins av/a5-dependent functions in melanoma cells by an ECD-disintegrin acurhagin-C", MATRIX BIOL, vol. 32, 2013, pages 152 - 159
SIRET, C.; TERCIOLO, C.; DOBRIC, A.; HABIB, M.C.; GERMAIN, S.; BONNIER, R.; LOMBARDO, D.; RIGOT, V.; ANDRE, F.: "Interplay between cadherins and a2(31 integrin differentially regulates melanoma cell invasion", BR J CANCER, vol. 113, 2015, pages 1445 - 53
SMITH, J.W.; PIOTROWICZ, R.S.; MATHIS, D.: "A mechanism for divalent cation regulation of beta 3-integrins", J BIOL CHEM., vol. 269, 1994, pages 960 - 7
SPRINGER, T.A.; WANG, J-H.: "The three-dimensional structure of integrins and their ligands, and conformational regulation of cell adhesion", ADV PROTEIN CHEM., vol. 68, 2004, pages 29 - 63
TOMASELLI, K.J.; HALL, D.E.; FLIER, L.A.; GEHLSEN, K.R.; TURNER, D.C.; CARBONETTO, S.; REICHARDT, L.F.: "A neuronal cell line (PC12) expresses two beta 1-class integrins-alpha 1 beta 1 and alpha 3 beta 1-that recognize different neurite outgrowth-promoting domains in laminin", NEURON, vol. 5, 1990, pages 651 - 62
VAN DER FLIER, A.; SONNENBERG, A.: "Function and interactions of integrins", CELL TISSUE RESEARCH, vol. 3, 2001, pages 285 - 298
VITA, C.; ROUMESTAND, C.; TOMA, F.; MENEZ, A.: "Scorpion toxins as natural scaffolds for protein engineering", BIOCHEMISTRY, vol. 92, 1995, pages 6404 - 6408, XP055058737, DOI: doi:10.1073/pnas.92.14.6404
WANG, W.J.: "Acurhagin-C, an ECD disintegrin, inhibits integrin alphavbeta3-mediated human endothelial cell functions by inducing apoptosis via caspase-3 activation", BR J PHARMACOL, vol. 160, 2010, pages 1338 - 51
XIONG, J.P.; STEHLE, T.; DIEFENBACH, B.; ZHANG, R.; DUNKER, R.; SCOTT, D.L.; JOACHIMIAK, A.; GOODMAN, S.L.; ARNAOUT, M.A.: "Crystal structure of the extracellular segment of integrin aV(33", SCIENCE (NY, vol. 294, 2001, pages 339 - 345, XP002970379, DOI: doi:10.1126/science.1064535
XIONG, J.P.; STEHLE, T.; ZHANG, R.; JOACHIMIAK, A.; FRECH, M.; GOODMAN, S.L.; ARNAOUT, M.A.: "Crystal structure of the extracellular segment of integrin αYβ3 in complex with an Arg-Gly-Asp ligand", SCIENCE (NY, vol. 296, 2002, pages 151 - 155, XP002262471, DOI: doi:10.1126/science.1069040
XUE, L.C.; RODRIGUES, J.P.; KASTRITIS, P.L.; BONVIN, A.M.; VANGONE, A.: "PRODIGY: a web server for predicting the binding affinity of protein-protein complexes", BIOINFORMATICS, vol. 32, 2016, pages 3676 - 3678
ZENG, X.C.; CORZO, G.; HARIN, R.: "Scorpion venom peptides without disulfide bridges", IUBMB LIFE, vol. 57, 2005, pages 13 - 21

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113384682A (en) * 2021-05-31 2021-09-14 南方医科大学 Application of scorpion venom polypeptide Smp43 in preparation of antitumor drugs

Similar Documents

Publication Publication Date Title
US10023613B2 (en) Peptidomimetic macrocycles as modulators of MCL-1
US10253087B2 (en) CCR2 antagonist peptides
ES2330312T3 (en) GUANILATE CYCLASS RECEIVER AGONISTS FOR THE TREATMENT OF TISSULAR INFLAMMATION AND CARCINOGENESIS.
AU2019336230B2 (en) Compositions and methods for the treatment of viral infections
ES2349043T3 (en) NEUBLASTIN VARIANTS.
CN110709414A (en) GIP receptor activating peptide
KR20160010446A (en) Peptide compound
US20190185518A9 (en) Warhead-containing peptidomimetic macrocycles as modulators of bfl-1
EP3982994A1 (en) Compositions and methods for the treatment of respiratory syncytial virus
HUT72896A (en) Cyclic compounds useful as inhibitors of platelet glycoprotein iib/iiia
WO2019186344A1 (en) Anti-cancer and disintegrin scorpion venoms
JP2024063019A (en) GIP receptor agonist peptide compounds and uses thereof
CN109311940B (en) DSG 2-derived peptides
US20230310539A1 (en) Treatment of disease with epor antagonist
KR102017973B1 (en) Anti-Hepatitis B Virus X Protein Polypeptide Pharmaceuticals
WO2023280960A1 (en) Cancer therapeutics
WO2003080103A1 (en) Antagonists of megalin or cubilin for use in preventing organ damage induced by therapeutic agents
US20220041657A1 (en) Anti-erythropoietin receptor peptide
US7345018B2 (en) Method of treating side effects induced by therapeutic agents
JP5898071B2 (en) Dermaseptin B2 as an inhibitor of tumor growth
CN117957239A (en) Cancer therapeutic agent
WO2024003313A1 (en) New peptides as selective il-23 receptor antagonists

Legal Events

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

Ref document number: 19722946

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19722946

Country of ref document: EP

Kind code of ref document: A1