WO2007079460A2 - Conception d'analogues de chimiokines cxc pour le traitement de maladies humaines - Google Patents

Conception d'analogues de chimiokines cxc pour le traitement de maladies humaines Download PDF

Info

Publication number
WO2007079460A2
WO2007079460A2 PCT/US2007/000436 US2007000436W WO2007079460A2 WO 2007079460 A2 WO2007079460 A2 WO 2007079460A2 US 2007000436 W US2007000436 W US 2007000436W WO 2007079460 A2 WO2007079460 A2 WO 2007079460A2
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
lys
natural
tyr
analog
Prior art date
Application number
PCT/US2007/000436
Other languages
English (en)
Other versions
WO2007079460A3 (fr
Inventor
Ahmed Merzouk
Hassan Salari
Original Assignee
Chemokine Therapeutics Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US11/388,542 external-priority patent/US7368425B2/en
Priority claimed from US11/393,769 external-priority patent/US7994114B2/en
Priority claimed from US11/494,232 external-priority patent/US20070066523A1/en
Priority claimed from US11/590,210 external-priority patent/US20070116669A1/en
Priority claimed from PCT/CA2006/001848 external-priority patent/WO2007053952A1/fr
Application filed by Chemokine Therapeutics Corporation filed Critical Chemokine Therapeutics Corporation
Publication of WO2007079460A2 publication Critical patent/WO2007079460A2/fr
Publication of WO2007079460A3 publication Critical patent/WO2007079460A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/521Chemokines
    • C07K14/522Alpha-chemokines, e.g. NAP-2, ENA-78, GRO-alpha/MGSA/NAP-3, GRO-beta/MIP-2alpha, GRO-gamma/MIP-2beta, IP-10, GCP-2, MIG, PBSF, PF-4, KC
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to the preparation, design, derivation, and use of peptide agonists and antagonists of CXC chemokines.
  • Receptors are macromolecules involved in chemical signaling between and within cells; they may be located on the cell surface membrane or within the cytoplasm. Activated receptors directly or indirectly regulate cellular biochemical processes (e.g., ion conductance, protein phosphorylation, DNA transcription, etc.) Molecules that bind to a receptor are called ligands, and identification of molecules that can control receptor activity can lead to new and desirable drugs.
  • a ligand may activate or inactivate a receptor; activation may either increase or decrease a particular cell function, and each ligand may interact with multiple receptor subtypes. Few if any drugs are absolutely specific for one receptor or subtype, but most have relative selectivity. Selectivity is the degree to which a drug acts on a given site relative to other sites and relates largely to the physicochemical binding of the drug to cellular receptors.
  • Chemokines a family of small cytokines, or proteins secreted by cells, potential sources of drugs because they are ligands that bind to cellular receptors. Chemokines induce directed chemotaxis in nearby responsive cells, hence the name chemotactic cytokines. Some chemokines are considered pro-inflammatory and can be induced during an immune response while others are considered homeostatic. All chemokines have molecular masses of between 8 and 10 kDa and are approximately 20-50% identical in that they share about 20-50% gene sequence and amino acid sequence homology with each other and share common tertiary structures. Their receptors are all integral membrane proteins containing seven membrane-spanning helices which are coupled to G proteins. All chemokines possess a number of conserved cysteine residues involved in intramolecular disulfide bond formation.
  • chemokines have been recognized as chemotactic agents that recruit leukocytes to the sites of injuries and have been found to have a wide variety of potential therapeutic uses. Chemokines have been found to participate in increasing the hemocrit, mobilizing stem cells, or in assisting in vaccine production or otherwise stimulating the immune system to effectuate tumor destruction.
  • CXC chemokines CXCL9 and CXCL11 have been shown to be natural antagonists for the receptor CCR3 (Loetscher et al., J. Bio. Chem 276:2986-91, 2001); useful in improving asthma symptoms following intravenous injection (Zimmermann et al., J. Allaergy Clin.
  • Inflammatory chemokines are released from a wide variety of cells in response to bacterial infection, viruses, and agents that cause physical damage such as, for example, silica or the urate crystals that occur in gout. They function mainly as chemoattractants for leukocytes, recruiting monocytes, neutrophils and other effector cells from the blood to sites of infection or damage. Chemokines can be released by many different cell types and serve to guide cells involved in innate immunity and also the lymphocytes of the adaptive immune system. The cells that are attracted by chemokines tend to follow a signal of increasing chemokine concentration to the site of infection or tissue injury. Some chemokines also have roles in the development of lymphocytes, migration and angiogenesis (the growth of new blood vessels).
  • chemokines have four characteristic cysteines (Cys), and members of the chemokine family are categorized into four groups: (1 ) the CC chemokines ( ⁇ - chemokines) with two adjacent cysteines near the amino terminus of the protein, (2) the C chemokines (Y chemokines), (3) the CX3C chemokines ( ⁇ chemokines), and (4) the CXC chemokines ( ⁇ -chemokines) in which the cysteines are separated by an amino acid.
  • the four groups of chemokines act on different receptors, and each class has a characteristic function.
  • the ⁇ -chemokines are potent chemoattractants and activators of leukocytes such as neutrophils
  • the ⁇ - chemokines are also potent chemoattractants and activators of monocytes.
  • the ⁇ and ⁇ chemokines have a low sequence homology of about 30-35% and, as such, are distinctive in their functions - the ⁇ chemokines cannot activate monocytes and the ⁇ chemokines have no effect on neutrophils. Since two disulfide bonds are characteristically formed between the first and third cysteine and between the second and fourth cysteine, it has generally been assumed that the disulfide bridges among four cysteines were required. See Clark-Lewis et ai, J. Biol. Chem. 269:16075-16081 , (1994). However, exceptions have been reported. For example, lymphotactin has only two cysteine residues, allowing only one disulfide bond. Regardless, lymphotactin manages to retain a functional structure with only the single disulfide bond.
  • the CC chemokines ( ⁇ -chemokines), CCL1-CCL28, bind to CC chemokine receptors, of which ten have been discovered to date and are designated CCR1-CCR10. These receptors are expressed on the surface of different cell types allowing their specific attraction by the chemokines.
  • the CC chemokines such as RANTES, MIP-1 -alpha, MCP-1, generally function as chemoattractants for monocytes, basophils, eosinophils, and T-cells but not neutrophils.
  • the CC chemokines induce the migration of monocytes and other cell types such as NK cells and dendritic cells.
  • CC chemokine is monocyte chemoattractant protein-1 (MCP-1 ) which induces monocytes to leave the bloodstream and enter the surrounding tissue, becoming tissue macrophages.
  • CCL28 attracts T cells and B cells that express CCR10, and eosinophils that express CCR3. It has also been implicated in anti-microbial activity.
  • CCR5, or chemokine (C-C motif) receptor 5 binds RANTES/CCL5.
  • the C chemokines ( ⁇ -chemokines) lymphotactin- ⁇ (CL-1 ) and lymphotactin- ⁇ (CL-2) are thought to attract T cell precursors to the thymus.
  • the CX3C chemokine ( ⁇ -chemokine) fractalkine (CX 3 CLI) is both secreted and tethered to the surface of the cell that expresses it, thereby serving as both a chemoattractant and as an adhesion molecule.
  • the CXC chemokines have tremendous therapeutic potential as agonists and antagonists of cellular response and, thus, are the subject of the present application.
  • the CXC subfamily has been divided into two groups depending on the presence of the ELR motif (Glu-Leu-Arg) preceding the first cysteine: the ELR + -CXC chemokines and ELR-CXC chemokines (see, e.g., Clark- Lewis, supra, and Belperio etaL, "CXC Chemokines in Angiogenesis," J. Leukoc. Biol. 68:1-8, 2000).
  • ELR + -CXC chemokines also known as ELR-CXC chemokines because they contain the ELR motif
  • ELR ' -CXC chemokines also known as non-ELR- CXC chemokines because they do not contain the ELR motif
  • neutrophil chemoattractants but rather a chemoattractant for lymphocytes.
  • the ELR-CXC chemokines such as IL-8, are generally strong neutrophil chemoattractants while the non-ELR chemokines such as, for example, IP-10 and SDF-1 , predominantly recruit lymphocytes.
  • All ELR + CXC chemokines including growth-regulated oncogene (GRO- ⁇ , - ⁇ and -y /CXCL1-CXCL3), ENA-78 (CXCL5), granulocyte chemotactic protein (GCP-2/CXCL6), neutrophil-activating protein 2 (NAP-2/CXCL7) and IL-8, stimulate endothelial cell chemotaxis in vitro and angiogenesis in vivo.
  • GRO- ⁇ , - ⁇ and -y /CXCL1-CXCL3 ENA-78
  • GCP-2/CXCL6 granulocyte chemotactic protein
  • NAP-2/CXCL7 neutrophil-activating protein 2
  • IL-8 stimulate endo
  • a chemokine falls into does not provide complete predictability about the scope of the chemokine's activity.
  • one non- ELR-CXC chemokine actually that actually stimulates, rather than inhibits, angiogenesis is stromal-derived factor 1 (SDF-1/CXCL12).
  • SDF-1/CXCL12 stromal-derived factor 1
  • the SDF- 1 receptor CXCR4 is expressed on endothelial cells, which undergo chemotaxis in response to SDF-1.
  • Levels of mRNA for the SDF-1 receptor on human endothelial cells are upregulated in response to vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), which are non-chemokine angiogenic factors.
  • VEGF vascular endothelial growth factor
  • bFGF basic fibroblast growth factor
  • SDF- 1 has been shown to induce angiogenesis from cross-sections of leukocyte-free rat aorta in vitro, and the formation of capillary-like structures by endothelial cells in culture. Another difference between SDF-1 and the non-ELR-CXC chemokines that inhibit angiogenesis is that the non-ELR CXC chemokines are induced by IFN and SDF-1 is not.
  • the receptors for the CXC chemokines are G-protein coupled seven- transmembrane receptors. These receptors have been named CXCR1-CXCR7. The receptors are listed in Table 1, along with corresponding chemokine ligands.
  • the CXCL1 chemokine is also known as growth-related oncogene-alpha (GRO- ⁇ ), or melanoma growth stimulatory activity-alpha (MGSA- ⁇ ), or neutrophil activating protein-3 (NAP-3) was first identified in 1989 as a chemokine with the ability to specifically activate neutrophils. May play a role in inflammation and exerts its effects on endothelial cells in an autocrine fashion. In vitro, the processed forms GRO- ⁇ (4-73), GRO- ⁇ (5-73) and GRO- ⁇ (6-73) show a 30-fold higher chemotactic activity.
  • GRO- ⁇ growth-related oncogene-alpha
  • MGSA- ⁇ melanoma growth stimulatory activity-alpha
  • NAP-3 neutrophil activating protein-3
  • the CXCL2 chemokine is also known as growth-related oncogene-beta (GRO- ⁇ ), or melanoma growth stimulatory activity-alpha (MGSA- ⁇ ), or macrophage inflammatory protein 2-alpha (MIP2- ⁇ ) was first identified in 1991. Produced by activated monocytes and neutrophils and expressed at sites of inflammation. Hematoregulatory chemokine, which, in vitro, suppresses hematopoietic progenitor cell proliferation. GRO- ⁇ (5-73) shows a highly enhanced hematopoietic activity. GRO- ⁇ (5-73) is available under the name Garnocestim as immunomodulator. It is used prior to hematopoietic transplantation for peripheral blood stem cell mobilization and reduction of incidence, duration, and/or severity of chemotherapy induced cytopenias.
  • GRO- ⁇ growth-related oncogene-beta
  • MGSA- ⁇ melanoma growth stimulatory activity-alpha
  • MIP2- ⁇ macrophage
  • the CXCL3 chemokine is also known as growth-related oncogene-gamma (GRO-Y), or melanoma growth stimulatory activity-gamma (MGSA- ⁇ ), or macrophage inflammatory protein 2-beta (MIP2- ⁇ ) was first identified in 1991.
  • GRO-Y growth-related oncogene-gamma
  • MGSA- ⁇ melanoma growth stimulatory activity-gamma
  • MIP2- ⁇ macrophage inflammatory protein 2-beta
  • the CXCL4 chemokine is also known as plate! et-factor-4 (PF-4).
  • Platelet factor-4 is a 70-amino acid protein that is released from the alpha-granules of activated platelets and binds with high affinity to heparin. Its major physiologic role appears to be neutralization of heparin-like molecules on the endothelial surface of blood vessels, thereby inhibiting local antithrombin III activity and promoting coagulation.
  • PF4 probably has a role in inflammation and wound repair. See Eisman, R., et al. Blood 76: 336-344 (1990).
  • the CXCL5 chemokine is also known as epithelial-derived neutrophil- activating protein 78 (ENA-78), or neutrophil-activating peptide.
  • ENA-78 was first identified in 1995 and is known as a chemotactic for neutrophil granulocytes.
  • N- terminal processed forms ENA-78(8-78) and ENA-78(9-78) are produced by proteolytic cleavage after secretion from peripheral blood monocytes.
  • the CXC chemokine ENA-78/CXCL5 has profound angiogenic potential mediated through the CXCR2 receptor.
  • CXCL5 production contributes to both enhanced proliferation and invasion of squamous cell carcinomas and that targeting of specific pathways that include CXCL5 may represent a potential therapeutic modality for these lesions.
  • G-CSF granulocyte colony-stimulating factor
  • ENA-78 might promote the accumulation of neutrophils that had migrated from the intravascular space into inflammatory tissues.
  • CXCL5 expression in bone cells has implications for inflammatory bone diseases such as arthritis and periodontal disease.
  • ENA-78 has been shown to contribute to the angiogenic activity found in the inflamed RA joint.
  • the CXCL6 chemokine is also known as granulocyte chemotactic protein 2 (GCP-2), or chemokine alpha 3 (CKA-3) was first identified in 1993. It is a chemotactic factor for neutrophil granulocytes. GCP-2 binds the CXCR1 and CXCR2 receptors, along with IL-8, both of which are co-induced in microvascular endothelial cells after stimulation with pro-inflammatory stimuli. Moreover, GCP-2 is considered to be a proangiogenic and has been shown to have an adverse affect on the inflammatory process of asthma.
  • GCP-2 granulocyte chemotactic protein 2
  • CKA-3 chemokine alpha 3
  • the CXCL7 chemokine is also known as neutrophil-activati ⁇ g peptide 2 (NAP-2), connective tissue-activating peptide III (CTAP-III), or beta-thromboglobulin (Beta-TG) and was first identified in 1986.
  • NAP-2 timulates DNA synthesis, mitosis, glycolysis, intracellular cAMP accumulation, prostaglandin E2 secretion, and synthesis of hyaluronic acid and sulfated glycosaminoglycan. It also stimulates the formation and secretion of plasminogen activator by human synovial cells.
  • NAP-2 is a ligand for CXCR1 and CXCR2, weakly competing with IL-8; and, NAP-2, NAP- 2(73), NAP-2(74), NAP-2(1-66), and the more potent NAP-2(1-63) are chemoattractants and activators for neutrophils. NAP-2 also appears to play a role in atherosclerosis, having potential therapeutic applications in another widespread disease.
  • the CXCL8 chemokine is also known as interleukin-8 (IL-8) and has been shown to have many potential therapeutic applications. They have been shown to have both anti-tumor and anti-infective therapeutic activity. IL-8s have shown to be responsible for the recruitment and activation of leukocytes and a mediator of acute inflammatory response. They have an ability, for example, to stimulate T-cell chemotaxis. The IL-8s have shown a profound angiogenic potential that is mediated through the CXCR2 receptor and have demonstrated an ability to contribute to the angiogenic activity found in the inflamed RA joint.
  • IL-8 interleukin-8
  • the CXCL9 chemokine is also known as gamma interferon-induced monokine (MIG) was first identified in 1994. Cytokine that affects the growth, movement, or activation state of cells that participate in immune and inflammatory response. Chemotactive for activated T-cells. Binds to CXCR3. Induced by interferon gamma. The induction is enhanced by TNF-alpha in dermal fibroblasts and vein endothelial cells.
  • MIG interferon-induced monokine
  • the CXCL10 chemokine is also known as interferon-inducible protein-10 (IP-10).
  • Interferon-inducible protein-10 (IP-10 or CXCL10) is induced by interferon- gamma and TNF-alpha, and is produced by keratinocytes, endothelial cells, fibroblasts and monocytes.
  • tP-10 is thought to play a role in recruiting activated T cells to sites of tissue inflammation (Dufour, et al., "IFN-gamma-inducible protein 10 (IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking," J Immunol., 168:3195-204, 2002).
  • the CXCL11 chemokine is also known as interferon-inducible T-cell alpha chemoattractant (I-TAC), or interferon-gamma-inducible protein 9 (IP-9), or H174, or Beta-R1 was first identified in 2000. It is chemotactive for interleukin-activated T cells but not unstimulated T cells, neutrophils or monocytes; induces calcium release in activated T cells; binds to CXCR3; may play an important role in CNS diseases which involve T cell recruitment; and may play a role in skin immune responses.
  • I-TAC interferon-inducible T-cell alpha chemoattractant
  • IP-9 interferon-gamma-inducible protein 9
  • the CXCL13 chemokine is also known as B cell-attracting chemokine 1 (BCA-1), or B lymphocyte chemoattractant (BLC) 1 or ANGIE was first identified in 2000.
  • BCA-1 is a chemotactive for B lymphocytes but not for T-lymphocytes, monocytes and neutrophils; does not induce calcium release in B lymphocytes; binds to BLR1/CXCR5; and has its highest levels in the liver, spleen, lymph node, appendix, and stomach. Low levels of BCA-1 are found in salivary glands, mammary glands, and fetal spleen.
  • the CXCL14 chemokine is also known as chemokine BRAK was first identified in 2000.
  • BRAK/CXCL14 is expressed at the mRNA level in certain normal tissues, such as the heart, brain, placenta, lung, liver, skeletal muscle, kidney and pancreas; but it is absent from many established tumor cell lines and human cancers.
  • BRAK expression in normal and tumor specimens from patients with squamous cell carcinoma (SCC) of the tongue and used recombinant BRAK (rBRAK) showed abundant expression of BRAK protein in suprabasal layers of normal tongue mucosa but an absence of such expression in tongue SCC.
  • BRAK protein is also found to be expressed strongly by stromal cells adjacent to tumors and is recognized as a potent inhibitor of in vivo angiogenesis stimulated by multiple angiogenic factors, including interleukin 8, basic fibroblast growth factor, and vascular endothelial growth factor. As such, a loss of BRAK expression from tumors may facilitate neovascularization and possibly contribute to immunologic escape.
  • rBRAK has been shown to block endothelial cell chemotaxis at concentrations as low as 1 nmol/L, suggesting a strong potential therapeutic use of BRAK for angiogenesis inhibition.
  • the CXCL15 chemokine is a mouse CXC chemokine known as lungkine and was first identified in 2000.
  • Lungkine is chemotactic for neutrophils and appears to be specifically expressed in the lung. See Rossi, et al. J, Immunol. 162:5490- 5497 (1999).
  • Expression of lungkine in fetal lungs has been found to exist at low levels, and increased levels can be induced by inflammation in the lung.
  • lungkine may be involved in lung-specific neutrophil trafficking during normal and inflammatory conditions, having expression that is restricted to the lung, produced by bronchoepithelial cells, and released into the airways.
  • the CXCL16 chemokine is also known as a scavenger receptor for phosphatidylserine and oxidized low density lipoprotein (SR-PSOX) was first identified in 2003. Acts as a scavenger receptor on macrophages, which specifically binds to OxLDL (oxidized low density lipoprotein), suggesting that it may be involved in pathophysiology such as atherogenesis. SRPSOX induces a strong chemotactic response, calcium mobilization, and binds to CXCR6/Bonzo. SRPSOX is expressed in T-cell areas, the spleen, lymph nodes, lung, kidney, small intestine, and thymus. It is expressed weakly in the heart and liver, and there is no expression in the brain and bone marrow.
  • SR-PSOX oxidized low density lipoprotein
  • the CXCL17 chemokine is also known as DMC (dendritic cell and monocyte chemokine-like protein), which attracts dendritic cells and monocytes.
  • DMC is predicted to have an IL-8-like chemokine fold and to be structurally and functionally related to CXCL8 and CXCL14.
  • DMC induces migration of monocytes and immature dendritic cells, and expression studies show that DMCis constitutively expressed in the lung, suggesting a potential role for DMC in recruiting monocytes and dendritic cells from blood into lung parenchyma.
  • the inventions taught herein are generally directed to the design, preparation, derivation, and use of mimetics of CXC chemokines in the prevention, treatment, and ameliorization of diseases and disorders.
  • the CXC chemokine analogs bind to CXC chemokine receptors and can be designed to affect the activity of the receptor, either as an agonist or an antagonist.
  • the invention includes a composition comprising an analog of a native CXC chemokine selected from a group consisting of CXCL1 , CXCL2, CXCL3, CXCL5, CXCL6, CXCL7, CXCL9, CXCL11, CXCL13, CXCL14, CXCL15, CXCL16, and CXCL17, wherein the analog has a length ranging from about 20 to about 37 amino acids.
  • the analog comprises an N-terminal region comprising a first conserved sequence consisting of about 13 to 17 of the first 17 of the native CXC chemokine N-terminal residues, or conservatively modified variants thereof, or a sequence having at least 90% homology to the first conserved sequence and capable of binding to a cellular receptor that binds to the first conserved sequence.
  • the analog also comprises a C-terminal region comprising a second conserved sequence consisting of about 6 to 16 of the last 16 of the native CXC chemokine C-terminal residues; or conservatively modified variants thereof, or a sequence having at least 90% homology to the second conserved sequence and capable of binding to a cellular receptor that binds to the second conserved sequence.
  • the analog further comprises a linker selected from a group consisting of from 1 to 4 natural or non-natural amino acids having the following structure:
  • R L is selected from a group consisting of saturated and unsaturated aliphatics and heteroaliphatics consisting of 20 or fewer carbon atoms that are optionally substituted with (i) a hydroxyl, carboxyl, amino, amido, or imino group, or (ii) an aromatic group having from 5 to 7 members in the ring; and -(CH 2 J n -, wherein n is an integer ranging from 1 to 20.
  • the analog is optionally modified with a modifier selected from a group consisting of a poly(ethylene glycol) or derivative thereof, a glycosaminoglycan, a diagnostic label, a radioactive group, an acyl group, an acetyl group, a peptide, a modifier capable of reducing the ability of the analog to act as a substrate for aminopeptidases, and a modifier capable of reducing the ability of the analog to act as a substrate for carboxypeptidases.
  • a modifier selected from a group consisting of a poly(ethylene glycol) or derivative thereof, a glycosaminoglycan, a diagnostic label, a radioactive group, an acyl group, an acetyl group, a peptide, a modifier capable of reducing the ability of the analog to act as a substrate for aminopeptidases, and a modifier capable of reducing the ability of the analog to act as a substrate for carboxypeptidases.
  • the analog is a non-ELR-CXC chemokine analog; wherein, the first conserved sequence consists of about 13 to 17 of the first 17 of the native CXC chemokine N-terminal residues, or conservatively modified variants thereof, or a sequence having at least 90% homology to the first conserved sequence and capable of binding to a cellular receptor that binds to the first conserved sequence, wherein the first conserved sequence does not include an ELR motif.
  • the second conserved, sequence consists of about 6 to 16 of the last 16 of the native CXC chemokine C-terminal residues, or conservatively modified variants thereof, or a sequence having at least 90% homology to the second conserved sequence and capable of binding to a cellular receptor that binds to the second conserved sequence.
  • the analog is an ELR-CXC chemokine analog, wherein the first conserved sequence consists of about 13 to 17 of the first 17 of the native CXC chemokine N-terminal residues, or conservatively modified variants thereof, or a sequence having at least 90% homology to the first conserved sequence and capable of binding to a cellular receptor that binds to the first conserved sequence, wherein the first conserved sequence includes an ELR motif.
  • the second conserved sequence consisting of about 6 to 16 of the last 16 of the native CXC chemokine C-terminal residues, or conservatively modified variants thereof, or a sequence having at least 90% homology to the second conserved sequence and capable of binding to a cellular receptor that binds to the second conserved sequence.
  • the C-terminal region of the analog is cyclized.
  • the linker is 11-aminoundecanoic acid or a combination of 4 natural amino acids, wherein the linker optionally contains an amino acid having a side chain bearing positive charge.
  • the invention is directed to a method of increasing the activity of a cell having a CXC receptor comprising binding the CXC receptor to an analog taught herein, wherein the increase is relative to the activity of the cell in the absence of the analog.
  • the invention is directed to a method of decreasing the activity of a cell having a CXC receptor comprising binding the CXC receptor to an analog taught herein, wherein the increase is relative to the activity of the cell in the absence of the analog.
  • the invention is directed to an antibody produced using an analog taught herein as the antigen.
  • FIG. 1 illustates the induction of [Ca 2+ Ji mobilization by select IP-10 analogs at a concentration of 100 ⁇ M according to some embodiments.
  • FIGs. 2A and 2B shows the incubation of SUP-T1 cells with SDF-1 according to some embodiments.
  • FIG. 3 shows a competitive dose response for binding to the SDF-1 receptor by native SDF-1 and the CXCR4 agonists (competing ligands) against 125 I- SDF-1 according to some embodiments.
  • FIG. 4 shows the CXCR2 receptor binding of the IL-8 mimetics as competing ligands according to some embodiments.
  • FIG. 5 shows the response of circulating neutrophil counts to the administration of varying doses of the test mimetic following one hour of treatmentaccording to some embodiments.
  • FIG. 6 describes the kinetics of the rise in circulating neutrophil counts in response to the administration of the test mimetic according to some embodiments.
  • FIG. 7 shows the response of circulating haematopoietic progenitor/stem cells to the administration of varying doses of the test mimetic according to some embodiments.
  • FIG. 8 describes the kinetics of the rise in haematopoietic progenitor/stem cells in response to the administration of the test mimetic according to some embodiments.
  • FlGs. 9-11 illustrate the efficacy of the PF-4 analogs as agonists according to some embodiments.
  • the present invention generally relates to the design, preparation, derivation, and use of mimetics of CXC chemokines in the prevention, treatment, and ameliorization of diseases and disorders.
  • this invention is directed to the design, synthesis, and use of chemokine analogs which bind to CXC chemokine receptors, such that the analogs can be designed to affect the activity of the receptor, either as an agonist or an antagonist.
  • this invention is directed to the synthesis or use of CXC chemokine analogs which bind to receptors for any of the 17 CXC chemokines to modulate cellular activity.
  • modulates refers to altering the function or activity of a chemokine receptor by contacting it with a chemokine or chemokine analog and thus increasing or decreasing the probability that a complex forms between the receptor and a natural binding partner.
  • the chemokine analogs can be designed to increase or decrease the probability that such a complex forms between a chemokine receptor and a natural binding partner, for example, and the relative effect can, in some embodiments, depend on the concentration of the chemokine analog exposed to the receptor.
  • CXC chemokine receptor refers to a CXC chemokine receptor as the term is used by one skilled in the art, as well as any other chemical moiety, such as a peptide, capable of binding to a CXC chemokine analog.
  • natural binding partner refers to G proteins, polypeptides, lipids, small molecules, or nucleic acids that bind to CXC chemokine receptors in cells or in the extracellular environment.
  • natural binding partner includes a substrate to be acted upon by the CXC chemokine receptor. A change in the interaction between a CXC chemokine receptor and a natural binding partner can result in a decreased or increased activity of the CXC chemokine receptor.
  • activate can refer to an interaction between a CXC chemokine analog and a CXC chemoki ⁇ e receptor that increases the cellular or extracellular function of a CXC chemokine receptor.
  • the CXC chemokine receptor function can be the interaction with a natural binding partner and can result in a catalytic activity.
  • inhibitor refers to decreasing the cellular or extracellular activity of the CXC chemokine receptor.
  • complex can refer to an assembly of at least two molecules bound to one another.
  • a signal transduction complex often contains at least two protein molecules bound to one another.
  • a protein tyrosine receptor protein kinase, GRB2, SOS, RAF, and RAS assemble to form a signal transduction complex in response to a mitogenic ligand.
  • a CXC chemokine analog is bound to a CXC chemokine receptor.
  • a G protein bound to a CXC chemokine receptor.
  • the terms "contact,” “contacted,” and “contacting” can refer to combining a solution or a composition comprising the CXC chemokine or CXC chemokine analog with a liquid medium bathing the polypeptide or cells comprising a CXC chemokine receptor.
  • the solution comprising the CXC chemokine or CXC chemokine analog may also comprise another component, such as dimethyl sulfoxide (DMSO), which can facilitate the uptake of the CXC chemokine or CXC chemokine analog into the cells of interest.
  • DMSO dimethyl sulfoxide
  • the solution comprising the CXC chemokine or CXC chemokine analog may be added to the medium bathing the cells by utilizing a delivery apparatus, such as a pipette-based device or syringe-based device.
  • the invention is directed to the synthesis, design, derivation, or use of CXC chemokin analogs of one or more of the 17 CXC chemokines.
  • the native sequences of the 17 CXC chemokines are provided in the attached Sequence Listings as SEQ ID NOs:1-17:
  • the N-terminal region of CXC chemokines is involved in the binding and activating site of its receptor, as well as is the carboxy terminal region.
  • the beta sheet structure that connects the two termini appears to play a role in the stabilization of the CXCR and assuring that the termini are in the proper conformation.
  • the CXC chemokine analogs contain structures corresponding to various regions or portions of the native CXC chemokines, or conservatively modified variants thereof.
  • the CXC chemokine analogs comprise an N-terminal region and a C-terminal region joined together using a linker.
  • the amino acid residues of the CXC chemokine or chemokine analog can be cyclized, for example, by etherification of lysine and serine residues or by any other means described herein or known in the art.
  • the CXC chemokine analog comprises a sequence derived from the corresponding wild-type CXC chemokine but with one or more of the cysteines replaced with another amino acid, which can include any natural or non-natural amino acids.
  • Some embodiments consist of linking from about 3 to about 17 amino acids of the wild-type N-terminal region to about 3 to about 17 amino acids of the wild-type C-terminal region directly with a linker.
  • the N-terminal can be acetylated and/or the C-terminal can be amidated.
  • the regions selected from the N-terminal, internal and C-terminal regions may be 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 20, 25, 30, 35, 40, 41 , or 45 amino acids in length, and this length can be independent to the N-terminal region, C-terminal region, linker, or a combination thereof.
  • the CXC chemokine analogs range from about 12 to about 20, from about 20 to about 40, from about 20 to about 35, from about 21 to about 34, from about 21 to about 28, or any range therein, amino acids in length.
  • the analogs are a hybrid structure that includes a first region from one CXC chemokine and a second region from a different CXC chemokine, wherein the first and second regions are connected using a linker.
  • CXC Chemokine analogs of the invention are useful for treating or preventing inflammatory conditions, autoimmune disorders, cancer, graft rejection, bacterial infection, viral infection, vascular conditions (for example, atherosclerosis, restenosis, systemic lupus erythematosis, and ischemia-reperfusion), sepsis, tumorigenesis, and angiogenesis, gene therapy, stem cell mobilization, vaccine production, and blood cell recovery following chemotherapy.
  • Inflammatory conditions can include acute and/or chronic inflammatory diseases.
  • the CXC Chemokine analogs may assist in gene therapy, for example, by providing a means for arresting the cell cycle.
  • the CXC chemokine analogs can be used for treatments that include, but are not limited to, treatment or management of arthritis, asthma, colitis/illeitis, psoriasis, atherosclerosis and the like; treatment or management of autoimmune conditions that include, but are not limited to, rheumatoid arthritis, multiple sclerosis and other autoimmunqlogical diseases; treatment or management of cancers that include, but are not limited to, human malignancy/ cancer cell metastasis and relapses; treatment or management in assisting with blood cell recovery that includes, but is not limited to, blood cell elevation after chemotherapy/radiotherapy and stem cell mobilization for transplantations; vaccine production that includes, but is not limited to, enhancement in humoral antibody production, increases in antigen presenting T-cells, increases in dendritic cells and immunological features known as vaccine induction; treatment or management of osteoporosis; or treatment or management of genetic disease through gene therapy.
  • the therapeutic uses are effective because a CXC chemokine analog can be designed to act as an agonist or antagonist to a native CXC chemokine.
  • the agonistic activity of the CXC chemokine analogs may include mimicking the biological activity normally induced by a native CXC chemokine-
  • the antagonistic activity of the CXC chemokine analogs may include inhibiting the biological activity normally induced by a native CXC chemokine.
  • the analog does not have to be an analog of the native chemokine in order to serve as an agonist or antagonist of a particular cellular function - an analog of a first native CXC chemokine can act as an agonist or antagonist with respect to the cellular activity normally induced by a second native CXC chemokine.
  • the CXC chemokine analogs can be used to prepare vaccines, to enhance humoral antibody production, to increase antigen- presenting T-cells, to increase dendritic cells and immunological features known as vaccine induction, and combinations thereof.
  • the term "antibody” can refer to any antibody-like molecule that has an antigen binding region, and includes antibody fragments such as Fab 1 , Fab, F(ab')2, single domain antibodies (DABs), Fv, scFv (single chain Fv), and the like. Techniques for preparing and using various antibody- based constructs and fragments are well known in the art as are techniques for preparing and characterizing antibodies.
  • the CXC chemokine analogs taught herein can be used as antigens to produce antibodies using methods well-known to those skilled in the art.
  • the antibody can be polyclonal or monoclonal. In some embodiments, the antibody is humanized.
  • the products of the present invention can be referred to using various terms, including “analog,” “mimetic,” “peptide,” “polypeptide,” “chemokine analog,” “chemokine mimetic,” “chemokine derivative,” and the like. These terms, and others that would share the same meaning to one of skill, can be used interchangeably herein.
  • the CXC chemokine analogs can comprise a sequence selected from any sequences taught herein and may comprise additional elements such as R-group substituents and a linker selected from the possibilities set forth herein. However, the analogs taught herein are not necessarily limited to the sequences taught herein, as they are taught by way of example.
  • the term biological activity can refer to any physiological or biological response produced by a CXC chemokine or CXC chemokine analog, whether the response is manifested as a symptom in a subject, measurable using an in vivo laboratory test, traceable using a biomarker or the like, or measurable using an in vitro method.
  • the activity can refer to what is referred to in the scientific reports known in the art such as, for example, the activities referred to in Bruce, L. er a/., "Radiolabeled Chemokine binding assays," Methods in Molecular Biology (2000) vol. 138, pp129-134; Raphaele, B. et al.
  • a biological activity can include, but is not limited to, receptor binding, chemotaxis, calcium mobilization, cellular apoptosis, an increase or decrease in a symptom of a disease relative to the degree of the symptom present prior to administration of a chemokine analog, along with any other such ligand/receptor activities recognized by those skilled in the art as a physiological or biological response.
  • amino acids are identified in the present application by the following conventional three-letter abbreviations shown in Table 2.
  • the single letter identifier is provided for ease of reference.
  • the three-letter abbreviations are generally accepted in the peptide art, recommended by the IUPAC-IUB commission in biochemical nomenclature, and are required by WIPO Standard ST.25:
  • peptide sequences are described using the generally accepted convention of placing the N-terminus on the left and the C-terminus on the right of the sequence listing as required by WlPO Standard ST.25. Amino acid substitutions are indicated using brackets and superscript numbers to indicate the position of the residue substituted. Cyclized regions are indicated using underlined residues to show the cyclic portion, as well as by using the term "cyclo" or the term "cyclic" to show the cyclized portion.
  • the analog can include a first conserved region and a second conserved region, wherein the first conserved region can include an N-terminal region, and the second conserved region can include a C-terminal region.
  • the N-terminal region can include a series of up to 17 of the first 17 amino acids of a native CXC chemokine, and the C-terminal region can include a series of up to 17 of the last 17 amino acids in the native chemokine.
  • the analog can comprise an N-terminal region having the first 15 residues of the native chemokine, and the C-terminal region can comprise the last 13 residues of the chemokine.
  • the first and second conserved regions can be linked using a linker:
  • CXCL1 (GRO- ⁇ ) chemokine analogs include:
  • X01 is L-or D-AIa, GIy, L- or D-Phe, the preferred amino acid residues is L- or D- AIa;
  • X02 is any natural or non natural amino acid different from L- or D-Cys, such as L- or D-Ser, L- or D-Thr, or L- or D-Tyr;
  • Xo3 is L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-Ser, or L- or D-Thr;
  • Xo4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-AIa, GIy, or D-L-Phe;
  • Xo5 is L- or D-Thr, L- or D-Ser, L- or D-Tyr, L- or D-Trp, or L- or D-His;
  • Xo6 is any natural or non natural amino acid residue different from L- or D-GIx, L- or D-Asx, L- or D-Arg, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-Ser, L- or D-Thr, such as L- or D-GIu, or L- or D-Asp;
  • Xo7 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-IIe, L- or D-L- or D-VaI, or L- or D-AIa, L- or D-Ph e, or L- or D-Tyr;
  • Xo8 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Arg, L- or D-Lys, L- or D-Orn- or D-AIa;
  • Xo9 is L- or D-Cys, L- or D-AIa, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, or L- or D-Tyr;
  • Xio is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, GIy 1 L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, or L- or D-Trp;
  • Xii is L- or D-Cys, L- or D-AIa, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, or L- or D-Tyr;
  • Xi2 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-IIe, L- or D-L- or D-VaI, or L- or D-AIa, L- or D-Phe, or L- or D-Tyr;
  • X- I 3 is any natural or non natural amino acid residue different from L- or D-Cys . residue, such as L- or D-GIx, L- or D-Asx, GIy, L- or D-VaI, L- or D-Leu, L- or D-He, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, or L- or D-Trp;
  • Xi4 is L- or D-Thr, L- or D-Ser, L- or D-Tyr, L- or D-Trp, or L- or D-His;
  • Xi5 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-IIe, L- or D-L- or D-VaI, or L- or D-AIa, L- or D-Phe, or L- or D-Tyr;
  • X-16 is one or up to six natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, GIy, L- or D-VaI, L- or D- Leu, L- or D-IIe, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, L- or D-Trp, L- or D-Lys, and L- or D-GIn 1 L- or D-(GIn-GIy), L- or D-(GIn-GIy-IIe), L- or D- (GIn-GIy-IIe-HiS), L- or D-(Gln-Gly-lle-His-Pro), or L- or D-(Gln-Gly-lle-His-Pro- Lys); and wherein, is any natural or non natural amino acid residue different from L- or D-Cys residue,
  • Yn is L- or D-Asx, L- or D-GIx, L- or D-Ser, L- or D-Thr, L- or D-Tyr, L- or D-Lys, or L or D-Arg;
  • Yi 2 is any natural or non natural amino acid different from L- or D-Cys, such as L- or D-Ser, L- or D-Asx, L- or D-GIx, or L- or D-Lys;
  • Yi3 is L-or D-GIx, L- or D-Asx, L- or D-Lys, L- or D- Ser, L- or D-Thr, L- or D-Tyr, or L- or Arg; and,
  • Yi4 is one or up to three natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Ser, L- or D- Asn, L- or D-Om-, or D-AIa, and L- or D-(Lys-Ser), L- or D-( Lys-Ser-Asn).
  • CXCL2 (GRO- ⁇ ) chemokine analogs include:
  • Xoi is L- or D-Thr, L- or D-Ser, L- or D-Tyr, L- or D-Trp, L- or D-His, L- or D-GIx, L- or D-Asx, L- or D-Leu, L- or D-IIe, or L- or D-AIa;
  • Xo2 is any natural or non natural amino acid residue different from L- or D-GIx, L- or D-Asx, L- or D-Arg, L- or D-Lys, L- or D-His, L- or D-Arg, L- or D-Ser, or L- or D-Thr;
  • Xo3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-IIe, L- or D-L- or D-VaI, or L- or D-AIa, L- or D-Phe, or L- or D-Tyr;
  • Xo 4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Arg, L- or D-Lys, L- or D-Om-, or D-AIa;
  • Xo5 is L- or D-Cys, L- or D-AIa 1 L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, or L- or D-Tyr;, 00436
  • Xos is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, GIy, L- or D-VaI 1 L- or D-Leu, L- or D-IIe, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, or L- or D-Trp;
  • Xo7 is L- or D-Cys, L- or D-AIa, L- or D-Phe, L- or D-His t L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, or L- or D-Tyr;
  • Xo ⁇ is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-IIe, L- or D-L- or D-VaI, or L- or D-AIa, L- or D-Phe, or L- or D-Tyr;
  • Xo9 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, GIy, L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, or L- or D-Trp;
  • Xio is L- or D-Thr, L- or D-Ser, L- or D-Tyr, L- or D-Trp, or L- or D-His;
  • Xii is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-IIe, L- or D-L- or D-VaI, or L- or D-AIa, L- or D-Phe, or L- or D-Tyr;
  • Xi 2 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, GIy, L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, or L- or D-Trp;
  • Xi3 is GIy, L- or D-AIa, L- or D-VaI, L- or D-Leu, or L- or D-IIe;
  • X- I4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-He, L- or D-leu, L- or D-L- or D-VaI, or L- or D-AIa, L- or D-Phe, or L- or D-Tyr;
  • X- 1 5 is L- or D-His, L- or D-Trp, L- or D-Tyr, L- or D-Arg, L- or D-Lys, or L- or D-Phe;
  • Xi6 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-He, L- or D-L- or D-VaI, or L- or D-AIa, L- or D-Phe, or L- or D-Tyr;
  • Yoi is L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-Ser, L- or D-Thr, L- or D-Met, L- or D-Lys, L- or D-Arg, or L- or D-Om; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Om-, or D-VaI; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-IIe, or L- or D-Leu; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-IIe, L- or D-leu, L-
  • CXCL3 (GRO- ⁇ ) chemoki ⁇ e analogs include:
  • X0 1 is L- or D-Thr, L- or D-Ser, L- or D-Tyr, L- or D-Trp, L- or D-His, L- or D-GIx, L- or D-Asx, L- or D-Leu, L- or D-He 1 or L- or D-AIa;
  • X0 2 is any natural or non natural amino acid residue different from L- or D-GIx, L- or D-Asx, L- or D-Arg, L- or D-Lys, L- or D-His, L- or D-Arg, L- or D-Ser, L- or D-Thr;
  • Xo3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-IIe, L- or D-L- or D-VaI, or L- or D-AIa, L- or D-Phe, L- or D-Tyr;
  • Xo 4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Arg, L- or D-Lys, L- or D-Orn- or D-AIa;
  • Xo 5 is L- or D-Cys, L- or D-AIa 1 L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr;
  • X06 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, GIy, L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, L- or D-Trp;
  • X07 is L- or D-Cys, L- or D-AIa, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr;
  • X08 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-IIe, L- or D-L- or D-VaI, or L- or D-AIa, L- or D-Phe, L- or D-Tyr; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, GIy, L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, L- or D-Trp; is L- or D-Thr, L- or D-Ser, L- or D-Tyr, L- or D-Trp, L- or D-His; L- or D-Ser, L- or D-Tyr; is
  • Yo 6 is any natural or non natural amino acid residue different from L- or D-GIx, L- or D-Asx, L- or D-Arg, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-Ser, L- or D-Thr, L- or D-IIe; L- or D-GIu, L- or D-Lys;
  • Yo7 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-GIu- or D-IIe;
  • Yo8 is L- or D-Lys, L- or D-IIe 1 L- or D-Leu, L- or D-AIa, L- or D-Phe, L- or D-Tyr, L- or D-Ser, L-or D-Arg, L- or D-Om;
  • Yog is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-IIe, L- or D-L- or D-Asx, or L- or D-GIx, L- or D-Phe, L- or D-Tyr, L- or D-NIe;
  • Yio is any natural or non natural amino acid residue different from L- or D-Cys residu or D-Leu, L- or D-IIe, such as L- or D-Lys, L- or D-Arg, L- or D-Leu- or D-Asx, L-or D-GIx;
  • Yii is L- or D-Asx, L- or D-GIx, L- or D-Arg, L- or D-Lys, L- or D-AIa, L- or D-Om, GIy;
  • Yi2 is any natural or non natural amino acid residue different from L- or D-Cys residue or GIy, L- or D-Asx, L- or D-GIx, L- or D-Ser, L- or D-Tyr, L- or D-Thr;
  • Y-13 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as GIy, L- or D-Ser, L- or D-Thr;
  • Yi 4 is one or up to three natural or non natural amino acid different from L- or D- Cys, such as L- or D-Asx, L-or D-GIx, L- or D-Ser, L- or D-Thr, L- or D-Tyr, L- or D-Lys, and L- or D-Ser, L- or D-(Ser-Thr), L- or D-(Ser-Thr-Asn);
  • the CXCL4 (PF-4) chemokine analogs include:
  • Xoi is an optional natural or non natural amino acid residue different from L- or D- Cys residue, such as L- or D-GIx, L- or D-Asx;
  • Xo 2 is any natural or non natural amino acid residue different from L- or D-Cys, L- or D-AIa, L- or D-VaI, L- or D-IIe, L- or D-Leu;
  • Xo3 is L- or D-GIx, L- or D-Asx, L- or D-AIa, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-Ser, L- or D-Thr;
  • Xo 4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, L- or D 1 L- or D-AIa, L- or D-VaI, L- or D-Phe;
  • Xo5 is L- or D- Asx, L- or D-GIx, L- or D-AIa, L- or D-Phe, L- or D-Tyr;
  • Xo ⁇ is any natural or non-natural amino acid residue different from L- or D-Cys, such as L- or D-AIa, L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-Phe, L- or D-Tyr, L- or D-GIx;
  • Xo 7 is L- or D- Asx, L- or D-GIx, L- or D-AIa, L- or D-Phe, L- or D-Tyr;
  • Xo ⁇ is any natural or non-natural amino acid residue different from L- or D-Cys, such as L- or D-Leu, L- or D-AIa, L- or D 1 L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-Phe, L- or D-Tyr;
  • Xo9 is L- or D-GIx, L- or D-Asx, L- or D-Arg, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-Ser, L- or D-Thr;
  • X-io is L- or D-Cys, L- or D-AIa 1 L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr;
  • Xii is any natural or non-natural amino acid residue different from L- or D-Cys, such as L- or D-GIx, L- or D-Asx, L- or D-GIy, L- or D-VaI, L- or D-Leu, L- or D- He, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, L- or D-Trp;
  • Xi 2 is L- or D-Cys, L- or D-AIa, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr;
  • X 13 is L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-AIa, L- or D-Phe, L- or D-Tyr;
  • Xi4 is optional, and may be any natural or non-natural amino acid residue different from L- or D-Cys, such as L- or D-Lys, L- or D-Arg, L- or D-His;
  • Xis is optional and may be L- or D-Thr, L- or D-Ser, L- or D-Tyr, L- or D-AIa, L- or D-Phe;
  • Xi6 is optional and may be any natural or non-natural amino acid residue different from L- or D-Cys, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, L- or D-AIa, L- or D-Phe;
  • Xi7 is optional and may be any natural or non-natural amino acid residue different from L- or D-Cys, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, L- or D-AIa, L- or D-Phe;
  • Yoi is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-AIa, L- or D-VaI, L- or D-IIe, L- or D-Leu;
  • Yo2 is L- or D-Pro, L- or D-AIa;
  • Yo3 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-Leu, L- or D-AIa, L- or D 1 L- or D-VaI, L- or D-Leu, L- or D-IIe 1 L- or D-Phe, L- or D-Tyr, L- or D-Ser, L- or D-Thr, L- or D-Phe;
  • Y 04 is L- or D-Tyr, L- or D-Ser, L- or D-Thr, L- or D-Phe;
  • Yes is any. natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-Lys, L- or D-Arg, L- or D-His;
  • Yo6 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-Lys, L- or D-Arg, L- or D-His;
  • Yo7 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-GIx, L- or D-Asx, L- or D-GIy, L- or D-VaI, L- or D-Leu, L- or D- He, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, L- or D-Trp;
  • Yo8 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-GIx, L- or D-Asx, L- or D-GIy, L- or D-VaI, L- or D-Leu, L- or D- He, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, L- or D-Trp;
  • Yii is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-Leu, L- or D-AIa, L- or D,L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-Phe, L- or D-Tyr;
  • Yi2 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-Leu, L- or D-AIa, L- or D 1 L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-Phe, L- or D-Tyr;
  • Yi3 is L- or D- Asx, L- or D-GIx, L- or D-AIa;
  • Yi4 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, L- or D-AIa, L- or D-Phe.
  • CXCL5. ENA-78 Compounds [0082]
  • the CXCL5 (ENA-78) chemokine analogs include:
  • X01 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-IIe, L- or D-L- or D-Arg, or L- or D-Lys, L- or D-Orn, L- or D-AIa;
  • X02 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-Arg, L- or D-Lys, L- or D-Om- or L- or D-GIx, L- or D-Asx, L- or D-Thr, L- or D-Tyr;
  • Xo3 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-GIx, L- or D-Asx, L- or D-Leu, L- or D-IIe, L- or D-Arg, L- or D- Ser, L- or D-Thr;
  • Xo4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-Arg, or L- or D-IIe, L- or D-Lys;
  • Xo5 is L- or D- Arg, L- or D-Cys, L- or D-Lys, L- or D-Om, L- or D-AIa residue, such as L- or D-Arg, L- or D-Cys, L- or D-Orn- or D-AIa, L- or D-Lys;
  • X 06 is L- or D-Cys, L- or D-VaI 1 L- or D-Ph e, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Om;
  • Xo7 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI 1 L- or D-IIe, L- or D-AIa, L- or D-Phe, L- or D-Tyr;
  • X 08 is L- or D-Cys, L- or D-Leu r L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-IIe, L- or D-Orn;
  • Xo9 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-GIx, or L- or D-Asx, L- or D-Lys, L-IIe;
  • Xio is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, L- or D-Thr, L- or D-Ser, L- or D-Leu, L- or D-IIe, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, L- or D-Trp;
  • Xii is L- or D-Thr, L- or D-Ser, L- or D-Tyr, L- or D-Trp, L- or D-His;
  • Xi2 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-GIx, L- or D-L- or D-Asx, L- or D-Tyr;
  • Xi3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, GIy, L- or D-VaI, L- or D-Leu, L- or D-He, L- or D-Pro, L- or D-Phe, L- or D-Tyr, L- or D-His, L- or D-Trp;
  • Xi4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as GIy, L- or D-VaI, L- or D-AIa;
  • Xi5 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-His, L- or D-Om- or D-Arg;
  • Xi6 is one or up to two natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-His, L- or D-Pro, GIy, L- or D-VaI, L- or D- Leu, L- or D-IIe, L- or D-AIa, L- or D-Phe, L- or D-Tyr;
  • L- or D-Cys residue such as L- or D-Phe, L- or D-leu, L- or D-Lys, L- or D-Tyr, or L- or D- His, L- or D-Trp; is L- or D-Leu, L- or D-IIe, L- or D-Lys, L- or D-Phe, L- or D-Tyr, L- or D-Ser, L- or D-Thr, L- or D-Orn, L- or D-Arg, L- or D-VaI; is any natural or non natural amino acid residue different from L- or D-Cys residue, such asL- or D-Lys, L- or D-Arg, L- or D-Om- or D-VaI, L- or D-IIe 1 L- or D-Leu; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Phe, L- or D-leu
  • Yi3 is any natural or non natural amino acid different from L- or D-Cys, such asGly, L- or D-Asx, L- or D-GIx, L- or D-Lys, L- or D-Arg; L- or D-GIu; and,
  • Yi4 is one or up.to four natural or non natural amino acid residue different from L- or D-Cys residue, such asL- or D-Asx, L- or D-GIx, L- or D-Lys, L- or D-Om- or D-AIa, the preferred amino acid residues are: L- or D-Asn, L- or D-(Asn-Lys), L- or D-(Asn-Lys-Glu), L- or D-(Asn-Lys-Glu-Asn).
  • CXCL6 CXCL6. GCP-2.
  • CXCL6 (GCP-2) chemokine analogs include:
  • X01 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as GIy, L- or D-Pro, L- or D-L- or D-VaI, or L- or D-AIa;
  • X02 is any natural or non natural amino acid residue residue different from L- or D- Cys, such as L- or D-Pro, L- or D-AIa, L- or D-Leu- or L- or D-IIe, L- or D-Phe, L- or D-Thr, L- or D-Tyr;
  • Xo3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-IIe, L- or D-AIa, L- or D-Phe, L- or D-Tyr;
  • Xo4 is any natural or non natural amino acid different from L- or D-Cys, such as L- or D-Asx, L-or D-GIx, L- or D-Ser, L- or D-Th r, L- or D-Tyr, L- or D-Lys;
  • Xo5 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-AIa, GIy, or D-L-Phe;
  • Xo6 is L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Om;
  • Xo7 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-Arg, or L- or D-IIe, L- or D-Lys;
  • Xo8 is L- or D-Thr, L- or D-Ser, L- or D-Tyr, L- or D-Trp, L- or D-His;
  • Xo9 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-GIx 1 L- or D-Asx, L- or D-Leu, L- or D-IIe, L- or D-Arg, L- or D- Ser, L- or D-Thr;
  • Xio is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-Arg, or L- or D-IIe, L- or D-Lys;
  • Xii is L- or D-Arg, L- or D-Lys, L- or D-Orn- or L- or D-GIx, L- or D-Asx, L- or D- Thr, L- or D-Tyr;
  • Xi2 is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Om, L- or D-Trp, L- or D-His, L- or D-Phe;
  • X-I3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-GIx, L- or D-L- or D-Asx, L- or D-Tyr;
  • Xi4 is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Om, L- or D-Trp, L- or D-His, L- or D-Phe;
  • Xi5 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-Arg, or L- or D-He, L- or D-Lys;
  • X-I ⁇ is one up to ten natural or non natural amino acid residues different from L- or D-Cys residue, such as L- or D-Arg, L- or D-VaI, L- or D-Thr, L- or D-Leu, L- or D-IIe, L- or D-Asx, L- or D-Pro, L- or D-Lys, and L- or D-Leu, L- or D-(Leu-Arg- VaI), L- or D-(Leu-Arg-Val-Thr-Leu-Arg), L- or D-( Leu-Arg-Val-Thr-Leu-Arg- Val-Asn-Pro-Lys); and wherein, is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Phe, L- or D-leu, L- or D-Lys, L- or D-Tyr, or L- or D- His, L- or D-Trp; is L
  • Yii is L- or D-Asx, L- or D-GIx, L- or D-Ser, L- or D-Thr, L- or D-Tyr, L- or D-Lys, L or D-Arg, GIy,
  • Yi 2 is any natural or non natural amino acid different from L- or D-Cys, such as L- or D-Asx, L-or D-GIx, L- or D-Ser, L- or D-Thr, L- or D-Tyr, L- or D-Lys;
  • Yi3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as GIy 1 L- or D-Pro, L- or D-L- or D-VaI, or L- or D-AIa;
  • Yi4 is one or up to four natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Asx, L- or D-GIx, L- or D-Lys, L- or D-Om- or D-AIa, and L- or D-Asn, L- or D-(Asn-Lys), L- or D-(Asn-Lys-Lys), L- or D- (Asn-Lys-Lys-Asn );
  • CXCL7 CXCL7. NAP-2.
  • CXCL7 (NAP-2) chemokine analogs include:
  • Xoi is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-AIa 1 GIy, or D-L-Phe;
  • Xo 2 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-GIx, L- or D-Asx, L- or D-Leu, L- or D-IIe, L- or D-Arg, L- or D- Ser, L- or D-Thr,
  • Xo 3 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-GIx, L- or D-Asx, L- or D-Lys, L- or D-IIe, L- or D-Leu, L- or D- AIa; is L- or D-Arg, L- or D-Lys, L- or D-Om- or L- or D-GIx, L- or D-Asx, L- or D- Thr, L- or D-Tyr; is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Om, L- or D-Trp, L- or D-His
  • Yoi is one or up to five natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Pro, L- or D-Asx, L- or D-AIa, L- or D-Arg, or L- or D-Phe, L- or D-Tyr, and L- or D-(Pro-Asp-Ala-Pro-Arg), L- or D-Arg;
  • Y 02 is L- or D-Leu, L- or D-IIe, L- or D-Lys, L- or D-Phe, L- or D-Tyr, L- or D-Ser, L- or D-Thr, L- or D-Orn, L- or D-Arg, L- or D-VaI;
  • Yo3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Orn- or D-VaI, L- or D-IIe 1 L- or D-Leu;
  • Yo4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Orn- or D-VaI, L- or D-IIe, L- or D-Leu;
  • Yo5 is L- or D-Leu, L- or D-IIe 1 L- or D-Lys, L- or D-Phe, L- or D-Tyr, L- or D-Ser, L- or D-Thr, L- or D-Orn, L- or D-Arg, L- or D-VaI;
  • Yo6 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Orn- or D-AIa, IL- or D-He, L- or D-VaI;
  • Yo 7 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-GIx, L- or D-Asx, L- or D-Lys, L- or D-IIe, L- or D-Leu, L- or D- AIa;
  • Yo8 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Orn- or D-VaI, L- or D-IIe, L- or D-Leu;
  • Yog is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Orn- or D-VaI, L- or D-IIe, L- or D-Leu; Yio no residue, or is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-GIx, L- or D-Asx, L- or D-Lys, L- or D-IIe, L- or D- Leu, L- or D-AIa;
  • Yii no residue or is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-AIa, Gty, or D-L-Phe;
  • L- or D-Cys such as L- or D-GIx, L- or D-Asx, L- or D-Leu, L- or D-IIe, L- or D-Arg, L- or D-Ser, L- or D-Thr; L- or D-(Glu-Ser), L- or D-(GIu- Ser-Ala), L- or D-(GI u-Ser-Ala-Asp),
  • the invention includes mimetics of human chemokine interleukin-8 (IL-8).
  • the analog can include a first conserved region and a second conserved region, wherein the first conserved region can include an N-terminal region, and the second conserved region can include a C- terminal region.
  • the N-terminal region can include a series of up to 17 of the first 17 amino acids of a native IL-8 chemokine
  • the C-terminal region can include a series of up to 17 of the last 17 amino acids in the native IL-8 chemokine.
  • these conserved regions can be linked using a linker.
  • the analog can comprise an N- terminal region having the first 15 residues of the native IL-8 chemokine, and the C- terminal region can comprise residues 56-71 of the native IL-8 chemokine:
  • X01 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-GIx, L- or D-L- or D-Asx, L- or D-Tyr;
  • X02 is any natural or non natural amino acid residue residue different from L- or D- Cys, such as L- or D-Pro, L- or D-AIa, L- or D-Leu- or L- or D-IIe, L- or D-Phe, L- or D-Thr, L- or D-Tyr;
  • X03 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Orn- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Xo4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Orn- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • X05 is L- or D-Arg, L- or D-Lys, L- or D-Orn- or L- or D-GIx, L- or D-Asx, L- or D- Thr, L- or D-Tyr;
  • X06 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Om- or D-VaI, L- or D-IIe, L- or D-Leu;
  • Xo7 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as GIy, L- or D-Pro, L- or D-L- or D-VaI, or L- or D-AIa;
  • X08 is L- or D-Arg, L- or D-Lys, L- or D-Orn- or L- or D-GIx, L- or D-Asx, L- or D- Thr, L- or D-Tyr;
  • X 09 is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-Hts, L- or D-Tyr, L- or D-AIa, L- or D-Om, L- or D-Trp, L- or D-His, L- or D-Phe;
  • Xio is any natural or non natural amino acid different from L- or D-Cys, such as L- or D-Asx, L-or D-GIx, L- or D-Ser, L- or D-Thr, L- or D-Tyr, L- or D-Lys;
  • Xii is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Orn, L- or D-Trp, L- or D-His, L- or D-Phe;
  • Xi2 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-IIe, L- or D-GIx, L- or D-Asx, L- or D-Leu, or L- or D- AIa, L- or D-Phe, L- or D-Tyr, L- or D-GIu, L- or D-Lys;
  • X-13 is any natural or non natural amino acid different from L- or D-Cys, such as L- or D-Asx, L-or D-GIx 1 L- or D-Ser, L- or D-Thr, L- or D-Tyr, L- or D-Lys;
  • Xi4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-GIx, L- or D-L- or D-Asx, L- or D-Tyr;
  • Xi5 is L- or D-Asx, L- or D-GIx, L- or D-Arg, L- or D-Lys, L- or D-AIa, L- or D-Orn, GIy;
  • Xi6 is any natural or non natural amino acid residue different from L- or D-Cys, such as L- or D-GIx, L- or D-Asx, L- or D-Lys, L- or D-IIe, L- or D-Leu, L- or D- AIa;
  • Yoi is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, L- or D-AIa, L- or D-Arg, or L- or D- Phe, L- or D-Tyr;
  • Yo2 is L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-Phe, L- or D-Tyr, L- or D-Arg, L- or D-AIa, L- or D-His, L- or D-Trp, L- or D-VaI;
  • Yo3 is L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-Phe, L- or D-Tyr, L- or D-Arg, L- or D-AIa, L- or D-His, L- or D-Trp, L- or D-VaI;
  • Yo4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, L- or D-Lys, L- or D-Arg, or L- or D- Phe, L- or D-Tyr;
  • Yo5 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI 1 L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Orn- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Yo6 is L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-Arg, L- or D-Tyr, L- or D-Leu, L- or D-IIe, L- or D-NIe, L- or D-AIa, L- or D-VaI;
  • Yo7 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe, L- or D-VaI; L- or D-Lys,
  • Yo8 is L- or D-Leu, L- or D-Thr, L- or D-Lys, L- or D-Arg, L- or D-Tyr, L- or D-Ser, L- or D-IIe, L- or D-NIe, L- or D-AIa, L- or D-VaI;
  • Yog is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Om- or D-VaI, L- or D-IIe, L- or D-Leu;
  • Yio is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Yii is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, L- or D-Lys, L- or D-Arg, or L- or D- Ser, L- or D-Tyr, L- or D-Thr;
  • Y-I 2 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, L- or D-Trp, L- or D-His, or L- or D- Phe, L- or D-Tyr, L- or D-Thr, L- or D-Ser;
  • Yi3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-He, L- or D-VaI; L- or D-Arg,
  • Yi4 is one or up to seven natural or non natural amino acid residues different from L- or D-Cys, such as L- or D-GIx, L- or D-Asx, L- or D-Lys, L- or D-Ser, L- or D-Arg, L- or D-AIa, L- or D-Thr, L- or D-Tyr, and L- or D-Lys, L- or D-Ser, L- or D-(Lys-Gln), L- or D-(Ser-Arg), L- or D-(Ser-Arg-Gln), L- or D-(Ser-Arg-Gln- Lys), L- or D-(Ser-Arg-Gln-Lys-Lys), L-or D-(Ser-Arg-Gln-Lys-Lys-Thr), L- or D-(Ser-Arg-Gln-Lys-Thr-Thr); CXCL10. IP-10.
  • the invention includes mimetics of human chemokine IP-10.
  • the analog can include a first conserved region and a second conserved region, wherein the first conserved region can include an N-terminal region, and the second conserved region can include a C- terminal region.
  • the N-terminal region can include a series of up to 17 of the first 17 amino acids of a native IP-10 chemokine
  • the C-terminal region can include a series of up to 17 of the last 17 amino acids in the native IP-10 chemokine.
  • these conserved regions can be linked using a linker.
  • the analog can comprise an N- terminal region having the first 14 residues of the native IP-10 chemokine, and the C- terminal region can comprise residues 55-67, 58-71 , 59-72, or 66-78, for example, of the native IP-10 chemokine:
  • IP-10 analogs are taught in detail in U.S. Patent Application Nos. 11/590,210 and 10/243,795, each of which is hereby incorporated by reference herein in its entirety.
  • Possible configurations for the IP-10 mimetics can include those shown in Table 4.
  • the N-terminus can be acetylated, and the C- terminal can be amidated.
  • linker can be any linker taught herein.
  • the IP-IOs can be cyclized in their C-terminal region using the methods taught herein.
  • oi is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Phe, L- or D-His, L- or D-L- or D-Trp, L- or D-Tyr;
  • Xo2 is any natural or non natural amino acid residue residue different from L- or D- Cys, such as L- or D-Pro, L- or D-AIa, L- or D-Leu- or L- or D-IIe, L- or D-Phe, L- or D-Thr, L- or D-Tyr;
  • Xo3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Met, L- or D-NIe, L- or D-Leu, L- or D-GIx, L- or D- Asx, L- or D-Orn- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • o4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Phe, L- or D-His, L- or D-L- or D-Trp, L- or D-Tyr;
  • o5 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Orn- or D-VaI, L- or D-IIe, L- or D-Leu;
  • o6 is L- or D-Arg
  • Xii is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Om, L- or D-Trp, L- or D-His, L- or D-Phe;
  • Xi2 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-IIe 1 L- or D-GIx 1 L- or D-Asx, L- or D-Leu, or L- or D- AIa, L- or D-Phe, L- or D-Tyr, L- or D-GIu, L- or D-Lys;
  • Xi3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as GIy, L- or D-Pro, L- or D-L- or D-VaI, or L- or D-AIa;
  • Xi4 is any natural or non natural amino acid residue residue different from L- or D- Cys, such as L- or D-Pro, L- or D-AIa, L- or D-Leu- or L- or D-IIe, L- or D-Phe, L- or D-Thr, L- or D-Tyr;
  • Xi5 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as GIy 1 L- or D-Pro, L- or D-L- or D-VaI, or L- or D-AIa;
  • Xi6 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Yoi is one or four natural or non natural amino acid residue different from L- or D- Cys residue, such as L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-Phe, L- or D-Tyr, L- or D-Arg, L- or D-AIa, L- or D-GIx, L- or D-Asx, L- or D-VaI, and L-or D-Lys, L- or D-(Asn-Arg-Ala-Ser);
  • Yo 2 is L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-Phe, L- or D-Tyr, L- or D-Arg, L- or D-AIa, L- or D-His, L- or D-Trp, L- or D-VaI;
  • Y 03 is L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-Phe, L- or D-Tyr, L- or D-Arg, L- or D-AIa, L- or D-His, L- or D-Trp, L- or D-VaI; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, L- or D-Lys, L- or D-Arg, or L- or D- Phe, L- or D-Tyr; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI 1 L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Orn- or D-AIa, IL- or D-IIe, L- or D-
  • the invention includes mimetics of human chemokine SDF-1.
  • the analog can include a first conserved region and a second conserved region, wherein the first conserved region can include an N-terminal region, and the second conserved region can include a C- terminal region.
  • the N-terminal region can include a series of up to 17 of the first 17 amino acids of a native SDF-1 chemokine
  • the C-terminal region can include a series of up to 17 of the last 17 amino acids in the native SDF-1 chemokine.
  • these conserved regions can be linked using a linker.
  • the analog can comprise an N- terminal region having the first 14 residues of the native SDF-1 chemokine, and the C-terminal region can comprise residues 55-67 of the native SDF-1 chemokine:
  • the 1inker can be any linker caught herein.
  • the SDF-1 mimetics can be cyclized in their C- terminal region using the methods taught herein.
  • the CXCL13 (BCA-1 ) chemokine analogs include:
  • Xoi is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx 1 L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Xo2 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Leu, L- or D-IIe, L- or D-AIa, L- or D-NIe, L- or D-VaI;
  • Xo3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Orn- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Xo* is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Xo5 is L- or D-Tyr, L- or D-Phe, L- or D-His- or L- or D-GIx, L- or D-Asx, L- or D- Thr, L- or D-Trp, L- or D-Ser;
  • Xo ⁇ is L- or D-Tyr, L- or D-Phe, L- or D-His- or L- or D-GIx 1 L- or D-Asx, L- or D- Thr, L- or D-Trp, L- or D-Ser;
  • Xo7 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, or L- or D-AIa;
  • X 08 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, or L- or D-AIa;
  • Xo9 is any natural or non natural amino acid different from L- or D-Cys, such as L- or D-Leu, L-or D-IIe, L- or D-AIa 1 L- or D-Thr, L- or D-Tyr, L- or D-NIe, L- or D- Ser;
  • Xto is L- or D-Arg, L- or D-Lys, L- or D-Om- or L- or D-GIx, L- or D-Asx, L- or D- Thr, L- or D-Tyr;
  • Xii is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Om, L- or D-Trp, L- or D-His, L- or D-Phe;
  • Xi 2 is L- or D-Arg, L- or D-Lys, L- or D-Orn- or L- or D-GIx, L- or D-Asx, L- or D- Thr, L- or D-Tyr;
  • X 13 is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Orn, L- or D-Trp, L- or D-His, L- or D-Phe;
  • Xi4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Orn- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Xi5 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Orn- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Xi6 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Yoi is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe, L- or D-VaI; L- or D-GIn;
  • Yo2 is L- or D-VaI, L- or D-Thr, L- or D-Lys, L- or D-Phe, L- or D-Tyr, L- or D-Arg, L- or D-AIa, L- or D-Ser, L- or D-Trp, L- or D-Leu, L- or D-IIe;
  • Yo3 is any natural or non natural amino acid different from L- or D-Cys, such as L- or D-Leu, L-or D-He, L- or D-AIa, L- or D-Thr, L- or D-Tyr, L- or D-NIe, L- or D- Ser, L- or D-GIx, L- or D-Asx;
  • Yo4 is L- or D-Arg, L- or D-Lys, L- or D-Om- or L- or D-GIx, L- or D-Asx, L- or D- Thr, L- or D-Tyr, L- or D- Trp, L- or D-His; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Orn- or D-AIa, IL- or D-IIe, L- or D-VaI; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-Arg, L- or D-Tyr, L- or D-Leu, L- or D-II
  • CXCL14 CXCL14. BRAK.
  • CXCL14 (BRAK) chemokine analogs include:
  • X0 1 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, or L- or D-AIa;
  • X02 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Om- or D-VaI, L- or D-Ue, L- or D-Leu;
  • Xo3 is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Orn, L- or D-Trp, L- or D-His, L- or D-Phe;
  • X0 4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Om- or D-VaI, L- or D-IIe 1 L- or D-Leu;
  • X05 is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Om, L- or D-Trp, L- or D-His, L- or D-Phe;
  • Xo ⁇ is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, or L- or D-AIa;
  • X0 7 is L- or D-Arg, L- or D-Lys, L- or D-Orn- or L- or D-GIx, L- or D-Asx, L- or D- Th r, L- or D-Tyr;
  • X0 8 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Om- or D-VaI, L- or D-IIe, L- or D-Leu;
  • Xo9 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as GIy, L- or D-Pro, L- or D-L- or D-VaI, or L- or D-AIa;
  • X-io is any natural or non natural amino acid residues different from L- or D-Cys, such as L- or D-Pro, L- or D-VaI, L- or D-Phe, L- or D-GIx 1 L- or D-Asx, L- or D-Lys, L- or D-Arg, L- or D-IIe, L- or D-Ser;
  • Xii is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Arg, L- or D-Orn- or D-VaI, L- or D-IIe, L- or D-Leu;
  • Xi2 is any natural or non natural amino acid residue different from L- or D-Cys ' residue, such as L- or D-IIe, L- or D-GIx, L- or D-Asx, L- or D-Leu, or L- or D- AIa, L- or D-Phe, L- or D-Tyr, L- or D-GIu, L- or D-Lys;
  • Xi3 is L- or D-Arg, L- or D-Lys, L- or D-Om- or L- or D-GIx, L- or D-Asx, L- or D- Th r, L- or D-Tyr;
  • Xi4 is L- or D-Tyr, L- or D-Phe, L- or D-His- or L- or D-GIx, L- or D-Asx, L- or D- Thr, L- or D-Trp, L- or D-Ser;
  • Xi5 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, or L- or D-AIa;
  • Xi6 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Orn- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Yoi is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Lys, L- or D-Trp, L- or D-Arg, L- or D-Phe, L- or D- Tyr, L- or D-His, L- or D-AIa, L- or D-GIx, L- or D-Asx, L- or D-VaI;
  • Yo 2 is L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-Phe, L- or D-Tyr, L- or D-Arg, L- or D-AIa, L- or D-His, L- or D-Trp, L- or D-VaI, L- or D-Leu, L- or D-IIe;
  • Yo3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-GIx, L- or D-Asx, L- or D-Lys, L- or D-Arg, or L- or D- Phe, L- or D-Tyr; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx 1 L- or D-Asx, L- or D-Ser- or D-AIa, IL- or D-Thr, L- or D-Tyr; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, or L- or D-Trp; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L-
  • the CXCL15 (Lungkine) chemokine analogs include:
  • X01 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Orn- or D-AIa, IL- or D-IIe;
  • X02 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe;
  • Xo3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-IIe, L- or D-GIx, L- or D-Asx, L- or D-Leu, or L- or D- AIa, L- or D-Phe, L- or D-Tyr, L- or D-GIu, L- or D-Lys;
  • X04 is L- or D-Arg, L- or D-Lys, L- or D-Om- or L- or D-GIx, L- or D-Asx, L- or D- Thr, L- or D-Tyr;
  • Xo5 is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Orn, L- or D-Trp, L- or D-His, L- or D-Phe;
  • X06 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-IIe, L- or D-GIx, L- or D-Asx, L- or D-Leu, or L- or D- AIa, L- or D-Phe, L- or D-Tyr, L- or D-GIu, L- or D-Lys;
  • X07 is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Om, L- or D-Trp, L- or D-His, L- or D-Phe;
  • X08 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-IIe, L- or D-GIx, L- or D-Asx, L- or D-Leu, or L- or D- AIa, L- or D-Phe, L- or D-Tyr, L- or D-GIu, L- or D-Lys; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI 1 L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe; is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx 1 L- or D-A
  • Yo5 is L- or D-Ser, L- or D-Leu, L- or D-He- or L- or D-GIx, L- or D-Asx, L- or D-Thr, L- or D-Tyr, L- or Ala, L- or D-Phe, L- or D-His;
  • Yo7 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-His, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D- Orn- or D-Phe, L- or D-IIe, L- or D-Trp;
  • Yo8 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Phe, L- or D-Ser, L- or D-Thr, L- or D-Tyr;
  • Y-io is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Yii is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Leu, L- or D-IIe, L- or D-GIx, L- or D-Asx, L- or D-Trp- or D-AIa, IL- or D-Phe, L- or D-His;
  • Y-I2 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Ser, L- or D-Thr, L- or D-AIa, L- or D-Tyr, L- or D-Arg, L- or D-Lys;
  • Y-I3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-His, L- or D-Leu, L- or D-IIe, L- or D-GIx, L- or D-Asx, L- or D-Trp- or D-AIa, L- or D-Phe;
  • Yi4 is one or up to five natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-Ser, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe, GIy, and L- or D-Asn, L- or D-Thr, L- or D-(Thr-Gly), L- or D-(Thr-Gly-Ser), (Thr-Gly-Ser-Asp), L- or D-(Thr-Gly- Ser-Asp-Ala),
  • the CXCL16 (SRPSOX) chemokine analogs include:
  • Xoi is any natural or non natural amino acid residue different from L- or D-Cys residue, such as GIy, L- or D-Pro, L- or D-L- or D-VaI, or L- or D-AIa;
  • Xo2 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, or L- or D-AIa;
  • Xo3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-AIa, L- or D-Leu- or D-IIe;
  • Xo4 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, or L- or D-AIa;
  • Xo5 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as GIy, L- or D-Pro, L- or D-L- or D-VaI, or L- or D-AIa;
  • Xo6 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, or L- or D-AIa;
  • Xo7 is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Om, L- or D-Trp, L- or D-His, L- or D-Phe; o8 is any natural or non natural amino acid residue different from L- or D-Cys residue, such asL- or D-Tyr, L- or D-Phe, L- or D-His- or L- or D-GIx, L- or D- Asx, L- or D-Thr, L- or D-Trp, L- or D-Ser; o9 is L- or D-Cys, L- or D-VaI, L- or D-Phe, L-
  • Yo6 is L- or D-Met, L- or D-NIe, L- or D-His- or L- or D-GIx, L- or D-Asx, L- or D-Thr, L- or D-Trp, L- or D-Ser, L- or D-Tyr, L- or D-Lys, L- or D-Om;
  • Yo7 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-Thr, L- or D-Ser, L- or D-Tyr, or L- or D-AIa;
  • Yo8 is L- or D-Cys, L- or D-VaI, L- or D-Phe, L- or D-His, L- or D-Trp, L- or D-Ser, L- or D-Thr, L- or D-Lys, L- or D-His, L- or D-Tyr, L- or D-AIa, L- or D-Orn, L- or D-Trp, L- or D-His, L- or D-Phe, L-or D-NIe;
  • Yog is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-IIe, L- or D-GIx, L- or D-Asx, L- or D-Leu, or L- or D- AIa, L- or D-Phe, L- or D-Tyr, L- or D-GIu, L- or D-Lys; .
  • Yio is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe, L- or D-VaI;
  • Yii is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-IIe, L- or D-GIx, L- or D-Asx, L- or D-Leu, or L- or D- AIa, L- or D-Phe, L- or D-Tyr, L- or D-GIu, L- or D-Lys;
  • Yi3 is any natural or non natural amino acid residue different from L- or D-Cys residue, such as L- or D-VaI, L- or D-Lys, L- or D-Arg, L- or D-GIx, L- or D-Asx, L- or D-Om- or D-AIa, IL- or D-IIe, L- or D-VaI; and
  • Yi 4 is one or up to six natural or non natural amino acid residues, such as L- or D- Cys, GIy, L- or D-His, L- or D-AIa, L- or D-Tyr, L- or D-Ser, L- or D-Phe, IL- or D-NIe, L- or D-Tyr, and L- or D-Cys, L- or D-(Cys-Gly), L- or D-(Cys-Gly-His), L- or D-(Cys-Gly-His-Ala), L- or D-(Cys-Gly-His-Ala-Tyr), L- or D-(Cys-Gly-His- Ala-Tyr-Ser).
  • CXCL17 DMC. Compounds
  • the invention includes mimetics of human chemokine DMC.
  • the analog can include a first conserved region and a second conserved region, wherein the first conserved region can include an N-terminal region, and the second conserved region can include a C- terminal region.
  • the N-terminal region can include a series of up to 17 of the first 17 amino acids of a native DMC chemokine
  • the C-terminal region can include a series of up to 17 of the last 17 amino acids in the native DMC chemokine.
  • these conserved regions can be linked using a linker.
  • the analog can comprise an N- terminal region having the first 14 residues of the native DMC chemokine, and the C- terminal region can comprise residues 55-67 of the native DMC chemokine:
  • Possible configurations for the DMC mimetics can include those shown in Table 6.
  • the N-terminus can be acetylated, and the C-terminal can be amidated.
  • linker can be any linker taught herein.
  • the DMC mimetics can be cyclized in their C- terminal region using the methods taught herein. Synthesis
  • CXC chemokine analog compounds of the invention may be prepared by standard techniques known in the art.
  • a peptide or polypeptide component of a CXC chemokine analog may comprise, at least in part, a peptide synthesized using standard techniques (such as those described by Clark-Lewis, I., Dewald, B., Loetscher, M., Moser, B., and Baggiolini, M., (1994) J. Biol. Chem., 269, 16075- 16081 ).
  • Automated peptide synthesizers are commercially available (e.g., Advanced ChemTech Model 396; Milligen/Biosearch 9600, Appliedbiosystems/Pioneer).
  • Peptides and polypeptides may be assayed for CXC chemokine receptor agonist or antagonist activity in accordance with standard methods.
  • Peptides and polypeptides may be purified by HPLC and analyzed by mass spectrometry.
  • Peptides and polypeptides may be dimerized. In some embodimens, the peptides are dimerized via a disulfide bridge formed by gentle oxidation of the cysteines using 10% DMSO in water. Following HPLC purification, dimer formation may be verified, by mass spectrometry.
  • One or more modifying groups may be attached to a peptidic component by standard methods, for example, using methods for reaction through an amino group (e.g., the alpha-amino group at the amino-terminus of a peptide), a carboxyl group (e.g., at the carboxy terminus of a peptide), a hydroxy! group (e.g., on a tyrosine, serine or threonine residue) or other suitable reactive group on an amino acid side chain.
  • an amino group e.g., the alpha-amino group at the amino-terminus of a peptide
  • a carboxyl group e.g., at the carboxy terminus of a peptide
  • a hydroxy! group e.g., on a tyrosine, serine or threonine residue
  • analogs derived from the C-terminal and N- terminal joined by a linker could be cyclized in their C-terminal moiety using side- chain to side-chain; side-chain to scaffold or, scaffold to scaffold cyclization.
  • lactamization, etherification, or RCM Ring Closing Methatesis
  • the CXC chemokine analogs may be cyclized using a lactam formation procedure by joining the ⁇ -carboxy side chain or the ⁇ -carboxy moiety of glutamate (GIu) residue to the ⁇ -amino side chain of lysine (Lys) residue, as indicated in the following sequences by underlining of linked residues.
  • Lactams may for example be formed between glutamic acid and lysine (Lys) in the C-terminal portion of the polypeptide (which does not correspond necessarily with the numbering of that residue in the native sequence).
  • a lysine (Lys) may be substituted by ornithine (Orn) or any other (Lor D) natural or (L or D) non-natural amino acid having an amino group on its side chain.
  • glutamate (GIu) may for example be substituted with aspartate (Asp), denoted by nomenclature such as (Glu-> Asp) indicating a substitution in a given position in the peptide wherein aspartate replaces glutamate.
  • the CXC chemokine analogs include sequences wherein one or more of the amino acids have been replaced by a conservative amino acid substitution.
  • conservative amino acid substitution refers to a polypeptide chain in which one of the amino acid residues is replaced with an amino acid residue having a side chain with similar properties. Families of amino acid residues having side chains with similar properties are well known in the art.
  • amino acids with acidic side chains e.g., aspartic acid, glutamic acid
  • basic side chains e.g., lysine, arginine, histidine
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenyla!anine,methioni ⁇ e, tryptophan
  • beta- branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • CXC chemokines, CXC chemokine fragments, or CXC chemokine analogs may also be synthesized, in whole or in part, by recombinant methods using expression vectors encoding all or part of a CXC chemokine.
  • Vectors, or preferably expression vectors may contain a gene encoding a polypeptide of the invention, a functional derivative thereof, or another useful polypeptide. These vectors may be employed to express the encoded polypeptide in either prokaryotic or eukaryotic cells.
  • vector in this application refers to a DNA molecule into which another DNA of interest can be inserted by incorporation into the DNA of the vector.
  • classes of vectors can be plasmids, cosmids, viruses, and bacteriophage.
  • vectors are designed to accept a wide variety of inserted DNA molecules and then used to transfer or transmit the DNA of interest into a host cell (e.g., bacterium, yeast, higher eukaryotic cell).
  • a vector may be chosen based on the size of the DNA molecule to be inserted, as well as based on the intended use. For transcription into RNA or transcription followed by translation to produce an encoded polypeptide, an expression vector would be chosen.
  • a cloning vector For the preservation or identification of a specific DNA sequence (e.g., one DNA sequence in a cDNA library) or for producing a large number of copies of the specific DNA sequence, a cloning vector would be chosen. If the vector is a virus or bacteriophage, the term vector may include the viral/bacteriophage coat.
  • all or part of the vector DNA, including the insert DNA, may be incorporated into the host cell chromosome, or the vector may be maintained extrachromosomally.
  • those vectors that are maintained extrachromosomally are frequently capable of autonomous replication in a host cell into which they are introduced (e.g., many plasmids having a bacterial origin of replication).
  • Other vectors are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
  • expression vector refers to a DNA construct which allows one to place a gene encoding a gene product of interest, usually a protein, into a specific location in a vector from which the selected gene product can be expressed by the machinery of the host cell, or alternately, by in vitro expression system.
  • This type of vector is frequently a plasmid, but other forms of expression vectors, such as bacteriophage vectors and viral vectors (e.g., adenoviruses, replication defective retroviruses, and adeno-associated viruses), may be employed.
  • bacteriophage vectors and viral vectors e.g., adenoviruses, replication defective retroviruses, and adeno-associated viruses
  • Prokaryotic hosts are, in generally, very efficient and convenient for the production of recombinant polypeptides and are, therefore, one type of preferred expression system. Prokaryotes most frequently are represented by various strains of E. coli, but other microbial strains may be used, including other bacterial strains. Recognized prokaryotic hosts include bacteria such as E. coli, Bacillus, Streptomyces, Pseudomonas, Salmonella, Serratia, and the like. However, under such conditions, recombinantly-produced polypeptides will not be glycosylated.
  • plant cells may also be utilized as hosts, and control sequences compatible with plant cells are available, such as the cauliflower mosaic virus 35S and 19S promoters, and nopaline synthase promoter and polyadenylation signal sequences.
  • control sequences compatible with plant cells such as the cauliflower mosaic virus 35S and 19S promoters, and nopaline synthase promoter and polyadenylation signal sequences.
  • the protein of interest may be expressed in plants which have incorporated the expression vector into their germ line.
  • prokaryotic vectors that contain replication sites and control sequences derived from a species compatible with the host may be used.
  • Preferred prokaryotic vectors include plasmids such as those capable of replication in E. coli (such as, for example, pBR322, CoIEI, pSC101, pACYC 184, pVX, pUC118, pUC119 and the like).
  • Suitable phage or bacteriophage vectors may include ⁇ gt10, ⁇ gt11, vectors derived from filamentous bacteriophage such as m13, and the like.
  • Suitable Streptomyces plasmids include p1 J101, and streptomyces bacteriophages such as fC31.
  • Bacillus plasmids include pC194, pC221 , pT127, and the like.
  • Suitable Pseudomonas plasmids have been reviewed by Izaki (Jpn. J. Bacteriol. 33:729-742, 1978) and John et al. (Rev. Infect. Dis. 8:693-704, 1986).
  • a protein in a prokaryotic cell it is necessary to operably link the sequence encoding the protease of the invention to a functional prokaryotic promoter.
  • promoters are either constitutive or inducible promoters, but commonly inducible promoters are used.
  • constitutive promoters include the int promoter of bacteriophage ⁇ , the bla promoter of the ⁇ -lactamase gene sequence of pBR322, and the cat promoter of the chloramphenicol acetyl transferase gene sequence of pPR325, and the like.
  • inducible prokaryotic promoters include the major right and left promoters of bacteriophage ⁇ (PL and PR), the trp, recA, lacZ, lacl, and gal promoters of E. coli, the ⁇ -amylase and the V-28-specific promoters of B. subtilis, the promoters of the bacteriophages of Bacillus, and Streptomyces promoters.
  • Prokaryotic promoters are reviewed by Glick (Ind. Microbiot. 1 :277-282, 1987), Cenatiempo (Biochimie 68:505-516, 1986), and Gottesman (Ann. Rev. Genet. 18:415-442, 1984). Additionally, proper expression in a prokaryotic cell also requires the presence of a ribosome-binding site upstream of the encoding sequence.
  • Recombinant protein expression in E. coli can be increased by expressing the protein or fusion protein in a host bacteria with an impaired proteolytic system so as to reduce the post-synthesis degradation of the recombinant protein (Gottesman, S., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990) 119-128).
  • Another strategy is to alter the mix of codons used in the coding sequence to reflect the usage of the individual codons for each amino acid in the host (e.g., E. coli (Wada et al., (1992) Nucleic Acids Res. 20:2111- 2118)).
  • Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques and may prove useful for a variety of prokaryotic and eukaryotic expression systems.
  • Suitable hosts may also include eukaryotic cells.
  • Preferred eukaryotic hosts include, for example, yeast, fungi, insect cells, and mammalian cells both in vivo and in tissue culture.
  • Useful mammalian cell hosts include HeLa cells, cells of fibroblast origin such as VERO or CHO-K1 , and cells of lymphoid origin and their derivatives.
  • Preferred mammalian host cells include SP2/0 and J558L, as well as neuroblastoma cell lines such as IMR 332, which may provide better capacities for correct post-translational processing.
  • eukaryotic organisms such as yeast provide substantial advantages in that they can also carry out post- translational modifications.35:365-404, 1981).
  • yeast expression systems may be potentially utilized which incorporate promoter and termination elements from the actively expressed sequences coding for glycolytic enzymes. These expression systems produce large quantities of proteins when yeast are grown in mediums rich in glucose. Known glycolytic gene sequences can also provide very efficient transcriptional control signals.
  • a number of recombinant DNA strategies utilize strong promoter sequences and high copy number plasmids which can be utilized for production of the desired proteins in yeast. Examples of vectors suitable for expression in S. cerivisae include pYepSed (Baldari, et al., (1987) Embo J.
  • the protein of interest may be expressed in insect cells for example the Drosophila larvae.
  • insect cells Using insect cells as hosts, the Drosophila alcohol dehydrogenase promoter may be used (Rubin, Science 240:1453-1459, 1988).
  • baculovirus vectors can be engineered to express large amounts of the protein of interest in cultured insect cells (e.g., Sf 9 cells)(Jasny, Science 238:1653, 1987; Miller et al., in: Genetic Engineering, Vol. 8, Plenum, Setlow et al., eds., pp. 277-297, 1986).
  • Vectors which may be used include the pAc series (Smith etal. (1983) MoI. Cell Biol. 3:2156-2165) and the pVL series (Lucklow and Summers (1989) Virology 170:31-39).
  • promoters are derived from Simian Virus 40 (SV40), polyoma, Adenovirus 2, and cytomegalovirus (CMV) viruses.
  • Preferred eukaryotic promoters include, for example, the promoter of the mouse metallothionein I gene sequence (Hamer et a/., J. MoI. Appl. Gen. 1:273-288, 1982); the TK promoter of Herpes virus (McKnight, Cell 31 :355-365, 1982); the SV40 early promoter (Benoist et al., Nature (London) 290:304-31 , 1981); and the yeast gal4 gene sequence promoter (Johnston etal., Proc. Natl. Acad. Sci.
  • promoters from mammalian expression products such as actin, collagen, myosin, and the like, may be employed.
  • Regulatory elements may also be derived from adenovirus, bovine papilloma virus, cytomegalovirus, simian virus, or the like.
  • Transcriptional initiation regulatory signals may be selected which allow for repression or activation, so that expression of the gene sequences can be modulated.
  • regulatory signals which are temperature-sensitive so that by varying the temperature, expression can be repressed or initiated, or are subject to chemical (such as metabolite) regulation.
  • Expression of proteins of interest in eukaryotic hosts requires the use of eukaryotic regulatory regions. Such regions will, in general, include a promoter region sufficient to direct the initiation of RNA synthesis.
  • a recombinant mammalian expression vector may also be designed to be capable of directing expression of the nucleic acid preferentially in a particular cell type (i.e., tissue-specific regulatory elements are used to control the expression).
  • tissue-specific promoters include the liver-specific albumin promoter (Pinkert et al. (1987) Genes Dev. 1 :268-277); lymphoid-specific promoters (e.g., Calame and Eaton (1988) Adv. Immunol.43:235-275), and in particular promoters of immunoglobulins and T cell receptors (Winoto and Baltimore (1989) EMBO J. 8:729- 733, Banerji et al.
  • Preferred eukaryotic plasmids include, for example, SV40, BPV, pMAM- neo, pKRC, vaccinia, 2-micron circle, and the like, or their derivatives.
  • Such plasmids are well known in the art (Botstein et al., Miami W ⁇ tr. Symp. 19:265-274, 1982; Broach, In: "The Molecular Biology of the Yeast Saccharomyces: Life Cycle and Inheritance," Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, p. 445- 470, 1981 ; Broach, Cell 28:203-204, 1982; Bollon et al., J. Clin. Hematol. Oncol. 10:39-48, 1980; Maniatis, In: Cell Biology: A Comprehensive Treatise, Vol. 3, Gene Sequence Expression, Academic Press, NY, pp. 563-608, 1980).
  • the DNA construct(s) may be introduced into an appropriate host cell by any of a variety of suitable means, i.e., transformation, transfection, conjugation, protoplast fusion, electroporation, particle gun technology, DEAE-dextran-mediated transfection, lipofection, calcium phosphate-precipitation, direct microinjection, and the like. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (2001). After the introduction of the vector, recipient cells are grown in a selective medium, which selects for the growth of vector-containing cells. Expression of the cloned gene(s) results in the production of a protein of interest, or fragments thereof.
  • a gene that encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest.
  • selectable markers include those which confer resistance to drugs, such as G418, hygromycin, neomycin, methotrexate, glyphosate, and bialophos.
  • Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding the protein of interest or can be introduced on a separate vector. Cells stably transformed with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).
  • Proteins may be expressed as fusion proteins. Genes for proteins expressed as fusion proteins ligated into expression vectors that add a number of amino acids to a protein encoded and expressed, usually to the amino terminus of the recombinant protein. Such a strategy of producing fusion proteins is usually adopted for three purposes: (1) to assist in the purification by acting as a ligand in affinity purification, (2) to increase the solubility of the product, and (3) to increase the expression of the product. Often, expression vectors for use in fusion protein production, a proteolytic cleavage site is included at the junction of the fusion region and the protein of interest to enable purification of the recombinant protein away from the fusion region following affinity purification of the fusion protein.
  • Such enzymes, and their cognate recognition sequences include Factor Xa, thrombin and enterokinase, and may also include trypsin or chymotrypsin.
  • Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith, D. B. and Johnson, K. S. (1988) Gene 67:31-40), pMAL (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N.J.) which fuse glutathione S-transferase (GST) 1 maltose E binding protein, or protein A, respectively, to the target recombinant protein.
  • GST glutathione S-transferase
  • the CXC chemokine analogs can be modified in a variety of ways.
  • the R-group consists of a hydrogen or is an N-terminal modifier comprising a component selected from a group consisting of a poly(ethylene glycol) or derivative thereof, a glycosaminoglycan, a diagnostic label, a radioactive group, an acyl group, an acetyl group, a peptide, and a modifier capable of reducing the ability of the analog to act as a substrate for aminopeptidases.
  • the C-terminus of the analogs can be amidated.
  • a side chain to side chain cyclization can be produced between amino acid residues in the C-terminal region and can include lactamization, etherification, thioetherification, or cyclization generated by Mitsunubo or Ring Closing Methathesis (RCM) type of reactions.
  • RCM Ring Closing Methathesis
  • the C- terminal region included in these peptides can form a stable ⁇ -helix moity;
  • the linker can consist of up to four amino acids, -Xaai-Xaa2-Xaa 3 -Xaa4-, wherein Xaai, Xaa 2 , Xaa 3 , and Xaa4 are each independently selected from a group consisting of (a) any natural amino acid, and (b) any non-natural amino acid having the following structure:
  • R L is selected from a group consisting of saturated and unsaturated aliphatics and heteroaliphatics consisting of 20 or fewer carbon atoms that are optionally substituted with (i) a hydroxyl, carboxyl, amino, amido, or imino group; or (ii) an aromatic group having from 5 to 7 members in the ring.
  • the R L group can have from 0 to 10 carbon atoms and bear a positive charge.
  • the linker can comprise at least one amino acid having a side chain bearing positive charge.
  • the natural amino acid is not L- or D-Cys.
  • amino acids used in the present invention may be organic compounds comprising an amino group and a carboxyl group, and the amino group may be primary or secondary.
  • amino acids include, but are not limited to, glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tyrosine, aspartic acid, glutamic acid, lysine, arginine, serine, threonine, cysteine, asparagine, proline, tryptophan, histidine and combinations thereof.
  • R L may be a substituted, unsubstituted, hetero-, straight-chained, branched, cyclic, saturated or unsaturated aliphatic radical; or a substituted, unsubstituted, or hetero- aromatic radical.
  • R L can be substituted, unsubstituted, or hetero- forms of methyl, iso-propyl, sec-butyl, iso-butyl, benzyl, or a combination thereof.
  • substitutents include, but are not limited to, hydroxyl, carboxyl, amino, imino groups and combinations thereof.
  • R L is heteroaliphatic
  • heteroatoms include, but are not limited to, sulfur, phosphorous, oxygen, nitrogen and combinations thereof.
  • R L can comprise substituted or unsubstituted poly(alkylene glycols), which include, but are not limited to, PEG and PEG derivatives functionalized to link to specific chemical groups (available from Nektar Therapeutics, San Carlos, CA), poly(ethylene oxide), PPG, poly(tetramethylene glycol), ⁇ oly(ethylene oxide-co-propylene oxide), or copolymers and combinations thereof.
  • the amino acids may be bifunctional or trifunctional amino acids.
  • the amino acids may be limited to diamines, triamines, monocarboxylics, dicarboxylics, aliphatics, aromatics, amides, or a combination thereof.
  • the amino acids may not include any amino acid or group of amino acids, such as, for example lysine and its conservative substitutions. It is to be appreciated that one skilled in the art should recognize that some of the groups, subgroups, and individual amino acids may not be used in some embodiments of the present invention.
  • R L can be a substituted or unsubstituted alkylene comprising C n carbons in the alkylene backbone, wherein n is an integer ranging from 1 to about 20; from about 2 to about 16; from about 3 to about 12; from about 4 to about 10; from about 3 to about 8, and any range therein.
  • the linker can be, for example, 11-aminoundecanoic acid.
  • the linker can include any combination of natural or non-natural amino acids, wherein the number of amino acids can range from 1 to about 20; from about 2 to about 20; from about 3 to about 15, from about 4 to about 12, or any range therein. In some embodiments, the number of amino acids can range from 1 to 4. In some embodiments, the linker comprises any combination of four natural or non-natural amino acids such as, for example, -(GIy) 4 - (SEQ ID NO:212). In some embodiments, the linker is not -(GIy) 4 - (SEQ ID NO.212).
  • the linker consists of four amino acids, -XaarXaa 2 - Xaa3-Xaa ⁇ r (SEQ ID NO:213), wherein Xaai, Xaa2, Xaa3, and Xaa 4 are each independently selected from a group consisting of (a) any natural amino acid, and (b) any non-natural amino acid.
  • natural amino acids include, but are not limited to, glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, .
  • tyrosine aspartic acid, glutamic acid, lysine, arginine, serine, threonine, cysteine, asparagine, proline, tryptophan, histidine and combinations thereof.
  • the natural amino acid is not L- or D-Cys.
  • the linker comprises at least one amino acid having a side chain bearing positive charge.
  • amino acids include Lys, Arg, His, and Orn.
  • Xaai, Xaa 2 , Xaa 3 , and Xaa 4 can each be independently selected from a group consisting of GIy, L- or D-Lys, L- or D-Arg, L- or D-His, and L- or D-Orn.
  • the linker can contain any combination of GIy and Lys, GIy and Arg, GIy and Orn, or GIy and His.
  • the linker can contain all Lys, all Arg, all His, or all Orn.
  • the mimetics can include portions of the CXC chemokines connected directly to each other through amide bonds; or disulfide bonds, such as the disulfide bonds that can form between Cys residues.
  • amino acid substitutions may also be made in the polypeptide sequences. Examples of such substitutions include, but are not limited to, substituting lysine for glutamic acid, lysine for aspartic acid, ornithine for glutamic acid, and ornithine for aspartic acid.
  • any of the analogs taught herein can have 75, 80, 85, 90, 95, 97, 99%, or any range therein, homology to the corresponding regions of the native chemokine sequence, so long as the function of the respective analog is preserved. Percent homology can be determined using any method known to one of skill, such as the NCBI BLAST tool and techniques, for example, which is available at www.ncbi.nlm.nih.gov.
  • the CXC chemokine analogs may include at least one modifying group connected either directly or indirectly somewhere on the analog structure.
  • modifying group refers to any chemical moiety that was either absent from the corresponding native chemokine or comprises an isolated sequence of less than five amino acids. Such sequences are "isolated” in that they are positioned differently in the CXC chemokine analog than they were positioned in the native chemokine.
  • a linker can also comprise a modifying group.
  • the CXC chemokine analog modifications can include, but are not limited to, modifications of an N-terminus; modifications of a C-terminus; modifications of an internal region; modifications of an N-terminal region containing a sequence Glu-Leu-Arg; modifications of an internal region containing three anti-parallel ⁇ -sheets in the structure; modifications of a C- terminal region containing an ⁇ -helical structure; modifications of a combination of N- terminal and C-terminal regions; combinations of these modifications linked together either directly or through a linker; combinations of N-terminal and internal regions and modifications thereof; combinations of internal and C-terminal regions and modifications thereof; combinations of N-terminal, internal and C-terminal regions and modifications thereof; and combinations thereof.
  • the N- terminal region of each sequence must include a sub-sequence of Glu-Leu-Arg.
  • the N-terminal region does not include a sub-sequence Glu- Leu-Arg.
  • a modifying group can be connected, for example, to the N-terminus or C- terminus of a peptide; to a peptidic or peptidomimetic region flanking the core domain; to a side chain of at least one amino acid residue such as, for example, an ⁇ -amino group of a lysyl residue, a carboxyl group of an aspartic acid or glutamic acid residue, a hydroxy group of a tyrosyl, serine or threonine residue, or other suitable reactive group on an amino acid side chain; or in-chain as a linker.
  • Examples of chemical connections used to attach the modifying groups can include, but are not limited to, ether, azide, amide, ester, anhydride, orthoester, alkylamine, sulphide, disulphide, carbamate, carbonate, urea bonds, and the like.
  • a modifying group can include any of the functional groups described herein, such as a "biotinyl structure", which includes biotinyl groups and analogues and derivatives thereof.
  • biotinyl structures include, but are not limited to, iminiobiotinyl structures such as, for example, a 2-iminobiotinyl group.
  • the modifications can control the pharmacokinetic or pharmacodynamic properties of a CXC chemokine analog without substantially reducing its bioactive function.
  • the modifications can alter in vivo stability, bioavailability, or half-life of a mimetic.
  • the modifications can provide a diagnostic capability such as, for example, by creating a means of detecting the presence or location of a mimetic in vivo or in vitro. Examples of detectable substances are described below.
  • the functional groups themselves can serve as a modifying group.
  • the functional groups of the present invention can be independently selected from substituted, unsubstituted, hetero-, straight-chained, branched, cyclic, saturated or unsaturated aliphatic radical; or a substituted, unsubstituted, or hetero- aromatic radicals.
  • a functional group can be selected from H; aliphatic hydrocarbon groups such as, for example, alkyl, alkenyl, and alkynyl groups; aromatic groups such as, for example, aryl, aralkyl, aralkenyl, and aralkynyl groups; and, various other groups as defined below.
  • the functional groups may include biobeneficial, bioactive, and/or diagnostic agents.
  • a “bioactive agent” is a functional group that can be connected to the CXC chemokine analog to provide a therapeutic effect, a prophylactic effect, both a therapeutic and a prophylactic effect, or other biologically active effect.
  • a “biobeneficial agent” is a functional group that can also be connected to a CXC chemokine analog to provide a biological benefit within a subject.
  • a biobeneficial agent can be non-inflammatory, such as, for example, by acting as a biomimic to passively avoid attracting monocytes and neutrophils, which leads to the cascade of events creating inflammation.
  • a “diagnostic agent” is a type of bioactive agent that can be used, for example, in diagnosing the presence, nature, or extent of a disease or medical condition in a subject.
  • a diagnostic agent can be any agent that may be used in connection with methods for imaging an internal region of a patient and/or diagnosing the presence or absence of a disease in a patient.
  • Diagnostic agents include, for example, contrast agents for use in connection with ultrasound imaging, magnetic resonance imaging (MRI), nuclear magnetic resonance (NMR), computed tomography (CT), electron spin resonance (ESR), nuclear medical imaging, optical imaging, elastography, radiofrequency (RF) and microwave laser. Diagnostic agents may also include any other agents useful in facilitating diagnosis of a disease or other condition in a patient, whether or not imaging methodology is employed.
  • the biobeneficial agents can have a reactive group that can be used to connect an agent to a CXC chemokine analog.
  • reactive groups include, but are not limited to, hydroxyl, carboxyl, and amino groups.
  • the biobeneficial agents can remain attached to the CXC chemokine analog or be controllably released from the CXC chemokine analog.
  • the molecular weight of an agent connected to a CXC chemokine analog should be at or below about 40,000 Daltons, or any range therein, to ensure elimination of the agent from a subject.
  • the molecular weight of the agent ranges from about 300 Daltons to about 40,000 Daltons, from about 8,000 Daltons to about 30,000 Daltons, from about 10,000 Daltons to about 20,000 Daltons, or any range therein. It is to be appreciated that one skilled in the art should recognize that some of the groups, subgroups, and individual biobeneficial agents may not be used in some embodiments of the present invention.
  • the aliphatic radicals have from about 1 to about 50 carbon atoms, from about 2 to about 40 carbon atoms, from about 3 to about 30 carbon atoms, from about 4 to about 20 carbon atoms, from about 5 to about 15 carbon atoms, from about 6 to about 10 carbon atoms, and any range therein.
  • the aromatic radicals have from about 6 to about 180 carbon atoms, from about 12 to about 150 carbon atoms, from about 18 to about 120 carbon atoms, from about 24 to about 90 carbon atoms, from about 30 to about 60 carbon atoms, and any range therein.
  • alkyl can be used interchangeably with the term “alkylene” in some contexts and refers to a straight-chained or branched hydrocarbon chain.
  • alkyl groups include lower alkyl groups such as, for example, methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, t-butyl or iso-hexyl; upper alkyl groups such as for example, n-heptyl, n-octyl, iso-octyl, nonyl, decyl, and the like; lower alkylene such as, for example, ethylene, propylene, butylenes, butadiene, pentene, n-hexene and iso-hexene; and upper alkylene such as, for example, n-heptene, n- octene, iso-octene, non
  • alkyl groups may also contain various substituents in which one or more hydrogen atoms can be replaced by a functional group, or the alkyl groups can contain an in-chain functional group.
  • alkenyl refers to a straight-chained or branched hydrocarbon chain where at least one of the carbon-carbon linkages is a carbon-carbon double bond.
  • alkynyl refers to a straight-chained or branched hydrocarbon chain where at least one of the carbon-carbon linkages is a carbon-carbon triple bond.
  • aryl refers to a hydrocarbon ring bearing a system of conjugated double bonds often comprising at least six ⁇ (pi) electrons.
  • aromatic groups include, but are not limited to, phenyl, pyrrolyl, furyl, thiophenyl, imidazolyl, oxazole, thiazolyl, triazolyl, pyrazolyl, pyridyl, pyrazinyl, pyridazinyl and pyrimidinyl, naphthyl, anysyl, toluyl, xylenyl, and the like.
  • aralkyl refers to an alkyl group substituted with at least one aryl group.
  • aralkyls examples include substituted benzyls such as, for example, phenylmethyl, 2-naphthylethyl, 2-(2-pyridyl) propyl, 5-dibenzosuberyl, and the like.
  • aralkenyl refers to an alkenyl group substituted with at least one aryl group. Those aryl groups having heteroatoms in the ring structure may also be referred to as "aryl heterocycles" or "heteroaromatics.”
  • the aromatics can be substituted at one or more ring positions and can also be part of a polycyclic group.
  • aryl groups can include fused aromatic moieties such as naphthyl, anthracenyl, quinolyl, indolyl, and the like.
  • the phrase "straight-chained or branched” includes any substituted or unsubstituted acyclic carbon-containing compounds including, but not limited to, alkanes, alkenes and alkynes.
  • a radical is "straight-chained” when it has less than 0.1 mole percent of sidechains having 1 or more carbon atoms. In some embodiments, a radical is straight-chained if it has less than 0.01 mole percent of such sidechains.
  • a radical is straight-chained if it has less than 0.001 mole percent of such sidechains.
  • a radical is "branched" when it has more than 0.1 mole percent of sidechains having 1 or more carbon atoms. In some embodiments, a radical is branched when it has more than 0.01 mole percent of such sidechains. In some embodiments, a radical is branched when it has more than 0.001 mole percent of such sidechains.
  • a straight chain or branched alkyl has from about 1 to about 20 carbon atoms, from about 2 to about 18 carbon atoms, from about 3 to about 17 carbon atoms, from about 5 to about 15 carbon atoms, from about 2 to about 10 carbon atoms, or any range therein.
  • a cycloalkyl may have a ring structure containing from about 2 to about 12 carbon atoms, from about 3 to about 11 carbon atoms, from about 4 to about 10 carbon atoms, or any range therein.
  • a functional group may comprise a cyclic or polycyclic group.
  • cyclic group refers to a ring structure that can be substituted, unsubstituted, hetero-, saturated or unsaturated and have from 3 to 24 carbon atoms, from 3 to 18 carbon atoms, from 3 to 12 carbon atoms, or any range therein.
  • Examples of cyclic groups include, but are not limited to, cycloalkyls such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cyclooctyl structures; cycloalkenes; and aromatics.
  • polycyclic group refers to two or more substituted, unsubstituted, hetero-, saturated or unsaturated cyclic rings in which two or more ring carbons are common among two adjoining rings such that the rings are "fused rings.”
  • the rings can also be “bridged rings” in that they are joined through atoms that are not common among the adjoining rings.
  • heterocyclic group includes cyclic saturated, unsaturated and aromatic groups having from 3 to 10; from 4 to 8; or 5, 6, or 7 carbon atoms, wherein the ring structure includes one or more heteroatoms, such as oxygen, nitrogen, sulfur, or combinations thereof.
  • Heterocyclic groups include pyrrolidine, oxolane, thiolane, imidazole, oxazole, piperidine, piperazine, and morpholine.
  • the heterocyclic ring may be substituted at one or more positions with such substituents as, for example, halogens, alkyls, cycloalkyls, alkenyls, alkynyls, aryls, arylalkyls, other heterocycles, hydroxyl, amino, nitro, thiol, amines, imines, amides, phosphonates, phosphines, carbonyls, carboxyls, silyls, ethers, thioethers, sulfonyls, selenoethers, ketones, aldehydes, esters, -CF 3 , -CN.
  • Heterocycles may also be bridged or fused to other cyclic groups.
  • a linker may also link the heterocyclic group to such substituents as, for example, halogens, alkyls, cycloalkyls, alkenyls, alkynyls, aryls, arylalkyls, heterocyctes, hydroxyls, aminos, nitros, thiols amines, imines, amides, phosphonates, phosphines, carbonyls, carboxyls, silyls, ethers, thioethers, sulfonyls, sulfonates, selenoethers, ketones, aldehydes, esters, -CF3, -CN.
  • alkylcarbonyl refers to -C(O)-alkyl.
  • arylcarbonyl refers to -C(O)-aryl.
  • alkyloxycarbonyl refers to the group -C(O)-O-alkyl, and the term “aryloxycarbonyl” refers to - C(O)-O-aryl.
  • acyloxy refers to -0-C(O)-R 7 , in which R 7 is alkyl, alkenyl, alkynyl, aryl, aralkyl or heterocydyl.
  • amino refers to -N(Ra)(Rp), in which R 0 and Rp are each independently hydrogen, alkyl, alkyenyl, alkynyl, aralkyl, aryl, or in which R 0 and Rp together with the nitrogen atom to which they are attached form a ring having 4-8 atoms.
  • amino includes unsubstituted, monosubstituted (e.g., monoalkylamino or monoarylamino), and disubstituted (e.g., dialkylamino or alkylarylamino) amino groups.
  • acylamino refers to -N(R' ⁇ )C(O)-R7, in which R 7 is as defined above and R 0 . is alkyl.
  • nitro means -NO2
  • halogen designates -F, -Cl, -Br or -I
  • sulfhydryl means -SH
  • hydroxyl means -OH.
  • the functional groups can include, but are not limited to, oxygen-containing groups such as, for example, alcohols, ethers, phenols, and derivatives thereof.
  • oxygen-containing groups include, but are not limited to, acetonides, alcohols, alkoxides, bisphenols, carbinols, cresols, diols, enols, enolates, epoxides, ethers, glycols, hydroperoxides, peroxides, phenols, phenolates, phenoxides, pinacols, trioxides, and ynols.
  • the functional groups can include, but are not limited to, oxygen-containing groups such as, for example, aldehydes, ketones, quinones and derivatives thereof.
  • oxygen-containing groups include, but are not limited to, acetals, acyloins, aldehydes, carbonyl compounds, diosphenols, dypnones, hemiacetals, hemiketals, ketals, ketenes, keto compounds, ketones, quinhydrones, quinomethanes, quinines, and combinations thereof.
  • the functional groups can include, but are not limited to, oxygen-containing groups such as, for example, carboxylic acids and derivatives thereof.
  • oxygen-containing groups include, but are not limited to, carboxylic acids, oxoacids, sulfonic acids, acid anhydrides, acid thioanhydrides, acyl groups, acyl halides, acylals, anhydrides, carboxylic acids, cyclic acid anhydrides, cyclic anhydrides, esters, fulgides, lactides, lactols, lactones, macrolides, naphthenic acids, ortho acids, ortho esters, oxo carboxylic acids, peroxy acids, and combinations thereof,
  • the functional groups can include, but are not limited to, nitrogen-containing groups containing one nitrogen such as, for example, aldimines, aldoximes, alkoxyamines, amic acids, amides, amines, amine oxides, amine ylides, carbamates, hemiaminals, carbonitriles, carboxamides, isocyanides, cyanates, isocyanates, diisocyanates, cyanides, cyanohydrins, diacylamines, enamines, fulminates, hemiaminals, hydroxamic acids, hydroximic acids, hydroxylamines, imides, imidic acids, imidines, imines, oximes, isoureas, ketenimines, ketimines, ketoximes, lactams, lactims, nitriles, nitro, nitroso, nitrosolic acids, oxime O-ethers, quaternary ammonium compounds, quinone
  • the functional groups can include, but are not limited to, sulfur-containing groups such as thio, thiol, thioether, sulfonyl, sulfido, sulfinamides, sulfilimines, sulfimines, sulfimides, sulfinamidines, sulfines, sulfinic acids, sulfinic anhydrides, sulf ⁇ nylamines, sulfonamides, suifones, sulfonediimines, sulfonic acids, sulfonic anhydrides, sulfoxides, sulfoximides;
  • sulfur-containing groups such as thio, thiol, thioether, sulfonyl, sulfido, sulfinamides, sulfilimines, sulfimines, sulfimides, sulfinamidines, sulfines,
  • the functional groups can include, but are not limited to, silyl groups, halogens, selenoethers, trifluoromethyls, thio-derivatives of urethanes where at least one oxygen atom is replaced by a sulfur atom; phosphoryls, phosphonates, phosphinates; and ethyleneically unsaturated groups such as, for example, allyl, acryloyl and methacrylol, and maleate and maleimido; and combinations thereof.
  • heteroatoms of the hetero- radicals include, but are not limited to, sulfur, phosphorous, oxygen, nitrogen and combinations thereof.
  • heterocyclic groups include, but are not limited to, pyrrolidine, oxolane, thiolane, imidazole, oxazole, piperidine, piperazine, and morpholine.
  • the heterocyclics may also be bridged or fused to other cyclic groups as described below.
  • the modifying groups can include, but are not limited to, O-modified derivatives including, but not limited to, C-terminal hydroxymethyl benzyl ether, and other C-terminal hydroxymethyl derivatives; N- modified derivatives including, but not limited to, substituted amides such as alkylamides; hydrazides and compounds in which a C-terminal phenylalanine residue is replaced with a phenethylamide analogue such as, for example, by replacing Ser-lle-Phe with Ser-lle-phenethyiamide.
  • the functional group may include a fluorescein- containing group.
  • fluorescein-containing groups include, but are not limited to, 5-(and 6-)-carboxyfluorescein succinimidyl ester and fluorescein isothiocyanate.
  • the modifying group may include a cholyl structure.
  • An example of a cholyl derivative is 3-(0-aminoethyl-iso)-cholyl (Aic).
  • the functional group may include N- acetylneuraminyl, trans-4-cotininecarboxyl, 2-imino-1-imidazolidineacetyl, (S)-(-)- indoline-2-carboxyl, 2-norbornaneacetyl, ⁇ -oxo-5-acenaphthenebutyryl, (-)-2-oxo-4- thiazolidinecarboxyl group, tetrahydro-3-furoyl group, 4-morpholinecarbonyl group, 2- thiopheneacetyl group, 2-thiophenesulfonyl group, diethylene-triaminepentaacetyl group, (O)-methoxyacetyl group, N-acetylneuraminyl group, and combinations thereof.
  • the functional group may include light scattering groups, magnetic groups, nanogold, other proteins, a solid matrix, radiolabels, carbohydrates, and combinations thereof.
  • biobeneficial agents include, but are not limited to, many of the polymers listed above such as, for example, carboxymethyl cellulose, poly(alkylene glycols), poly(N-vinyl pyrrolidone), poly(acrylamide methyl propane sulfonic acid), poly(styrene sulfonate), sulfonated dextran, polyphosphazenes, poly(orthoesters), ⁇ oly(tyrosine carbonate), dermatan sulfate, hyaluronic acid, heparin, and any derivatives, analogs, homologues, congeners, salts, copolymers and combinations thereof.
  • polymers listed above such as, for example, carboxymethyl cellulose, poly(alkylene glycols), poly(N-vinyl pyrrolidone), poly(acrylamide methyl propane sulfonic acid), poly(styrene sulfonate), sulfonated dextran, polyphosphazene
  • heparin derivatives include, but are not limited to, earth metal salts of heparin such as, for example, sodium heparin, potassium heparin, lithium heparin, calcium heparin, magnesium heparin, and low molecular weight heparin.
  • Other examples of heparin derivatives include, but are not limited to, heparin sulfate, heparinoids, heparin-based compounds and heparin derivatized with hydrophobic materials.
  • hyaluronic acid derivates include, but are not limited to, sulfated hyaluronic acid such as, for example, O-sulphated or N-sulphated derivatives; esters of hyaluronic acid wherein the esters can be aliphatic, aromatic, arylaliphatic, cycloaliphatic, heterocyclic or a combination thereof; crosslinked esters of hyaluronic acid wherein the crosslinks can be formed with hydroxyl groups of a polysaccharide chain; crosslinked esters of hyaluronic acid wherein the crosslinks can be formed with polyalcohols that are aliphatic, aromatic, arylaliphatic, cycloaliphatic, heterocyclic, or a combination thereof; hemiesters of succinic acid or heavy metal salts thereof; quaternary ammonium salts of hyaluronic acid or derivatives such as, for example, the O-sulphated or N-sulphated derivatives; esters of
  • poly(alkylene glycols) and its derivatives include, but are not limited to, PEG, mPEG, poly(ethylene oxide), poly(propylene glycol)(PPG), poly(tetramethylene glycol), and any derivatives, analogs, homologues, congeners, salts, copolymers and combinations thereof.
  • the poly(alkylene glycol) is poly(ethylene glycol-co-hydroxybutyrate).
  • copolymers that may be used as biobeneficial agents include, but are not limited to, any derivatives, analogs, homologues, congeners, salts, copolymers and combinations of the foregoing examples of agents.
  • copolymers that may be used as biobeneficial agents in the present invention include, but are not limited to, dermatan sulfate, which is a copolymer of D-glucuronic acid or L-idur ⁇ nic acid and N-acetyl-D-galactosamine; poly(ethylene oxide-co-propylene oxide); copolymers of PEG and hyaluronic acid; copolymers of PEG and heparin; copolymers of PEG and hirudin; graft copolymers of poly(L-lysine) and PEG; copolymers of PEG and a poly(hydroxyalkanoate) such as, for example, poly(ethylene glycol-co-hydroxybutyrate); and, any derivatives, analogs, con
  • the bioactive agents can be any moiety capable of contributing to a therapeutic effect, a prophylactic effect, both a therapeutic and prophylactic effect, or other biologically active effect in a subject.
  • a bioactive agent can also have diagnostic properties.
  • the bioactive agents include, but are not limited to, small molecules, nucleotides, oligonucleotides, polynucleotides, amino acids, oligopeptides, polypeptides, and proteins.
  • Bioactive agents include, but are not limited to, antiproliferatives, antineoplastics, antimitotics, antiinflammatories, antiplatelets, anticoagulants, antifibrins, antithrombins, antibiotics, antiallergics, antioxidants, and any prodrugs, codrugs, metabolites, analogs, homologues, congeners, derivatives, salts and combinations thereof. It is to be appreciated that one skilled in the art should recognize that some of the groups, subgroups, and individual bioactive agents may not be used in some embodiments of the present invention.
  • Antiproliferatives include, for example, actinomycin D, actinomycin IV, actinomycin 11, actinomycin X1, actinomycin C1, and dactinomycin (Cosmegen®, Merck & Co., Inc.).
  • Antiplatelets, anticoagulants, antifibri ⁇ , and antithrombins include, for example, sodium heparin, low molecular weight heparins, heparinoids, hirudin, argatroban, forskolin, vapiprost, prostacyclin and prostacyclin analogues, dextran, D-phe-pro-arg-chloromethylketone (synthetic antithrombin), dipyridamole, glycoprotein llb/llla platelet membrane receptor antagonist antibody, recombinant hirudin, and thrombin inhibitors (Angiomax®, Biogen, Inc.), and any prodrugs, codrugs, metabolites, analogs, homologues, congeners, derivatives, salts and combinations thereof.
  • Cytostatic or antiproliferative agents include, for example, angiopeptin, angiotensin converting enzyme inhibitors such as captopril (Capoten® and Capozide®, Bristol-Myers Squibb Co.), cilazapril or lisinopril (Prinivil® and Prinzide®, Merck & Co., Inc.),' calcium channel blockers such as nifedipine; colchicines; fibroblast growth factor (FGF) antagonists, fish oil (omega 3-fatty acid); histamine antagonists; lovastatin (Mevacor®, Merck & Co., Inc.); monoclonal antibodies including, but not limited to, antibodies specific for Platelet-Derived Growth Factor (PDGF) receptors; hitroprusside; phosphodiesterase inhibitors; prostaglandin inhibitors; suramin; serotonin blockers; steroids; thioprotease inhibitors; PDGF antagonists including, but not limited to, triazo
  • bioactive agents useful in the present invention include, but are not limited to, free radical scavengers; nitric oxide donors; rapamycin; everolimus; tacrolimus; 40-O-(2-hydroxy)ethyl-rapamycin; 40-O-(3-hydroxy)propyl-rapamycin; 40-O-[2-(2-hydroxy)ethoxy]ethyl-rapamycin; tetrazole containing rapamycin analogs such as those described in U.S. Pat. No.
  • 6,329,386 estradiol; clobetasol; idoxifen; tazarotene; alpha-interferon; host cells such as epithelial cells; genetically engineered epithelial cells; dexamethasone; cytokines; chemokines, chemokine mimetics, chemokine receptor ligands, and, any prodrugs, codrugs, metabolites, analogs, homologues, congeners, derivatives, salts and combinations thereof.
  • Free radical scavengers include, but are not limited to, 2,2',6,6'- tetramethyl-1-piperinyloxy, free radical (TEMPO); 4-amino-2,2',6,6'-tetramethyl-1- piperinyloxy, free radical (4-amino-TEMPO); 4-hydroxy-2,2',6,6'-tetramethyl- piperidene-1-oxy, free radical (TEMPOL), 2,2',3,4,5,5'-hexamethyl-3-imidazoliniu ⁇ n- 1-yloxy methyl sulfate, free radical; 16-doxyl -stearic acid, free radical; superoxide dismutase mimic (SODm) and any analogs, homologues, congeners, derivatives, salts and combinations thereof.
  • TEMPO free radical
  • 4-amino-TEMPO 4-hydroxy-2,2',6,6'-tetramethyl- piperidene-1-oxy, free radical
  • SODm superoxide dis
  • Nitric oxide donors include, but are not limited to, S- nitrosothiols, nitrites, N-oxo-N-nitrosamines, substrates of nitric oxide synthase, diazenium diolates such as spermine dtazenium diolate and any analogs, homologues, congeners, derivatives, salts and combinations thereof.
  • Chemokines include, but are not limited to, IL-8, IP-10, PF-4, MIP-1 ⁇ , RANTES, I-309, MCP-1, CCL28, and SDF-1.
  • Chemokine mimetics include, but are not limited to, those taught in U.S. Patent Application Publication Nos. 2002/0156034, 2002/0165123, and 2003/0148940; and U.S. Patent Application No. 10/243,795; each of which is incorporated by reference herein in its entirety.
  • Chemokine receptor ligands include, but are not limited to, those taught in U.S. Patent Nos. 6,515,001 and 6,693,134; and U.S. Patent Application Publication Nos. 2003/0004136, 2003/0045550, 2003/0092674, and 2003/0125380; each of which is incorporated by reference herein in its entirety.
  • the paramagnetic agents include, but are not limited to, gadolinium chelated compounds.
  • fluorescent agents include, but are not limited to, indocyanine green, umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin.
  • agents detectable by ultrasound include, but are not limited to, perflexane, Albunex ® and Optison ® .
  • agents used in PET include, but are not limited to, fluorodeoxyglucose, sodium fluoride, methionine, choline, deoxyglucose, butanol, raclopride, spiperone, bromospiperone, carfentanil, and flumaze ⁇ il.
  • detectable substances include, but are not limited to, various enzymes and prosthetic groups.
  • suitable enzymes include, but are not limited to, horseradish peroxidase, alkaline phosphatase, ⁇ -galactosidase, or acetylcholinesterase.
  • suitable prosthetic group complexes include, but are not limited to, streptavidin/biotin and avidin/biotin.
  • Labeled CXC chemokine analogs can be used to assess in vivo pharmacokinetics, as well as detect the progression of a disease or the propensity of a subject to develop a disease.
  • chemokine receptors for tissue distribution can be detected using a labeled CXC chemokine analog either in vivo or in an in vitro sample derived from a subject.
  • a CXC chemokine analog may be radioactively labeled with 14 C, either by incorporation of 14 C into the modifying group or one or more amino acid structures in the CXC chemokine analog.
  • a modifying group can be chosen to provide a chelation site for a diagnostic label.
  • the modifying group can be the Aic derivative of cholic acid, which provides a free amino group; a tyrosine residue within a CXC chemokine sequence may be substituted with radioactive iodotyrosyl; or a CXC chemokine analog may be labeled with radioactive technetium or iodine.
  • any isotope of radioactive iodine may be incorporated to create a diagnostic agent.
  • 123 I has a half-life of 13.2 hours and can be used for whole body scintigraphy; 124 I has a half life of 4 days and can be used for PET; 125 I has a half life of 60 days and can be used for metabolic turnover studies; and, 131 I has a half life of 8 days and can be used for whole body counting and delayed low resolution imaging studies.
  • a modification may be introduced at the C-terminus of a peptide, the N-terminus of a peptide, in the region between the C-terminus and N-terminus, or a combination thereof.
  • a modification to the C-terminus may reduce the ability of a CXC chemokine analog to act as a substrate for carboxypeptidases.
  • C-terminal modifiers include, but are not limited to, an amide group, an ethylamide group and various non-natural amino acids such as, for example, D-amino acids and ⁇ -alanine.
  • a modification of a C-terminus may be accompanied by a modification to the N- termi ⁇ us to reduce the ability of a CXC chemokine analog to act as a substrate for aminopeptidases.
  • N-terminus modifiers include, but are not limited to acyl, alkyl, aryl, arylalkyl, hydroxyalkyl, alkanoyl groups, alkanoics, diacids, and other modifiers having a carboxyl functional group.
  • the modification to an N-terminus can be deamidation.
  • aminopeptidases and carboxypeptidases have been found to have important functions in biological activities such as, for example, diabetes, memory and learning, antigen formation, and angiogenesis.
  • aminopeptidase refers to a multifunctional enzyme that cleaves proteins from the N-terminus.
  • Aminopeptidases can be classified into a number families such as, for example, the zinc-containing (M1) aminopeptidase family which consists of nine aminopeptidases that include, but are not limited to, placental leucine aminopeptidase (P-LAP), adipocyte-derived leucine aminopeptidase (A-LAP) and leukocyte-derived arginine aminopeptidase (L-RAP).
  • P-LAP placental leucine aminopeptidase
  • A-LAP adipocyte-derived leucine aminopeptidase
  • L-RAP leukocyte-derived arginine aminopeptidase
  • Modulation of aminopeptidase activity can have many therapeutic and prophylactic applications.
  • control of the activity of P-LAP can control the inducement of uterine contractions and treat or prevent disorders such as premature delivery and spontaneous abortion, as well as other disorders associated with water resorption, memory and learning and glucose metabolism.
  • control of the activity of A-LAP can treat disorders associated with antigen production, blood pressure and inflammation.
  • control of the activity of L-RAP can treat disorders association with antigen formation.
  • carboxypeptidases Although both aminopeptidases and carboxypeptidases can terminate biological activity, the carboxypeptidases clearly predominate in such terminations.
  • the term "carboxypeptidase” refers to a multifunctional enzyme that cleaves proteins from the C-terminus. Carboxypeptidases are derived from the zymogens, procarboxypeptidase A and B. Modulation of carboxypeptidase activity can have many therapeutic and prophylactic applications. In one example, control of the activity of the carboxypeptidases such as kininase Il (angiotensin-converting enzyme), carboxypeptidase M, and carboxypeptidase N, can potentially control hypertensive disorders relating to cardiovascular and kidney disorders.
  • kininase Il angiotensin-converting enzyme
  • carboxypeptidase M can potentially control hypertensive disorders relating to cardiovascular and kidney disorders.
  • compositions include a CXC chemokine analog compound to be used in treating diseases or disorders selected from the group consisting of autoimmune diseases, acute chronic inflammation, cancer, cardiovascular disease, infectious disease, and inflammatory disorders including rheumatoid arthritis, chronic inflammatory bowel disease, chronic inflammatory pelvic disease, multiple sclerosis, asthma, osteoarthritis, atherosclerosis, psoriasis, rhinitis, autoimmunity, and organ transplant rejection.
  • diseases or disorders selected from the group consisting of autoimmune diseases, acute chronic inflammation, cancer, cardiovascular disease, infectious disease, and inflammatory disorders including rheumatoid arthritis, chronic inflammatory bowel disease, chronic inflammatory pelvic disease, multiple sclerosis, asthma, osteoarthritis, atherosclerosis, psoriasis, rhinitis, autoimmunity, and organ transplant rejection.
  • compositions include a CXC chemokine analog compound in a therapeutically or prophylactically effective amount sufficient to be used to increase the hemocrit, assist in mobilizing and recovering stem cells, stimulate the production of blood cells, assist in vaccine production, or assist in gene therapy.
  • the amount of a mimetic used in the compositions can vary according to factors such as type of disease, age, sex, and weight of the subject. Dosage regimens may be adjusted to optimize a therapeutic response. In some embodiments, a single bolus may be administered; several divided doses may be administered overtime; the dose may be proportionally reduced or increased; or any combination thereof, as indicated by the exigencies of the therapeutic situation and factors known one of skill in the art. It is to be noted that dosage values may vary with the severity of the condition to be alleviated.
  • Dosage regimens may be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and the dosage ranges set forth herein are exemplary only and do not limit the dosage ranges that may be selected by medical practitioners.
  • administration refers to a method of incorporating a compound into the cells or tissues of a subject, either in vivo or ex vivo to diagnose, prevent, treat, or ameliorate a symptom of a disease.
  • a compound can be administered to a subject in vivo parenterally.
  • a compound can be administered to a subject by combining the compound with cell tissue from the subject ex vivo for purposes that include, but are not limited to, cell expansion and mobilization assays.
  • the terms "administration” or “administering” can include sequential or concurrent incorporation of the compound with the other agents such as, for example, any agent described above.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include, but are not limited to, parenteral such as, for example, intravenous, intradermal, intramuscular, and subcutaneous injection; oral; inhalation; intranasal; transdermal; transmucosal; and rectal administration.
  • An "effective amount" of a compound of the invention can be used to describe a therapeutically effective amount or a prophylactically effective amount.
  • a “therapeutically effective amount” refers to an amount that is effective at the dosages and periods of time necessary to achieve a desired therapeutic result and may also refer to an amount of active compound, prodrug or pharmaceutical agent that elicits any biological or medicinal response in a tissue, system, or subject that is sought by a researcher, veterinarian, medical doctor or other clinician that may be part of a treatment plan leading to a desired effect.
  • the therapeutically effective amount may need to be administered in an amount sufficient to result in amelioration of one or more symptoms of a disorder, prevention of the advancement of a disorder, or regression of a disorder.
  • a therapeutically effective amount can refer to the amount of a therapeutic agent that improves a subject's condition by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 100%.
  • therapeutic effect refers to the inhibition or activation of factors causing or contributing to the abnormal condition (including a disease or disorder).
  • a therapeutic effect relieves or prevents to some extent one or more of the symptoms of the abnormal condition.
  • a therapeutic effect can refer to one or more of the following: (a) an increase or decrease in the number of lymphocytic cells present at a specified location, (b) an increase or decrease in the ability of lymphocytic cells to migrate, (c) an increase or decrease in the response of lymphocytic cells to a stimulus, (d) an increase or decrease in the proliferation, growth, and/or differentiation of cells; (e) inhibition (i.e., slowing or stopping) or acceleration of cell death; (f) relieving, to some extent, one or more of the symptoms associated with an abnormal condition; (g) enhancing or inhibiting the function of the affected population of cells; (h) activating an enzyme activity present in cells associated with the abnormal condition; and (i) inhibiting an enzyme activity present in cells associated with the abnormal condition.
  • abnormal condition refers to a function in the cells or tissues of an organism that deviates from their normal functions in that organism and includes, but is not limited to, conditions commonly referred to as diseases or disorders.
  • An abnormal condition can relate to cell proliferation, cell differentiation, cell survival, cell migration or movement, or the activities of enzymes within a cell.
  • Diseases and disorders may include inflammatory disorders including rheumatoid arthritis, chronic inflammatory bowel disease, chronic inflammatory pelvic disease, multiple sclerosis, asthma, osteoarthritis, atherosclerosis, psoriasis, rhinitis, autoimmunity, organ transplant rejection, and genetic diseases.
  • a prophylactically effective amount refers to an amount that is effective at the dosages and periods of time necessary to achieve a desired prophylactic result.
  • a prophylactic dose is used in a subject prior to the onset of a disease, or at an early stage of the onset of a disease, to prevent or inhibit onset of the disease or symptoms of the disease.
  • a prophylactically effective amount may be less than, greater than, or equal to a therapeutically effective amount.
  • the administration can be oral. In some embodiments, the administration can be subcutaneous injection. In some embodiments, the administration can be intravenous injection using a sterile isotonic aqueous buffer. In some embodiments, the administration can include a solubilizing agent and a local anesthetic such as lignocaine to ease discomfort at the site of injection. In some embodiments, the administrations may be parenteral to obtain, for example, ease and uniformity of administration.
  • the compounds can be administered in dosage units.
  • dosage unit refers to discrete, predetermined quantities of a compound that can be administered as unitary dosages to a subject.
  • a predetermined quantity of active compound can be selected to produce a desired therapeutic effect and can be administered with a pharmaceutically acceptable carrier.
  • the predetermined quantity in each unit dosage can depend on factors that include, but are not limited to, (a) the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of creating and administering such dosage units.
  • a "pharmaceutically acceptable carrier” is a diluent, adjuvant, excipient, or vehicle with which the mimetic is administered.
  • a carrier is pharmaceutically acceptable after approval by a state or federal regulatory agency or listing in the U.S. Pharmacopeia! Convention or other generally recognized sources for use in subjects.
  • the pharmaceutical carriers include any and all physiologically compatible solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. Examples of pharmaceutical carriers include, but are not limited to, sterile liquids, such as water, oils and lipids such as, for example, phospholipids and glycolipids.
  • sterile liquids include, but are not limited to, those derived from petroleum, animal, vegetable or synthetic origin such as, for example, peanut oil, soybean oil, mineral oil, sesame oil, and the like.
  • Water can be a preferred carrier for intravenous administration.
  • Saline solutions, aqueous dextrose and glycerol solutions can also be liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include, but are not limited to, starch, sugars, inert polymers, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol mo ⁇ ostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol, and the like.
  • the composition can also contain minor amounts of wetting agents, emulsifying agents, pH buffering agents, or a combination thereof.
  • the compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • Oral formulations can include standard carriers such as, for example, pharmaceutical grades mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. See Martin, E.W. Remington's Pharmaceutical Sciences. Supplementary active compounds can also be incorporated into the compositions.
  • the carrier is suitable for parenteral administration.
  • the carrier can be suitable for intravenous, intraperitoneal, intramuscular, sublingual or oral administration.
  • the pharmaceutically acceptable carrier may comprise pharmaceutically acceptable salts, such as acid addition salts.
  • salts for purposes of the present invention, the term “salt” and “pharmaceutically acceptable salt” can be used interchangeably in most embodiments.
  • Pharmaceutically acceptable salts are non-toxic at the concentration in which they are administered and include those salts containing sulfate, hydrochloride, phosphate, sulfonate, sulfamate, sulfate, acetate, citrate, lactate, tartrate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, cyclohexylsulfonate, cyclohexylsulfamate, and quinate.
  • Pharmaceutically acceptable salts can be obtained from acids, such as hydrochloric acid, sulfuric acid, phosphoric acid, sulfonic acid, sulfonic acid, sulfamic acid, acetic acid, citric acid, lactic acid, tartaric acid, malonic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, cyclohexylsulfo ⁇ ic acid, cyclohexylsulfamic acid, and quinic acid.
  • acids such as hydrochloric acid, sulfuric acid, phosphoric acid, sulfonic acid, sulfonic acid, sulfamic acid, acetic acid, citric acid, lactic acid, tartaric acid, malonic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic
  • Such salts can be prepared, for example, by reacting the free acid or base form of the product with one or more equivalents of the desired base or acid in a solvent in which the salt is insoluble, or in water that is later removed using a vacuum. Ion exchange can also be used to prepare desired salts.
  • compositions for parenteral administration may include liposomes.
  • Liposomes and emulsions are delivery vehicles or carriers that are especially useful for hydrophobic drugs.
  • additional strategies for protein stabilization may be employed.
  • one may administer the drug in a targeted drug delivery system such as, for example, in a liposome coated with target-specific antibody.
  • the liposomes will bind to the target protein and be taken up selectively by the cell expressing the target protein.
  • compositions typically must be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable for a high drug concentration.
  • the carrier can be a solvent or dispersion medium including, but not limited to, water; ethanol; a polyol such as for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like; and, combinations thereof.
  • the proper fluidity can be maintained in a variety of ways such as, for example, using a coating such as lecithin, maintaining a required particle size in dispersions, and using surfactants.
  • isotonic agents can be used such as, for example, sugars; polyalcohols that include, but are not limited to, mannitol, sorbitol, glycerol, and combinations thereof; and sodium chloride.
  • Sustained absorption characteristics can be introduced into the compositions by including agents that delay absorption such as, for example, monostearate salts, gelatin, and slow release polymers.
  • Carriers can be used to protect active compounds against rapid release, and such carriers include, but are not limited to, controlled release formulations in implants and microencapsulated delivery systems.
  • Biodegradable and biocompatible polymers can be used such as, for example, ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, polylactic acid, polycaprolactone, polyglycolic copolymer (PLG), and the like. Such formulations can generally be prepared using methods known to one of skill in the art.
  • the mimetics are administered by injections that can include intramuscular, intravenous, intra-arterial, intracoronary, intramyocardial, intrapericardial, intraperitoneal, subcutaneous, intrathecal, or intracerebrovascular injections.
  • the compounds may be administered as suspensions such as, for example, oily suspensions for injection.
  • Lipophilic solvents or vehicles include, but are not limited to, fatty oils such as, for example, sesame oil; synthetic fatty acid esters, such as ethyl oleate or triglycerides; and liposomes.
  • Suspensions that can be used for injection may also contain substances that increase the viscosity of the suspension such as, for example, sodium carboxymethyl cellulose, sorbitol, or dextran.
  • a suspension may contain stabilizers or agents that increase the solubility of the compounds and allow for preparation of highly concentrated solutions.
  • a sterile and injectable solution can be prepared by incorporating an effective amount of an active compound in a solvent with any one or any combination of desired additional ingredients described above, filtering, and then sterilizing the solution.
  • dispersions can be prepared by incorporating an active compound into a sterile vehicle containing a dispersion medium and any one or any combination of desired additional ingredients described above.
  • Sterile powders can be prepared for use in sterile and injectable solutions by vacuum drying, freeze-drying, or a combination thereof, to yield a powder that can be comprised of the active ingredient and any desired additional ingredients.
  • the additional ingredients can be from a separately prepared sterile and filtered solution.
  • a mimetic may be prepared in combination with one or more additional compounds that enhance the solubility of the mimetic.
  • the compounds can be administered by inhalation through an aerosol spray or a nebulizer that may include a suitable propellant such as, for example, dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide, or a combination thereof.
  • a dosage unit for a pressurized aerosol may be delivered through a metering valve.
  • capsules and cartridges of gelatin for example, may be used in an inhaler and can be formulated to contain a powderized mix of the compound with a suitable powder base such as, for example, starch or lactose.
  • a therapeutically or prophylactically effective amount of a mimetic may range in concentration from about 0.001 nM to about 0.1 M; from about 0.001 nM to about 0.05 M; from about 0.01 nM to about 15 ⁇ M; from about 0.01 nM to about 10 ⁇ M, or any range therein.
  • the mimetics may be administered in an amount ranging from about 0.001 mg/kg to about 50 mg/kg; from about 0.005 mg/kg to about 40 mg/kg; from about 0.01 mg/kg to about 30 mg/kg; from about 0.01 mg/kg to about 25 mg/kg; from about 0.1 mg/kg to about 20 mg/kg; from about 0.2 mg/kg to about 15 mg/kg; from about 0.4 mg/kg to about 12 mg/kg; from about 0.15 mg/kg to about 10 mg/kg, or any range therein, wherein a human subject is assumed to average about 70 kg.
  • the mimetics can be administered as a diagnostic, therapeutic or prophylactic agent in a combination therapy with the administering of one or more other agents.
  • the agents of the present invention can be administered concomitantly, sequentially, or cyclically to a subject. Cycling therapy involves the administering a first agent for a predetermined period of time, administering a second agent for a second predetermined period of time, and repeating this cycling for any desired purpose such as, for example, to enhance the efficacy of the treatment.
  • the agents can also be administered concurrently.
  • the term "concurrently" is not limited to the administration of agents at exactly the same time, but rather means that the agents can be administered in a sequence and time interval such that the agents can work together to provide additional benefit.
  • Each agent can be administered separately or together in any appropriate form using any appropriate means of administering the agent or agents.
  • each of the agents described herein can be administered to a subject in combination therapy.
  • the agents can be administered at points in time that vary by about 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours, 12 hours, 18 hours, 24 hours, 48 hours or 1 week in time.
  • at least one of the agents is an immunomodulatory agent.
  • the agents can include antiproliferatives, antineoplastics, antimitotics, antiinflammatories, antiplatelets, anticoagulants, antifibrins, antithrombins, antibiotics, antiallergics, antioxidants, and any prodrugs, codrugs, metabolites, analogs, homologues, congeners, derivatives, salts and combinations thereof.
  • the invention includes sustained release formulations for the administration of one or more agents.
  • the sustained release formulations can reduce the dosage and/or frequency of the administrations of such agents to a subject.
  • a CXC chemokine analog may be prepared in a "prodrug" form, wherein the mimetic begins acting upon its metabolism in vivo, in which the mimetic can become, for example, an agonist or an antagonist.
  • the prodrugs can have, for example, an alkyl group attached through a hydrolyzable linkage, such as an ester or anhydride linkage that must hydrolyze before the analog can be active.
  • the analog is a pharmaceutically acceptable salt form of the analog.
  • a CXC chemokine analog can also be hydrolyzably connected to an additional agent and, thus, deliver the additional agent in vivo upon the hydrolysis of the analog from the additional agent; such a construct is known as a "codrug" form of the analog.
  • agents include the bioactive agents, biobeneficial agents, diagnostic agents, and additional CXC chemokine analogs.
  • the agent comprises a glycosaminoglycan such as for example, heparin, hirudin, hyaluronic acid, and any prodrugs, codrugs, metabolites, analogs, homologues, congeners, derivatives, salts and combinations thereof.
  • the agent comprises a phospholipid such as, for example, phosphatidylcholine (lecithin).
  • the phospholipids can be conjugated to any functional group on a CXC chemokine analog, wherein the phospholipid and/or the CXC chemokine analog can be modified as necessary.
  • the phospholipids can be connected to an amino functional group, such as for example the N-terminus of a CXC chemokine analog. It is to be appreciated that one skilled in the art should recognize that some of the groups, subgroups, and individual biobeneficial agents described herein may not be used in some embodiments of the present invention.
  • Phosphatidylcholine is a phospholipid that is a major constituent of cell membranes. Phosphatidylcholine may have hepatoprotective activity, is important for normal cellular membrane composition and repair, and is the major delivery form of the essential nutrient choline, which is a precursor in the synthesis of the neurotransmitter acetylcholine. Phosphatidylcholine's role in the maintenance of cell-membrane integrity is vital to all of the basic biological processes such as, for example, information flow that occurs within cells in the transcription of DNA to RNA; the translation of RNA to proteins; the formation of cellular energy; and intracellular communication or signal transduction.
  • Phosphatidylcholine has a fluidizing effect on cellular membranes, which is important in that a decrease in cell-membrane fluidization, a breakdown of cell-membrane integrity, and an impairment of cell- membrane repair mechanisms are associated with a number of disorders, including, but not limited to liver disease, neurological diseases, various cancers, cell death.
  • the CXC chemokine could be administered with phosphatidylcholine to treat a disease.
  • the disease can include or be associated with liver disease.
  • the liver diseases may include, but are not limited to, alcoholic and non-alcoholic liver disorders such as, for example, fibrosis; cirrhosis; and hepatitis A, B, C and E.
  • the disease can be neurological disease.
  • the neurological diseases include, but are not limited to, manic conditions; cognitive disorders such as old-age memory loss, short-term memory loss, and Alzheimer's Disease; and tardive dyskinesia.
  • the disease can be any cancer that is associated with a deficiency in choline and phosphatidylcholine such as, for example, liver cancer.
  • the disease can be a choline deficiency that results in apoptosis, atherosclerosis or a loss of memory.
  • an effective amount of phosphatidylcholine is a daily administration that ranges from about 10 mg/kg to about 1000 mg/kg, from about 20 mg/kg to about 800 mg/kg, from about 30 mg/kg to about 600 mg/kg, from about 40 mg/kg to about 400 mg/kg, from about 40 mg/kg to about 200 mg/kg, from about 50 mg/kg to about 100 mg/kg, or any range therein.
  • a CXC chemokine analog compound of the invention may be co-administered with a second agent by administering the CXC chemokine before, at the same time, or after the administration of the second agent.
  • the concurrent administration can be made using a co-drug form of the CXC chemokine and second agent, where the separate activities of the two drugs are not realized until the codrug is broken down in vivo, such that the separation of the linkage between the two compounds creates the two separate activities.
  • Peptides of the invention may be synthesized chemically from the C- terminus to the N-terminus ("reverse sequence") using the Fmoc/tBu strategy either manually or automatically using a batchwise or continuous flow peptide synthesizer.
  • Main Solvent a grade certified, ACS spectroanalyzed, N, N- dimethylformamide (DMF) (Fisher, D131-4).
  • the DMF is treated with activated molecular sieves, type 4A (BDH, B54005) for at least two weeks and then tested with 2,4-dinitrofluorobenzene (FDNB) (Eastman).
  • FDNB 2,4-dinitrofluorobenzene
  • Equal volumes of an FDNB solution (1 mg/ml of FDNB in 95% EtOH) and DMF are mixed and allowed to stand for 30 minutes.
  • the absorbance of the mixture is then taken at 381 nm over an FDNB blank solution (no DMF), and if the absorbance is approximately 0.2, then the DMF is suitable for the synthesis.
  • Deblocking Agent 20% piperidine (Aldrich, 10,409-4) in DMF containing 0.5 % (v/v) triton X100 (Sigma, T-9284).
  • Activating Agents 2-(H-benzotriazol-lyl)-1,1,3,3-tetramethyluronium tetrafluoroborate (TBTU) (Quantum RichelLeu, R0139); hydroxybenzotriazole (HOBt) (Quantum RichelLeu, R0166-100), each at a concentration of 0.52 M in DMF; and 4- methylmorpholine (NMM) (Aldrich, M5655-7) at a concentration of 0.9 M in DMF.
  • TBTU 2-(H-benzotriazol-lyl)-1,1,3,3-tetramethyluronium tetrafluoroborate
  • HOBt hydroxybenzotriazole
  • NMM 4- methylmorpholine
  • An Fmoc-L-amino derivative is prepared with protected side-chains.
  • the side-chains are protected using t-butoxycarbonyl (Boc), t-butyl (tBu), and triphenyl methyl (Trt) groups in a 4 fold excess (Peptides Int'l; Bachem; Novabiochem; Chem-lmpex, Inc).
  • the residues to be cyclized for example GIu 60 and Lys 56 in some embodiments, are Allyl-protected (MiIIi pore/Perseptive Biosys.).
  • the synthesis starts from the C-terminus, and the residues are double coupled automatically at ambient temperature using a 4-fold excess of the residues and the coupling reagents, TBTU and HOBt in DMF 1 for each coupling. Double coupling is used to ensure a high yield of coupling and can be a second coupling step that follows single coupling.
  • the synthesis can be interrupted after select residues for cyclization, such as Leu 55 , for lactamization of residues GIu 60 and Lys 66 away from the column.
  • the peptide bound to the support is cyclized by first removing the lateral allyl groups from protected residues, such as GIu 60 and Lys 56 , as described below. The peptide synthesis is then resumed.
  • the support-bound peptide is removed from the column and a 3-fold solution (347mg) of tetrakis(triphenylphosphine) palladium(O) (Pd(PPh 3 M (Sigma- Aldrich, 21,666-6) and 0.1 mmol of the peptide attached to the resin is dissolved in 5% acetic acid.
  • the peptide is activated using 2.5% NMM in CHCI 3 at a concentration of 0.14 M under an argon purge.
  • the solution is added to the support- bound peptide in a reaction vial containing a small magnetic bar for gentle stirring. The mixture is flushed with argon, sealed and stirred at room temperature for 6 hours.
  • the support-bound peptide is transferred to a filter funnel and subject to a series of washes: (i) the first wash is with a 30 ml of a 0.5% (w/w) solution of sodium diethyldithiocarbonate in DMF; (ii) the second wash is with DCM alone; (iii) the third wash is with a 1/1 (v/v) mixture of DCM/DMF; and (iv) the fourth wash is with DMF alone.
  • a positive Kaiser test indicated the deprotection of the amino side chained of the Lys 56 .
  • the support-bound peptide is removed from the synthesizer, placed in a medium filter funnel, washed with DCM to replace the non-volatile DMF, and thoroughly dried under high vacuum for at least two hours, or preferably, overnight.
  • Cleavage Mixture (reagent K): 100 ml of a trifluoroacetic acid (TFA)/Phenol/Water/Thio-Anisol/EDT (82/5/5/5/2.5)(v/v) mixture is prepared.
  • the support-bound peptide (0.5 g) is poured into 7.5 ml of reagent K with gentle agitation on a rocker, allowed to react for 4 hours at room temperature, filtered, and washed with neat TFA.
  • the 7.5 ml of reagent K contains the following:
  • the precipitate is collected at room temperature in a screw-capped 50 ml polypropylene vial after ce ⁇ trifugation for 4 minutes at 2000 rpm in a bench-top centrifuge.
  • the pellets of free peptide were washed 3x with cold ether, centrifuged and dried under a flow of argon.
  • the precipitate was dissolved in 20% acetonitrile with 0.1% TFA and lyophilized.
  • the product is purified and characterized using an analytical HPLC procedure.
  • a Vydac 218TP54 column (C18 reversed-phase, 4.6 mm x 150 mm inner column dimensions, and 5 ⁇ m particle size).
  • a multisolvent mobile phase is used, and the eluants are a 0.1 % TFAZH 2 O (solvent A) and a 0.1 % TFA/acetonitrile (solvent B).
  • Elution Conditions A multisolvent delivery system is used and combines solvent A and solvent B to alter the polarity of the mobile phase during elution.
  • the mobile phase is delivered at a flow rate of 1.0 ml/min and at a concentration of 20- 50% B for 40 minutes; at a concentration of 60-90% B for 5 minutes; at a concentration of 90-20% B for 5 minutes; and at a concentration of 20% B for 10 minutes.
  • the detector is set at 214 nm to read 0.5 absorbance units over a full scale.
  • Agents can be attached as modifying groups that are pendant or in-chain with a CXC chemokine mimetic.
  • a trifunctional amino acid for example, can be incorporated into the CXC chemokine mimetic as a linker and the third functionality can be connected to an agent.
  • Protecting groups can be used to selectively attach an agent to the trifunctional amino acid.
  • Benzyl esters are one type of protecting group that can be used for a lysine carboxyl, for example, and f-butoxycarbonyl can be used for amino groups such as, for example, the amino group in glutamic acid.
  • Amino, hydroxyl and carboxyl groups can be used, for example, as a connecting site for agents.
  • Carboxyl groups can be used as a connecting site for agents having, for example, amino, hydroxyl, or thiol groups.
  • Coupling agents include, but are not limited to, 1-ethyl-3(3-dimethylaminopropyl) carbodiimide (EDC) and 1 ⁇ -dicyclohexylcarbodiimide (DCC).
  • an amine functional compound is 4-amino-TEMPO, an antioxidant and antihypertensive that can be administered as a codrug in combination with a CXC chemokine mimetic.
  • Such an amine functional compound may be connected to an amino acid sequence containing free carboxyls such as, for example, the lysine-derived carboxyls, by first activating the carboxyls and coupling the amine in a solvent under agitation.
  • the carboxyls may be activated with, for example, N-hydroxysuccinimide (NHS) and DCC in a solvent such as, for example, THF or chloroform, which produces N-hydroxysuccinimidyl ester.
  • Examples of the solvent that may be used to couple the amine to the carboxyls include, but are not limited to, THF and DMF.
  • linkages can be preselected and created in order to increase the rate of release of a desired agent from a CXC chemokine mimetic such as, for example, an ester or an anhydride linkage.
  • the reaction can occur at a temperature ranging from about 5°C to about 50 0 C, from about 15°C to about 35 0 C, from about 2O 0 C to about 30 0 C, or any range therein.
  • the reaction time can range from about 0.5 hours to about 24 hours, from about 1 hour to about 18 hours, from about 4 hours to about 16 hours, from about 6 hours to about 12 hours, or any range therein.
  • a benzyl ester protecting group can be removed from a lysine carboxyl by hydrogenolysis with hydrogen gas over a catalyst such as, for example, palladium or platinum on carbon.
  • a catalyst such as, for example, palladium or platinum on carbon.
  • suitable solvents include, but are not limited to, ethanol, methanol, isopropanol, and THF.
  • the reaction may be conducted under about 1 atm of hydrogen for about 6 hours to about 24 hours, for about 8 hours to about 16 hours, for about 10 hours to about 14 hours, or any range therein.
  • a glycosaminoglycan can be connected to an amine functional group as an aldehyde-terminated heparin, for example, to provide additional control over the behavior of the CXC chemokine mimetic in vivo and/or to provide a codrug form of the mimetic.
  • An example of an aldehyde-terminated heparin is represented by the following formula:
  • p is an integer not equal to 0.
  • the aldehyde-terminated heparin can be combined with the amine functional group in a DMF/water solvent and subsequently reduced with NaCNBH 3 to produce a heparin linked to a CXC chemokine mimetic through an amide bond.
  • CXC chemokines and CXC chemokine analogs of the invention may be modified by the addition of polyethylene glycol (PEG).
  • PEG modification may lead to improved circulation time, improved solubility, improved resistance to proteolysis, reduced antigenicity and immunogenicity, improved bioavailability, reduced toxicity, improved stability, and easier formulation (Fora review see, Francis et al., International Journal of Hematology 68:1-18, 1998). PEGylation may also result in a substantial reduction in bioactivity.
  • PEG sizes and derivatives that are commercially designed for specific applications such as, for example, attachment to a variety of different chemical functionalities including, but not limited to, amines, thiols, hydroxyls, sulfhydryls, and carboxyls.
  • an amine group of a CXC chemokine mimetic can be combined with a carboxyl-terminated PEG (Nektar Corp.) in the presence of, for example, EDC or DCC to form a pegylated structure through formation of an amide bond between the CXC chemokine mimetic and the PEG.
  • succinimidyl derivative of mPEG (Nektar Corp.) or an isocyanate-terminated mPEG (Nektar Corp.) can be combined with an CXC chemokine mimetic under conditions known to those of skill in the art.
  • the carboxyl group of an CXC chemokine mimetic can be activated with, for example, EDC or DCC and combined with an amino-terminated mPEG (Nektar Corp.)
  • an amine group of an CXC chemokine mimetic can be combined with a methacrylate-terminated mPEG (Nektar Corp.) in the presence of an initiator capable of undergoing thermal or photolytic free radical decomposition.
  • suitable initiators include benzyl-N,N-diethyldithiocarbamate or p- xylene-N,N-diethyldithiocarbamate.
  • IP-10 CXC chemokines are the subject of U.S. Application No.
  • SEQ ID NOs: 1641 -1645 were prepared for testing their ability to bind to an IP-10 receptor and their efficacy in mediating intracellular calcium mobilization ([Ca 2+ ],) at a variety of concentrations.
  • Binding and Calcium Mobilization Suspensions of CXCR-3/300-19 cells were used to assess binding and intracellular calcium mobilization induced by IP-10 analogs. These are mouse pre-B lymphocytes transfected with the CXCR3 receptor, ⁇ Moser, et al). The cells were washed in RPMI media and resuspended in RPMI media supplemented with 10% FCS, then plated at 1.2 X 10 5 cells per well of 96-well black wall/clear bottom plates coated with poly-D-lysine (Becton Dickinson) and loaded with 100 uL fluorescent calcium indicator FLIPR Calcium 3 assay kit component A (Molecular Probes) for 1hr at 37 0 C. The cells on the plates were then spun at 1000 rpm for 15 minutes at room temperature.
  • FIG. 1 illustates the induction of [Ca 2+ ]J mobilization by select IP-10 analogs at a concentration of 100 ⁇ M according to some embodiments.
  • the results are representative of three independent experiments.
  • SEQ ID NOs: 1641-1645 all bound to the receptor and affected calcium mobilization.
  • SEQ ID NOs: 1641, 1643, and 1644 however, increased calcium mobilization by 300 to over 500%.
  • the results are compared to a recombinant human IP-10 chemokine, as described above.
  • IP-10 analog that is supported by these results would range from about 21 to about 34 amino acids in length and comprise:
  • linker having up to 4 amino acids, wherein the linker is preferably 11-aminoundecanoic acid.
  • Fluo-4,AM loaded SUP-T1 cells (5x10 6 cells/ml), a human lymphoid cell line, were stimulated with SDF-1 and Compound A (SEQ ID NO:809), Compound B (SEQ ID NO:810), Compound C (SEQ ID NO:811 ), Compound D (SEQ ID NO:812) and Compound E (SEQ ID NO:813) at the concentrations indicated.
  • the values represent the mean +/- one S.D. of a representative experiment from three independent experiments.
  • FIGs. 2A and 2B shows the incubation of SUP-T1 cells with SDF-1 according to some embodiments.rief
  • the mimetics used include Compound A (SEQ ID NO:809), Compound B (SEQ ID NO:810), Compound C (SEQ ID NO:811), Compound D (SEQ ID NO:812) or Compound E (SEQ ID NO:813), and the results showed a receptor-mediated induction of [Ca 2+ ]j mobilization.
  • the underlined residues in the structures depicted below were cyclized by a lactamization reaction between lysine and glutamic acid residues.
  • SEQ ID NOs.:809-813 were prepared for testing their ability to bind and activate an SDF-1 receptor, for example, mediate intracellular calcium mobilization ([Ca 2+ ]j) at a variety of concentrations, etc.
  • the [Ca 2+ Jj mobilization assays were conducted as follows. Briefly, SUP- T1 cells (ATCC, Manassas, VA), a human lymphoid cell line, were cultured in RPMI containing phenol red (Invitrogen, Burlington, Ontario, Canada) with 10% fetal bovine serum and antibiotics consisting of 100 U/ml penicillin G sodium and 100 ⁇ g/ml streptomycin sulfate (Invitrogen) at a density between 2x10 5 and 8x10 5 cells/ml.
  • SUP- T1 cells ATCC, Manassas, VA
  • RPMI containing phenol red (Invitrogen, Burlington, Ontario, Canada) with 10% fetal bovine serum and antibiotics consisting of 100 U/ml penicillin G sodium and 100 ⁇ g/ml streptomycin sulfate (Invitrogen) at a density between 2x10 5 and 8x10 5 cells/ml.
  • SDF-1 Compound A (SEQ ID NO:809) F Compound B (SEQ ID NO:810), Compound C (SEQ ID NO:811 ), Compound D (SEQ ID NO:812) or Compound E (SEQ ID NO:813) at the concentrations indicated were injected into aliquots of 5x10 5 cells. Changes in the level of cellular fluorescence were read in a Thermo Labsystems Fluorskan Acsent fluorescence plate reader (VWR, Mississauga, Ontario, Canada). Controls include cells treated with the recombinant chemokine or plain medium. Data is expressed with 100% being the level of fluorescence in plain medium. The values represent the mean +/- one S. D. of a representative experiment from three independent experiments.
  • FIG. 3 shows a competitive dose response for binding to the SDF-1 receptor by native SDF-1 and the CXCR4 agonists (competing ligands) against 125 I-SDF-I according to some embodiments.
  • the Durapore membrane of the Millipore Multiscreen plates was moistened with blocking buffer containing 0.5% BSA (Sigma), 50 mM HEPES, 150 mM NaCI, 5 mM MgCI 2 , 1 mM CaCl 2 and 0.02% sodium azide for 40 min before use.
  • BSA 0.5% BSA
  • 50 mM HEPES 50 mM HEPES
  • 150 mM NaCI 5 mM MgCI 2 , 1 mM CaCl 2
  • 0.02% sodium azide sodium azide
  • This example illustrates the efficacy of SDF-1 peptide analogs (as represented by Compound A (SEQ ID NO:809) and Compound B (SEQ ID NO:810)) in mobilizing circulating neutrophils in a mouse model.
  • This study consisted of three groups of female Balb/c mice (Charles River, Wilmington, MA): an untreated control group of 6 mice and two 18-mouse test groups. Before the start of the study, 20-23g mice were randomly grouped in appropriately labeled cages and identified by cage markings and shaved marks on the dorsal region. The two test groups were treated one time intravenously with SDF-1 analogs at a dose of 2.5 mg/kg in volumes approximating 200 ⁇ l. The evaluated end points included moribundity and complete blood counts with differentials.
  • SEQ ID NOs:3-32 have been prepared to use in the prevention, treatment, and ameliorization of diseases that can benefit from therapeutic angiogenesis.
  • the efficacy of the SDF-1 mimetics of the invention to bind to mammalian, cells and compete with SDF-1 was measured.
  • the experiments include contacting an SDF-1 mimetic with a cell, and the experiments were performed using a human lymphoid cell line of SUP-T1 cells (American Type Culture Collection or ATCC) at a concentration of 5 x 10 s cells/ml.
  • a DURAPORE membrane and Millipore Multiscreen 96-well plates were used in the binding assay, and the membrane was blocked with a PVP/Tween-based blocking buffer before use.
  • the efficacy of the chemokine analogs of the invention to activate mammalian cell receptors is demonstrated by their ability to mobilize intracellular calcium in SUP-T1 cells.
  • the experiments include contacting an SDF-1 mimetic with a cell.
  • SUP-T1 cells (ATCC) were plated on the day of the experiment using 1.2 x 10 5 cells per well in 96-well black-wall/clear-bottom plates coated with poly-D-lysine (BD Biosciences) and loaded using a fluorescent calcium indicator.
  • the indicator used was from a FLIPR Calcium 3 assay kit, component A, (Molecular Probes) and was loaded in the cell for 1hr at 37°C.
  • the SDF-1 mimetics were also shown to induce the survival of Human Umbilical Vein Endothelial Cells (HUVEC) in a serum free medium using an MTT assay to analyse cell viability after peptide treatment.
  • the SDF-1 mimetics were shown to induce the differentiation of Human Vein Endothelial Cells using a matrigel tube formation assay, and they were also shown to induce neo-vessel formation in an aortic ring assay.
  • neovascularization was measured to show the effect on angiogenesis and the ability to induce a vascular supply to promote wound healing, and this was shown using a MATRIGEL plug assay.
  • the IL-8 CXC chemokines are the subject of U.S. Application Nos. 10/932,208 and 10/243,795, each of which is hereby incorporated herein by reference in its entirety.
  • the cross-reference SEQ ID NOs from the source application are used in the explanation and in any associated table or figure, and the SEQ ID NOs used in the present application are provided to allow for location of the sequences in the formal sequence listing of the instant application.
  • SEQ ID NOs 1642-1675 have been prepared to use in the prevention, treatment, and ameliorization of diseases.
  • a competitive-dose-response binding assay was used to compare the ability of the native IL-8 to bind to the CXCR1/CXCR2 receptors with the ability of IL- 8 agonists to bind to the CXCR1/CXCR2 receptors.
  • An 125 I radiolabeled derivative of native IL-8 (“ 125 l-IL-8") was used to measure the binding activity of native IL-8.
  • the competitive dose response is shown in FIG.4.
  • FIG. 4 shows the CXCR2 receptor binding of the IL-8 mimetics as competing ligands according to some embodiments.
  • Differentiated HL-60 cells were assessed for 125 l-IL-8 binding following 2 hours of incubation with IL-8 or its agonist, and 125 l-IL-8.
  • the 125 l-IL-8 was added at a concentration of 2nM in the presence of native IL-8 and the IL-8 mimetics at their respective concentrations as shown.
  • the results are expressed as percentages of the maximal specific binding that was determined without competing ligand and are representative of one independent experiment.
  • HL-60 cells American Type Culture Collection
  • RPMI culture medium containing phenol red, 10% fetal bovine serum, and antibiotics consisting of 100U/ml penicillin G sodium and 100 ⁇ g/ml streptomycin sulfate.
  • the cells were added at a density ranging from about 2x10 5 to about 8x10 5 cells/ml.
  • the cells were then induced to differentiate and express CXCR2 by treating the cells for 3-7 days with 1.25% DMSO.
  • Millipore Multiscreen plates and a Durapore ® membrane (Millipore Corp.) were used for high throughput binding assays.
  • the binding buffer used for the assay consisted of 0.5% (w/v) bovine serum albumin (BSA) (e.g., 0.5 g BSA/100 ml buffer), 5OmM 4-(2-hydroxyethyl)-1 - piperazineethanesulfonic acid (HEPES), 15OmM NaCI, 5mM MgCI 2 , 1mM CaCb and 0.02% sodium azide.
  • BSA bovine serum albumin
  • HEPES 4-(2-hydroxyethyl)-1 - piperazineethanesulfonic acid
  • the HL-60 cells were harvested, washed with plain RPMI, and resuspended in binding buffer at density of about 5x10 6 cells/ml. The cells were preincubated with the IL-8 mimetics for 30 minutes.
  • the binding buffer, the 125 I-IL-S, and the cells incubating with native IL-8 or IL-8 mimetic were then added to wells used to hold the cells in the assay. The cells were then incubated in the wells for another 2 hours with shaking.
  • the IL-8 mimetics were used in concentrations indicated in FIG. 4 with a competitive dose of 2 nM of radiolabeled 125 l-IL-8.
  • Controls include (i) wells with only binding buffer and radiolabeled IL-8 chemokine for background, and (ii) wells with binding buffer, an unlabelled native IL-8 chemokine standard, radiolabeled chemokine and cells for standardization.
  • a dose response curve is developed using a range of native IL-8 concentrations, and a concentration of 0 ⁇ g/ml is included.
  • Each data point is expressed as a percentage of the maximal specific binding that was determined using the radiolabeled native IL-8 without the competing IL-8 mimetics ("IL-8 agonist") and represents measurements obtained from two or three wells in a representative experiment.
  • IL-8 agonist the radiolabeled native IL-8 without the competing IL-8 mimetics
  • FIG. 4 A concentration-dependent inhibition of 125 l-IL-8 binding is shown in FIG. 4 and summarized in Table 11 and indicates the affinity of native IL-8 and IL-8 mimetics for the receptor.
  • Table 11 provides (i) IC50 values for a variety of IL-8 mimetics to show the concentration of a particular IL-8 mimetic that is necessary to provide 50% of the maximal inhibition of 125 l-IL-8 binding that can be obtained with a particular IL-8 mimetic; and (ii) the maximal inhibition of the percent of 125 I-IL-S bound to CXCR2 receptors on differentiated HL-60 cells for both native IL-8 and IL-8 mimetics.
  • the inhibition of 125 l-IL-8 binding by IL-8 mimetics is indicativfe of the relative ability of the analogs to bind to CXCR1/CXCR2 receptors.
  • the results of calcium mobilization assays can be used to show the agonistic activation of the IL-8 receptor by the native IL-8 and IL-8 mimetics.
  • the HL-60 cells are cultured as described above, harvested and suspended in Tyrode's salt solution at a density of about 2x10 6 cells/ml.
  • the Tyrode's salt solution contains about 137 mM NaCI, 2.7mM KCI, 1 mM MgCI 2 , 1 mM CaCI 2 , 0.2 mM NaH 2 PO 4 , 12 mM NaHCO 3 , and 5.5 mM glucose.
  • the cells are labeled with 4 ⁇ M of Fluo-4/AM (Molecular Probes, Inc.) for 45 minutes at 37 ⁇ C to measure calcium mobilization from cells.
  • the label is a dye that fluoresces when bound to calcium.
  • the cells are labeled with the dye to obtain a measure of the amount of calcium released by the cells when the cells are treated with the IL-8 mimetic or native IL-8.
  • An increase in fluorescence indicates an increase in calcium mobilization.
  • the cells are washed three times with the Tyrode's salt solution after labeling and re-suspended at 5 x 10 6 cells/ml.
  • the native IL-8 and IL-8 mimetics are injected to produce a final concentration of about 10 ⁇ g/ml to about 200 ⁇ g/ml in aliquots containing about 5 x 10 5 cells. Changes in the level of cellular fluorescence are read in a Thermo Labsystems Fluorskan Acsent fluorescence plate reader (VWR Scientific Prod's).
  • the controls include cells treated with either the native chemokine or the plain medium of Tyrode's Salt Solution. Data is expressed using 1.0 as the standard level of fluorescence in the plain medium. The reported values represent the mean of at least duplicate measurements from wells in one or more experiments.
  • Table 12 provides a summary of the average fold increase of calcium mobilization in differentiated HL-60 cells over the control wells for native IL-8 and IL- 8 mimetics. Table 12.
  • This example illustrates the efficacy of the IL-8 mimetic a161 (SEQ ID NO:1647) ("the test mimetic") in increasing the number of circulating neutrophils and hematopoietic progenitor/stem cells in a mouse model.
  • the results are shown in FIGs. 5-8.
  • the experiments consisted of the following groups of female Balb/c mice (Charles River Lab's): (1) an untreated control group of 10 mice; and (2) test groups of 10 mice each.
  • mice were randomly grouped in appropriately labeled cages and identified by cage markings and ear punch.
  • the test groups were treated one time subcutaneously with the test analog at doses of 1 , 5, 10, 15, 20, or 25mg/kg in volumes of approximately 200 ⁇ l.
  • the mice were anesthetized immediately before blood collection. Blood samples were obtained from the mice at 30 minutes, 1 hour, 4 hours, 6 hours, 24 hours and/or 48 hours after administration of the test mimetic. Blood was collected with an EDTA S-Monovette syringe (Sarstedt) and 25G needle through a cardiac puncture. Blood was mixed gently by 5 inversions then expelled into a microcentrifuge tube.
  • CFU colony forming units
  • haematopoietic progenitor/stem cells were determined as follows. The volume of blood in each microfuge tube was determined and nine times the volume of ammonium chloride was added. Cells were incubated on ice for 10 minutes to lyse the red blood cells. The cells were washed twice and resuspended in 300 ⁇ L of Iscove's Modified Dulbecco's Medium (IMDM) containing 2% fetal bovine serum.
  • IMDM Iscove's Modified Dulbecco's Medium
  • CFU-GM colony-forming unit granulocyte-macrophage
  • BFU-E burst-forming unit erythroid
  • CFU-GEMM colony-forming unit granulocyte erythrocyte macrophage megakaryocyte
  • FIG. 6 describes the kinetics of the rise in circulating neutrophil counts in response to the administration of the test mimetic according to some embodiments.
  • the test mimetic was administered by subcutaneous injection into female Balb/c mice at 25 mg/kg at time intervals of 30 minutes, 1 hour, 4 hours, and 24 hours. The mice were euthanized and blood was collected by cardiac puncture at each time interval. Complete blood counts and differentials were determined using a Hemavet ® . The values represent the mean +/- one standard deviation for 10 animals per treatment group. Statistically significant elevations as determined using a p value of ⁇ 0.05 are indicated in FIG. 6 by a "*".
  • FIG. 7 shows the response of circulating haematopoietic progenitor/stem cells to the administration of varying doses of the test mimetic according to some embodiments.
  • the test mimetic was administered by subcutaneous injection into female Balb/c mice in amounts of 1 , 5, 10, 15, 20, and 25mg/kg. At 1 hour post- injection, the mice were euthanized and blood was collected by cardiac puncture.
  • CFU-GM colony forming unit granulocyte- macrophage
  • BFU-E burst-forming unit erythroid
  • CFU-GEMM colony forming unit granulocyte erythrocyte macrophage megakaryocyte
  • FIG. 8 describes the kinetics of the rise in haematopoietic progenitor/stem cells in response to the administration of the test mimetic according to some embodiments.
  • the test mimetic was administered by subcutaneous injection into female Balb/c mice at 25 mg/kg. At 30 minutes, 1 hour, 4 hours, 6 hours, 24 hours or 48 hours post-injection, mice were euthanized and blood collected by cardiac puncture.
  • CFU-GM colony forming unit granulocyte-macrophage
  • BFU-E burst-forming unit erythroid
  • CFU-GEMM colony forming unit granulocyte erythrocyte macrophage megakaryocyte
  • PF-4 CXC chemokines are the subject of PCT Application No. PCT/CA2006/001848, which claims the benefit of U.S. Provisional Application No. 60/735,186, each of which is hereby incorporated herein by reference in its entirety.
  • the cross-reference SEQ ID NOs from the source application are used in the explanation with regard to any associated figure or table, and the SEQ ID NOs used in the present application are provided to allow for location of the sequences in the formal sequence listing.
  • FIGs. 9-11 illustrate the efficacy of the PF-4 analogs as agonists according to some embodiments. The efficacy is demonstrated through their ability to inhibit growth of human endothelial cells. The inhibition of endothelial cell growth is an important function of angiostatic compounds. The growth and survival of endothelial cells is tightly regulated by growth factors. The present examples illustrate the ability of PF-4 analogs to inhibit the growth stimulating effects of basic Fibroblast Growth Factor (bFGF) on HUVEC cells.
  • bFGF basic Fibroblast Growth Factor
  • PF-4 analogs can be demonstrated by their ability to block the proliferation of human erythroleukemia cell lines (HEL) with megakaryocyte phenotype.
  • HEL human erythroleukemia cell lines
  • the three analogs tested inhibited the growth of HEL cells at a concentration of 0.1 ⁇ g/ml, as determined using the MTT assay and as illustrated in FIGs 8-10.

Landscapes

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

Abstract

La présente invention concerne de manière générale la conception, la préparation, la dérivation et l'utilisation de mimétiques des chimiokines CXC (CXCL1 à CXCL17) dans la prévention, le traitement et l'amélioration de maladies et de troubles très divers. Généralement parlant, l'invention concerne la conception, la synthèse et l'utilisation d'analogues de chimiokines qui se lient aux récepteurs CXCR1 à CXCR7 des chimiokines CXC, de sorte que ces analogues peuvent être conçus pour influer sur l'activité du récepteur soit comme un agoniste, soit comme un antagoniste. Ces analogues peuvent être utiles pour traiter des maladies et des troubles très divers et ils peuvent également servir d'appoint dans le traitement de maladies et troubles divers.
PCT/US2007/000436 2006-01-04 2007-01-04 Conception d'analogues de chimiokines cxc pour le traitement de maladies humaines WO2007079460A2 (fr)

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
US75585906P 2006-01-04 2006-01-04
US60/755,859 2006-01-04
US11/388,542 2006-03-24
US11/388,542 US7368425B2 (en) 2006-03-24 2006-03-24 Cyclic peptides for modulating growth of neo-vessels and their use in therapeutic angiogenesis
US11/393,769 2006-03-29
US11/393,769 US7994114B2 (en) 2000-09-14 2006-03-29 Chemokine mimetics synthesis and their use
US11/494,232 US20070066523A1 (en) 2002-09-13 2006-07-26 Chemokine analogs for the treatment of human diseases
US11/494,232 2006-07-26
US11/590,210 2006-10-30
US11/590,210 US20070116669A1 (en) 2002-09-13 2006-10-30 Interferon-inducible protein-10 (IP-10 or CXCL10) chemokine analogs for the treatment of human diseases
CAPCT/CA2006/001848 2006-11-10
PCT/CA2006/001848 WO2007053952A1 (fr) 2005-11-10 2006-11-10 Analogues du facteur plaquettaire 4 (pf-4) et leurs utilisations

Publications (2)

Publication Number Publication Date
WO2007079460A2 true WO2007079460A2 (fr) 2007-07-12
WO2007079460A3 WO2007079460A3 (fr) 2007-09-13

Family

ID=38190734

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/000436 WO2007079460A2 (fr) 2006-01-04 2007-01-04 Conception d'analogues de chimiokines cxc pour le traitement de maladies humaines

Country Status (1)

Country Link
WO (1) WO2007079460A2 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7696309B2 (en) 2006-10-23 2010-04-13 The Brigham And Women's Hospital, Inc. Protease resistant mutants of stromal cell derived factor-1 in the repair of tissue damage
EP2182976A1 (fr) * 2007-08-31 2010-05-12 Massachusetts Institute of Technology Traitement d'une maladie auto-immune
US9308277B2 (en) 2010-02-25 2016-04-12 Mesoblast International Sàrl Protease-resistant mutants of stromal cell derived factor-1 in the repair of tissue damage
EP3305304A4 (fr) * 2015-05-29 2019-01-23 Seikagaku Corporation Composition comprenant un dérivé de glucosaminoglycane et un régulateur d'activité de récepteur de chimiokine
US10662234B2 (en) 2011-06-07 2020-05-26 Mesoblast International Sàrl Methods for repairing tissue damage using protease-resistant mutants of stromal cell derived factor-1

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000059928A1 (fr) * 1999-04-07 2000-10-12 Thomas Jefferson University Peptides synthetiques issus de chimiokines
WO2001076615A2 (fr) * 2000-04-12 2001-10-18 The University Of British Columbia Traitement de cellules hematopoietiques utilisant des agonistes de cxcr4
WO2001085196A2 (fr) * 2000-05-09 2001-11-15 The University Of British Columbia Traitement de cellules hématopoiétiques basé sur des antagonistes du cxcr4
WO2004024088A2 (fr) * 2002-09-13 2004-03-25 Chemokine Therapeutics Corp. Conception d'analogues de chimiokines permettant de traiter des maladies humaines

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000059928A1 (fr) * 1999-04-07 2000-10-12 Thomas Jefferson University Peptides synthetiques issus de chimiokines
WO2001076615A2 (fr) * 2000-04-12 2001-10-18 The University Of British Columbia Traitement de cellules hematopoietiques utilisant des agonistes de cxcr4
WO2001085196A2 (fr) * 2000-05-09 2001-11-15 The University Of British Columbia Traitement de cellules hématopoiétiques basé sur des antagonistes du cxcr4
WO2004024088A2 (fr) * 2002-09-13 2004-03-25 Chemokine Therapeutics Corp. Conception d'analogues de chimiokines permettant de traiter des maladies humaines

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BELPERIO J A ET AL: "CXC chemokines in angiogenesis." JOURNAL OF LEUKOCYTE BIOLOGY JUL 2000, vol. 68, no. 1, July 2000 (2000-07), pages 1-8, XP002440747 ISSN: 0741-5400 *
CLARK-LEWIS I ET AL: "STRUCTURE-ACTIVITY RELATIONSHIPS OF CHEMOKINES" JOURNAL OF LEUKOCYTE BIOLOGY, FEDERATION OF AMERICAN SOCIETIES FOR EXPERIMENTAL, US, vol. 57, no. 5, 1 May 1995 (1995-05-01), pages 703-711, XP000569546 ISSN: 0741-5400 *
DORSEY RUSSELL ET AL: "Immunotherapy with interleukin-10 depends on the CXC chemokines inducible protein-10 and monokine induced by IFN-gamma." CANCER RESEARCH 1 MAY 2002, vol. 62, no. 9, 1 May 2002 (2002-05-01), pages 2606-2610, XP002440745 ISSN: 0008-5472 *
GALOPPINI C ET AL: "A new class of pseudopeptide antagonists of the kinin B1 receptor containing alkyl spacers." JOURNAL OF MEDICINAL CHEMISTRY 11 FEB 1999, vol. 42, no. 3, 11 February 1999 (1999-02-11), pages 409-414, XP002440744 ISSN: 0022-2623 *
HOUSHMAND PANTEA ET AL: "Therapeutic applications in the chemokine superfamily." CURRENT OPINION IN CHEMICAL BIOLOGY AUG 2003, vol. 7, no. 4, August 2003 (2003-08), pages 457-460, XP002440746 ISSN: 1367-5931 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7696309B2 (en) 2006-10-23 2010-04-13 The Brigham And Women's Hospital, Inc. Protease resistant mutants of stromal cell derived factor-1 in the repair of tissue damage
US7999067B2 (en) 2006-10-23 2011-08-16 The Brigham And Women's Hospital, Inc. Protease resistant mutants of stromal cell derived factor-1 in the repair of tissue damage
US9631005B2 (en) 2006-10-23 2017-04-25 The Brigham And Women's Hospital, Inc. Protease resistant mutants of stromal cell derived factor-1 in the repair of tissue damage
US10774124B2 (en) 2006-10-23 2020-09-15 The Brigham And Women's Hospital, Inc. Protease resistant mutants of stromal cell derived factor-1 in the repair of tissue damage
EP2182976A1 (fr) * 2007-08-31 2010-05-12 Massachusetts Institute of Technology Traitement d'une maladie auto-immune
EP2182976A4 (fr) * 2007-08-31 2012-01-25 Massachusetts Inst Technology Traitement d'une maladie auto-immune
US9308277B2 (en) 2010-02-25 2016-04-12 Mesoblast International Sàrl Protease-resistant mutants of stromal cell derived factor-1 in the repair of tissue damage
US10456451B2 (en) 2010-02-25 2019-10-29 Mesoblast International Sàrl Protease-resistant mutants of stromal cell derived factor-1 in the repair of tissue damage
US10662234B2 (en) 2011-06-07 2020-05-26 Mesoblast International Sàrl Methods for repairing tissue damage using protease-resistant mutants of stromal cell derived factor-1
EP3305304A4 (fr) * 2015-05-29 2019-01-23 Seikagaku Corporation Composition comprenant un dérivé de glucosaminoglycane et un régulateur d'activité de récepteur de chimiokine
US11253540B2 (en) 2015-05-29 2022-02-22 Seikagaku Corporation Composition including glycosaminoglycan derivative and chemokine receptor activity regulator

Also Published As

Publication number Publication date
WO2007079460A3 (fr) 2007-09-13

Similar Documents

Publication Publication Date Title
US20070160574A1 (en) Design of CXC chemokine analogs for the treatment of human diseases
AU762472B2 (en) Therapeutic chemokine receptor antagonists
EP1286684B1 (fr) Utilisation d'antagonistes du cxcr4 pour traiter les maladies autoimmunes et le cancer
US6875738B1 (en) Therapeutic chemokine receptor antagonists
EP1834961A2 (fr) Conception d'analogues de chimiokines permettant de traiter des maladies humaines
US20060014682A1 (en) CXCR4 antagonist treatment of hematopoietic cells
AU2001252081B9 (en) CXCR4 Agonist Treatment of Hematopoietic Cells
WO2000009152A1 (fr) Antagonistes therapeutiques du recepteur de la chimiokine
EP1642588A2 (fr) Mimétiques de l'interleukine-8 et méthodes pour leur utilisation dans la prévention, le traitement, le diagnostic et l'amélioration des symptomes d'une maladie
US7378098B2 (en) CXC chemokine receptor 4 agonist peptides
AU2001252081A1 (en) CXCR4 Agonist Treatment of Hematopoietic Cells
EP1515990B1 (fr) Antagonistes de chimiokines cxc a liaison cxcr3
WO2007079460A2 (fr) Conception d'analogues de chimiokines cxc pour le traitement de maladies humaines
US7939498B2 (en) Cyclic peptides for preserving or enhancing the function of an organ allograft
WO1996009062A1 (fr) Agonistes et antagonistes polypeptidiques de l'interleukine-8 humaine
AU2005201244B2 (en) CXCR4 agonist treatment of hematopoietic cells
AU2006252080A1 (en) Design of interferon-inducible protein-10 (IP-10 or CXCL10) chemokine analogs for the treatment of human diseases
CA2322764A1 (fr) Antagonistes therapeutiques du recepteur de chemokine
AU2003279715A1 (en) Design of chemokine analogs for the treatment of human diseases
CA2405907A1 (fr) Traitement de cellules hematopoietiques utilisant des agonistes de cxcr4
WO2005056581A2 (fr) Peptide susceptible de se lier specifiquement a un recepteur de chimiokine et son utilisation

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07717703

Country of ref document: EP

Kind code of ref document: A2