WO2010092440A1 - Antagonistes du récepteur cxcr2 pour le traitement ou la prévention de la résistance à l'insuline - Google Patents

Antagonistes du récepteur cxcr2 pour le traitement ou la prévention de la résistance à l'insuline Download PDF

Info

Publication number
WO2010092440A1
WO2010092440A1 PCT/IB2009/052312 IB2009052312W WO2010092440A1 WO 2010092440 A1 WO2010092440 A1 WO 2010092440A1 IB 2009052312 W IB2009052312 W IB 2009052312W WO 2010092440 A1 WO2010092440 A1 WO 2010092440A1
Authority
WO
WIPO (PCT)
Prior art keywords
urea
hydroxy
cxcl5
alkyl
cxcr2
Prior art date
Application number
PCT/IB2009/052312
Other languages
English (en)
Inventor
Lluis Fajas
Carine Chavey
Original Assignee
Inserm (Institut National De La Sante Et De La Recherche Medicale)
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Inserm (Institut National De La Sante Et De La Recherche Medicale) filed Critical Inserm (Institut National De La Sante Et De La Recherche Medicale)
Priority to PCT/IB2009/052312 priority Critical patent/WO2010092440A1/fr
Publication of WO2010092440A1 publication Critical patent/WO2010092440A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/17Amides, e.g. hydroxamic acids having the group >N—C(O)—N< or >N—C(S)—N<, e.g. urea, thiourea, carmustine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin

Definitions

  • the present invention relates to CXCR2 receptor antagonists or inhibitors of CXCR2 receptor gene expression for the treatment or the prevention of insulin resistance.
  • Insulin resistance is the condition in which normal amounts of insulin are inadequate to produce a normal insulin response from adipose, muscle and liver cells.
  • Insulin resistance in fat cells reduces the effects of insulin and results in elevated hydrolysis of stored triglycerides in the absence of measures which either increase insulin sensitivity or which provide additional insulin. Increased mobilization of stored lipids in these cells elevates free fatty acids in the blood plasma.
  • Insulin resistance in muscle cells reduces glucose uptake (and so local storage of glucose as glycogen), whereas insulin resistance in liver cells results in impaired glycogen synthesis and a failure to suppress glucose production. Elevated blood fatty acid levels (associated with insulin resistance and diabetes mellitus Type 2), reduced muscle glucose uptake, and increased liver glucose production all contribute to elevated blood glucose levels. High plasma levels of insulin and glucose due to insulin resistance are believed to be the origin of metabolic syndrome and type 2 diabetes, including its complications.
  • WAT White adipose tissue
  • WAT is primarily involved in energy storage in the form of triglycerides, and energy release in the form of free fatty acids.
  • WAT is no longer considered only as a fat storage organ. Instead, WAT secrete numerous factors that play a role in several physiological and pathological processes including insulin resistance (reviewed in (Ahima and Flier, 2000; Trujillo and Scherer, 2006)). WAT can therefore be considered as a sensory organ. External factors such as nutritional status, stress, physical exercise, infection or trauma, will affect WAT biology through various signaling pathways including growth factors and fatty acids. In turn, WAT will then signal other organs in an endocrine manner, thereby transmitting a metabolic, proliferative, growth, or differentiation response.
  • Adipokines are factors secreted by the different cell compartments of WAT, such as adipocytes or macrophages, and were initially characterized as regulators of metabolic processes, such as regulation of food intake, energy homeostasis, adipocyte differentiation, or insulin sensitivity. Subsequently, it was found that adipokines could modulate inflammatory processes. These adipokines include WAT-specific factors, such as leptin, adiponectin, and well-known cytokines secreted by several cell types, such as TNF ⁇ , IL-6, IL-8, IL-1 , or monocyte chemoattractant protein-1 (MCP-1 ).
  • WAT-specific factors such as leptin, adiponectin
  • cytokines secreted by several cell types, such as TNF ⁇ , IL-6, IL-8, IL-1 , or monocyte chemoattractant protein-1 (MCP-1 ).
  • Increased WAT mass is correlated with chronic systemic inflammatory response, demonstrated by increased infiltration of macrophages in WAT (Weisberg et al., 2003; Xu et al., 2003).
  • This inflammatory state results in the development of obesity-associated pathologies including atherosclerosis, hypertension, non alcoholic steatohepatitis, and insulin resistance.
  • These obesity-associated pathologies are the result, at least in part, of changes in the secretion of adipokines, and illustrate the link between inflammatory state and metabolic response (Hotamisligil, 2006).
  • Chemokines are proinflammatory cytokines that stimulate leukocyte chemoattraction and are produced in response to infectious and other inflammatory stimuli by a number of different cell types. To date more than 50 proinflammatory cytokines or chemokines have been identified and have been classified into four groups according to the location of the conserved cysteine residues: CXCL, CCL, CL, and CX3CL (Zlotnik et al., 2006). These chemokines play differential roles in specifically recruiting different cell types to an inflammatory site.
  • CXCL5 or epithelial neutrophil activating peptide is a cytokine belonging to the family of chemokines that is mainly implicated in the chemotaxis of inflammatory cells through the generation of local concentration gradients (WaIz et al., 1991 ; WaIz et al., 1997). It has been shown, to be a recruiter of neutrophils and involved in their activation, through interaction with the CXCR2 receptor. This C-X-C chemokine has been implicated in pulmonary disease, lung cancer, arthritis, and other pathological states (WaIz et al., 1991 ; WaIz et al., 1993; Wislez et al., 2004).
  • the present invention relates to a CXCR2 receptor antagonist for the treatment or the prevention of insulin resistance.
  • the present invention also relates to an inhibitor of CXCR2 receptor gene expression for the treatment or the prevention of insulin resistance.
  • the present invention further relates to a method for diagnosing and/or prognosing insulin resistance in a patient comprising detecting the presence and/or quantifying CXCL5 in a biological sample obtained from said patient.
  • CXCR2 or "CXCR2 receptor” are used interchangeably and have their general meaning in the art.
  • the CXCR2 receptor can be from any source, but typically is a mammalian (e.g., human and non-human primate) CXCR2 receptor, particularly a human CXCR2 receptor.
  • CXCL5 can be from any source, but typically is a mammalian (e.g., human and non-human primate) CXCL5, particularly a human CXCL5.
  • an “inhibitor of gene expression” refers to a natural or synthetic compound that has a biological effect to inhibit or significantly reduce the expression of a gene. Consequently an “inhibitor of CXCR2 receptor gene expression” refers to a natural or synthetic compound that has a biological effect to inhibit or significantly reduce the expression of the gene encoding for the CXCR2 receptor.
  • receptor antagonist is meant a natural or synthetic compound that has a biological effect opposite to that of a receptor agonist.
  • the term is used indifferently to denote a “true” antagonist and an inverse agonist of a receptor.
  • a "true” receptor antagonist is a compound which binds the receptor and blocks the biological activation of the receptor, and thereby the action of the receptor agonist, for example, by competing with the agonist for said receptor.
  • An inverse agonist is a compound which binds to the same receptor as the agonist but exerts the opposite effect. Inverse agonists have the ability to decrease the constitutive level of receptor activation in the absence of an agonist.
  • CXCR2 receptor antagonist includes any entity that, upon administration to a patient, results in inhibition or down-regulation of a biological activity associated with activation of the CXCR2 receptor by CXCL5 in the patient, including any of the downstream biological effects otherwise resulting from the binding to CXCR2 receptor with CXCL5.
  • Such CXCR2 receptor antagonists include any agent that can block CXCR2 receptor activation or any of the downstream biological effects of CXCR2 receptor activation.
  • a CXCR2 receptor antagonist e.g.
  • an antibody directed against CXCR2 can act by occupying the ligand binding site or a portion thereof of the CXCR2 receptor, thereby making CXCR2 receptor inaccessible to its natural ligand, CXCL5, so that its normal biological activity is prevented or reduced.
  • CXCR2 receptor antagonist includes also any agent able to interact with the natural ligand of CXCR2, namely CXCL5.
  • Said agent may be an antibody directed against CXCL5 which can block the interaction between CXCL5 and CXCR2 or which can block the activity of CXCL5 (“neutralizing antibody").
  • CXCR2 receptor antagonists are small organic molecules
  • said antagonists are preferably selective for the CXCR2 receptor as compared with the other CXC receptors, such as CXCR1 receptor.
  • selective it is meant that the affinity of the antagonist for the CXCR2 receptor is at least 10-fold, preferably 25-fold, more preferably 500-fold, still preferably 150-fold higher than the affinity for the other CXC receptor (CXCR1 ).
  • the affinity of an antagonist for CXCR2 receptor may be quantified by measuring the activity of CXCR2 receptor in the presence a range of concentrations of said antagonist in order to establish a dose-response curve.
  • an IC 5 O value may be deduced which represents the concentration of antagonist necessary to inhibit 50% of the response to an agonist in defined concentration.
  • the IC 5 O value may be readily determined by the one skilled in the art by fitting the dose-response plots with a dose-response equation as described by De Lean et al. (1979). IC 5 O values can be converted into affinity constant (Ki) using the assumptions of Cheng and Prusoff (1973).
  • the term "small organic molecule” refers to a molecule of a size comparable to those organic molecules generally used in pharmaceuticals. The term excludes biological macromolecules (e. g., proteins, nucleic acids, etc.). Preferred small organic molecules range in size up to about 5000 Da, more preferably up to 2000 Da, and most preferably up to about 1000 Da.
  • a subject denotes a mammal, such as a rodent, a feline, a canine, and a primate.
  • a subject according to the invention is a human.
  • insulin resistance refers to a condition where a normal amount of insulin is unable to produce a normal physiological or molecular response and preferably to obesity-induced insulin resistance
  • treating refers to reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.
  • prevention of insulin resistance may refer to the administration of the compounds of the present invention prevent the symptoms of insulin resistance.
  • detecting means determining if CXCL5 is present or not in a biological sample and “quantifying” means determining the amount of CXCL5 in a biological sample.
  • “Pharmaceutically” or “pharmaceutically acceptable” refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate.
  • a pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type
  • the present invention provides methods and compositions (such as pharmaceutical compositions) for treating or preventing insulin resistance.
  • the invention relates to a CXCR2 receptor antagonist for treating or preventing insulin resistance.
  • the CXCR2 receptor antagonist may be a low molecular weight antagonist, e. g. a small organic molecule.
  • CXCR2 antagonists that are contemplated by the invention include but are not limited to those described in International Patent Application Nos
  • the CXCR2 receptor antagonists described in the international Patent Application WO96/25157 have the following formula:
  • X is oxygen or sulfur
  • R is any functional moiety having an ionizable hydrogen and a pKa of IO or less;
  • Ri is independently selected from hydrogen; halogen; nitro; cyano; halosubstituted CMO alkyl; CMO alkyl; C 2- io alkenyl; CMO alkoxy; halosubstituted C MO alkoxy; azide; S(OJtR 4 : hydroxy; hydroxy Ci -4 alkyl; aryl; aryl Ci -4 alkyl; aryloxy; aryl Ci -4 alkyloxy; heteroaryl; heteroarylalkyl; heterocyclic; heterocyclic Ci -4 alkyl; heteroaryl Ci -4 alkyloxy; aryl C 2- io alkenyl; heteroaryl C2-10 alkenyl; heterocyclic C2-10 alkenyl; NR 4 R 5 ; C2-10 alkenyl;
  • C(O)NR 4 R 5 ; C(O)NR 4 R 5 ; C(O)NR 4 Ri 0 ; S(O) 3 H; S(O) 3 R 8 ; CM 0 alkyl C(O)Rn; C2-10 alkenyl C(O)Rn; C2-10 alkenyl C(O)ORn; C(O)Rn; C(O)ORn; C(O)ORi 2 ; OC(O) Ri 1 ; NR 4 C(O)Rn; or two Ri moieties together may form O-(CH 2 ) S O or a 5 to 6 membered unsaturated ring; t is 0 or an integer having a value of 1 or 2; s is an integer having a value of 1 to 3; R 4 and R 5 are independently hydrogen, optionally substituted Ci -4 alkyl, optionally substituted aryl, optionally substituted aryl Ci -4 alkyl, optionally substituted heteroaryl, optionally substituted heteroaryl Ci
  • Y is independently selected from hydrogen; halogen; nitro; cyano; halosubstituted C MO alkyl; C MO alkyl; C 2- io alkenyl; C M 0 alkoxy; halosubstituted C M 0 alkoxy; azide; S(O)tR 4 : hydroxy; hydroxy C i -4 alkyl; aryl; aryl C i -4 alkyl; aryloxy; aryl C i -4 alkyloxy; heteroaryl; heteroarylalkyl; heteroaryl C i -4 alkyloxy; heterocyclic; heterocyclic Ci -4 alkyl; aryl C 2- io alkenyl; heteroaryl C 2- io alkenyl; heterocyclic C 2- i 0 alkenyl; NR 4 R 5 ; C 2- i 0 alkenyl
  • OC(O) R 11 ; NR 4 C(O)Rn; or two Ri moieties together may form O-(CH 2 ) S O or a 5 to 6 membered unsaturated ring; n is an integer having a value of 1 to 3; m is an integer having a value of 1 to 3;
  • R 8 is hydrogen or Ci -4 alkyl
  • R 10 is Ci-IO alkyl C(O) 2 R 8 ;
  • Rn is hydrogen, Ci -4 alkyl, optionally substituted aryl, optionally substituted aryl Ci -4 alkyl, optionally substituted heteroaryl, optionally substituted heteroaryl Ci -4 alkyl, optionally substituted heterocyclic, or optionally substituted heterocyclic Ci -4 alkyl;
  • Ri 2 is hydrogen, C MO alkyl, optionally substituted aryl or optionally substituted arylalkyl; or a pharmaceutically acceptably salt thereof.
  • the receptor CXCR2 antagonists are selected from those described in the international Patent Application WO96/25157 and that are listed below:
  • N-(2-Hydroxy-4-nitrophenyl)-N'-(2-phenylphenyl)urea N-(2-Hydroxy-4-nitrophenyl)-N'-(2-methylthiophenyl)urea, N-(2-Hydroxy-4-nitrophenyl)-N'-(2,3-dichlorophenyl)urea, N-(2-Hydroxy-4-nitro phenyl) N'-(2-chloro phenyl)urea, N-(2-Hydroxy-4-nitrophenyl)-N'-(2,3-methylenedioxyphenyl)urea,
  • N-(2-Hydroxy-4-nitrophenyl)-N'-(2-methoxy-3-chlorophenyl)urea N-(2-Hydroxy 4-nitro phenyl) N'-(2-phenyloxy phenyl)urea, N-(3-Chloro-2-hydroxyphenyl)-N'-(bromophenyl)urea, N-(2-Hydroxy-3-glycmemethylestercarbonylphenyl)-N'-(2-bromophenyl)urea, N-(3-Nitro-2-hydroxyphenyl)-N'-(2-bromophenyl)urea,
  • N-(2-Hydroxy-4-cyanophenyl)-N'-(2-bromophenyl)urea N-(2-Hydroxy-3,4-dichlorophenyl)-N'-(2-bromophenyl)urea, N-(3-Cyano-2-hydroxyphenyl)-N'-(2-bromophenyl)urea, N-(2-Hydroxy-4-cyanophenyl)-N'-(2-methoxyphenyl)urea, N-(2-Hydroxy-4-cyanophenyl)-N'-(2-phenylphenyl)urea,
  • N-(2-Hydroxy-4-cyanophenyl-N'-(2,3-dichlorophenyl)urea N-(2-Hydroxy-4-cyanophenyl)-N'-(2-methylphenyl)urea, N-(2-Hydroxy-3-cyano-4-methylphenyl)-N'-(2-bromophenyl)urea, N-(4-Cyano-2-hydroxyphenyl)-N'-(2-thfluoromethylphenyl)urea, N-(3-Trifluoromethyl-2-hydroxyphenyl)-N'-(2-bromophenyl)urea,
  • N-(3-Phenylaminocarbonyl-2-hydroxyphenyl)-N'-(2-bromophenyl)urea N-(2-Hydroxy 4-nitro phenyl) N'-(2-iodo phenyl) urea, N-(2-Hydroxy 4-nitro phenyl) N'(2-bromo phenyl)thiourea, N-(2-Phenylsulfonamido)-4-cyanophenyl-N'(2-bromo phenyl)urea, (E)-N-[3-[(2-Aminocarbonyl)ethenyl)]-2-hydroxyphyenyl]-N'-(2- bromophenyl)urea,
  • N-(2-Hydroxy.3.4-dichlorophenyl)-N'-(2-methoxyphenyl)urea N-(2-Hydroxy.3.4-dichloropyenyl)-N'-(2-phenylphenyl)urea
  • N-(2-Hydroxy.3.4-dichlorophenyl)-N'-(2.3-dichlorophenyl)urea N-(2-Hydroxy-5-nitrophenyl)-N'-(2,3-dichlorophenyl)urea
  • N-(2-Hydroxy-3-cyanophenyl)-N'-(2,3-dichlorophenyl)urea N-(2-Hydroxy-3-cyanophenyl)-N'-(2,3-dichlorophenyl)urea.
  • the CXCR2 receptor antagonist is N-(2-Hydroxy-4- nitrophenyl)-N'-(2-bromophenyl)urea (also named SB 225002) described by White
  • the CXCR2 receptor antagonist may consist in an antibody (the term including antibody fragment) that can block CXCR2 receptor activation.
  • the CXCR2 receptor antagonist may consist in an antibody directed against the CXCR2 receptor or CXCL5, in such a way that said antibody impairs the binding of a CXCL5 to CXCR2.
  • the CXCR2 receptor antagonist may be an anti- CXCL5 neutralizing antibody (Arenberg et al., 1998; Halloran et al., 1999).
  • Antibodies directed against the CXCR2 receptor or CXCL5 can be raised according to known methods by administering the appropriate antigen or epitope to a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others.
  • a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others.
  • Various adjuvants known in the art can be used to enhance antibody production.
  • antibodies useful in practicing the invention can be polyclonal, monoclonal antibodies are preferred.
  • Monoclonal antibodies against CXCR2 receptor or CXCL5 can be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture.
  • Techniques for production and isolation include but are not limited to the hybridoma technique originally described by Kohler and Milstein (1975); the human B-cell hybridoma technique (Cote et al., 1983); and the EBV-hybhdoma technique (Cole et al. 1985).
  • techniques described for the production of single chain antibodies can be adapted to produce anti-CXCR2, or anti- CXCL5 single chain antibodies.
  • CXCR2 receptor antagonists useful in practicing the present invention also include anti- CXCR2, or anti-CXCL5 antibody fragments including but not limited to F(ab')2 fragments, which can be generated by pepsin digestion of an intact antibody molecule, and Fab fragments, which can be generated by reducing the disulfide bridges of the F(ab')2 fragments.
  • F(ab')2 fragments which can be generated by pepsin digestion of an intact antibody molecule
  • Fab fragments which can be generated by reducing the disulfide bridges of the F(ab')2 fragments.
  • Fab and/or scFv expression libraries can be constructed to allow rapid identification of fragments having the desired specificity to CXCR2 receptor.
  • Humanized anti-CXCR2 receptor or anti-CXCL5 antibodies and antibody fragments therefrom can also be prepared according to known techniques.
  • Humanized antibodies are forms of non-human (e.g., rodent) chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (CDRs) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity.
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • the CXCR2 receptor antagonist is an aptamer directed against CXCR2 or CXCL5.
  • Aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition.
  • Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.
  • Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library, as described in Tuerk C. and Gold L., 1990.
  • the random sequence library is obtainable by combinatorial chemical synthesis of DNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence.
  • Peptide aptamers consists of a conformationally constrained antibody variable region displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods (Colas et al., 1996).
  • Another aspect of the invention relates to an inhibitor of CXCR2 receptor gene expression for treating or preventing insulin resistance.
  • Inhibitors of CXCR2 receptor gene expression for use in the present invention may be based on anti-sense oligonucleotide constructs.
  • Anti-sense oligonucleotides including anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of CXCR2 receptor mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of CXCR2 receptors, and thus activity, in a cell.
  • antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence encoding CXCR2 receptor can be synthesized, e.g., by conventional phosphodiester techniques and administered by e.g., intravenous injection or infusion.
  • Methods for using antisense techniques for specifically inhibiting gene expression of genes whose sequence is known are well known in the art (e.g. see U.S. Pat. Nos. 6,566,135; 6,566,131 ; 6,365,354; 6,410,323; 6,107,091 ; 6,046,321 ; and 5,981 ,732).
  • Small inhibitory RNAs can also function as inhibitors of CXCR2 receptor gene expression for use in the present invention.
  • CXCR2 receptor gene expression can be reduced by contacting a subject or cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that CXCR2 receptor gene expression is specifically inhibited (i.e. RNA interference or RNAi).
  • dsRNA small double stranded RNA
  • RNAi RNA interference
  • Methods for selecting an appropriate dsRNA or dsRNA-encoding vector are well known in the art for genes whose sequence is known (e.g. see Tuschl, T. et al. (1999); Elbashir, S. M. et al. (2001 ); Hannon, GJ.
  • Ribozymes can also function as inhibitors of CXCR2 receptor gene expression for use in the present invention. Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. The mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of CXCR2 receptor mRNA sequences are thereby useful within the scope of the present invention.
  • Specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site can be evaluated for predicted structural features, such as secondary structure, that can render the oligonucleotide sequence unsuitable.
  • the suitability of candidate targets can also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using, e.g., ribonuclease protection assays.
  • antisense oligonucleotides and ribozymes useful as inhibitors of CXCR2 receptor gene expression can be prepared by known methods. These include techniques for chemical synthesis such as, e.g., by solid phase phosphoramadite chemical synthesis. Alternatively, anti-sense RNA molecules can be generated by in vitro or in vivo transcription of DNA sequences encoding the RNA molecule. Such DNA sequences can be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Various modifications to the oligonucleotides of the invention can be introduced as a means of increasing intracellular stability and half-life.
  • Antisense oligonucleotides siRNAs and hbozymes of the invention may be delivered in vivo alone or in association with a vector.
  • a "vector" is any vehicle capable of facilitating the transfer of the antisense oligonucleotide siRNA or ribozyme nucleic acid to the cells and preferably cells expressing CXCR2 receptor.
  • the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector.
  • the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the the antisense oligonucleotide siRNA or ribozyme nucleic acid sequences.
  • Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus; adenovirus, adeno-associated virus; SV40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and RNA virus such as a retrovirus.
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus
  • adenovirus adeno
  • Non-cytopathic viruses include retroviruses (e.g., lentivirus), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA.
  • Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replication- deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle).
  • retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo.
  • adeno-viruses and adeno- associated viruses are double-stranded DNA viruses that have already been approved for human use in gene therapy.
  • the adeno-associated virus can be engineered to be replication deficient and is capable of infecting a wide range of cell types and species. It further has advantages such as, heat and lipid solvent stability; high transduction frequencies in cells of diverse lineages, including hemopoietic cells; and lack of superinfection inhibition thus allowing multiple series of transductions.
  • the adeno-associated virus can integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection.
  • adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event.
  • the adeno-associated virus can also function in an extrachromosomal fashion.
  • Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g. Sambrook et al., 1989. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigen-encoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operatively encoded within the plasmid.
  • Plasmids may be delivered by a variety of parenteral, mucosal and topical routes.
  • the DNA plasmid can be injected by intramuscular, intradermal, subcutaneous, or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally.
  • the plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendhmers, cochleate and microencapsulation.
  • Another object of the invention relates to a method for treating or preventing insulin resistance comprising administering a subject in need thereof with a CXCR2 receptor antagonist or an inhibitor of CXCR2 receptor gene expression.
  • CXCR2 receptor antagonists or inhibitors of CXCR2 receptor gene expression may be administered in the form of a pharmaceutical composition, as defined below.
  • said antagonist or inhibitor is administered in a therapeutically effective amount.
  • a “therapeutically effective amount” is meant a sufficient amount of the CXCR2 receptor antagonist or inhibitor of CXCR2 receptor gene expression to treat or to prevent insulin resistance at a reasonable benefit/risk ratio applicable to any medical treatment.
  • the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed, the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidential with the specific polypeptide employed; and like factors well known in the medical arts.
  • the daily dosage of the products may be varied over a wide range from 0.01 to 1 ,000 mg per adult per day.
  • the compositions contain 0.01 , 0.05, 0.1 , 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from 1 mg to about 100 mg of the active ingredient.
  • An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
  • Inhibitors of the invention can be further identified by screening methods described in the state of the art.
  • the screening methods of the invention can be carried out according to known methods.
  • the screening method may measure the binding of a candidate compound to the receptor, or to cells or membranes bearing the receptor, or a fusion protein thereof by means of a label directly or indirectly associated with the candidate compound.
  • a screening method may involve measuring or, qualitatively or quantitatively, detecting the competition of binding of a candidate compound to the receptor with a labelled competitor (e.g., antagonist or agonist). Further, screening methods may test whether the candidate compound results in a signal generated by an antagonist of the receptor, using detection systems appropriate to cells bearing the receptor.
  • Antagonists can be assayed in the presence of a known agonist (e.g. CXCL5) and an effect on activation by the agonist by the presence of the candidate compound is observed.
  • a known agonist e.g. CXCL5
  • Competitive binding using known agonist such as CXCL5 is also suitable.
  • the CXCR2 receptor antagonist or inhibitor of CXCR2 receptor gene expression may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions.
  • the active principle in the pharmaceutical compositions of the present invention, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings.
  • Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms.
  • the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • saline solutions monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts
  • dry, especially freeze-dhed compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the CXCR2 receptor antagonist or inhibitor of CXCR2 receptor gene expression of the invention can be formulated into a composition in a neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • the carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active polypeptides in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed.
  • aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage could be dissolved in 1 ml of isotonic NaCI solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion. Some variation in dosage will necessarily occur depending on the condition of the subject being treated.
  • the person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
  • the CXCR2 receptor antagonist or inhibitor of CXCR2 receptor gene expression of the invention may be formulated within a therapeutic mixture to comprise about 0.0001 to 1.0 milligrams, or about 0.001 to 0.1 milligrams, or about
  • parenteral administration such as intravenous or intramuscular injection
  • other pharmaceutically acceptable forms include, e.g. tablets or other solids for oral administration; liposomal formulations; time release capsules; and any other form currently used.
  • Another aspect of the invention is a method for diagnosing and/or prognosing insulin resistance in a subject in need thereof, said method comprising detecting the presence and/or quantifying CXCL5 in a biological sample obtained from said subject.
  • the biological sample susceptible to contain CXCL5 is a biological sample, such as cell lysates (WAT, liver and muscle) or culture medium, or is a body fluid such as serum, plasma.
  • a biological sample such as cell lysates (WAT, liver and muscle) or culture medium, or is a body fluid such as serum, plasma.
  • said sample is WAT.
  • said sample is a serum sample or a plasma sample.
  • the detection and/or the quantification of CXCL5 is carried out by immunological detection.
  • the immunological detection or quantification of CXCL5 is achieved by any methods known in the art using at least one antibody that binds specifically to CXCL5 .
  • said methods include, but are not limited to, standard electrophoretic and immunodiagnostic techniques such as western blots, immuno- precipitation assay, radioimmunoassay, ELISA (enzyme-linked immunosorbant assay), "sandwich” immunoassay, gel diffusion precipitation reaction, immunodiffusion assay, precipitation reaction, agglutination assay (such as gel agglutination assay, hemagglutination assay, etc.), complement fixation assay, protein A assay, immunoelectrophoresis assay, high performance liquid chromatography, size exclusion chromatography, solid-phase affinity, etc.
  • standard electrophoretic and immunodiagnostic techniques such as western blots, immuno- precipitation assay, radioimmunoassay, ELISA (enzyme-linked immunosorbant assay), "
  • an antibody that binds specifically to CXCL5 is an antibody that does not cross react with others C-X-C chemokines.
  • Examples of said antibody include, but are not limited to, the antibody described in Arenberg et al., 1998; Halloran et al., 1999.
  • the antibody that binds to CXCL5 may be labelled with a detectable molecule or substance.
  • suitable labels for this purpose include a chemiluminescent agent, a colohmetric agent, an energy transfer agent, an enzyme, a substrate of an enzymatic reaction, a fluorescent agent, or a radioisotope.
  • the label may be coupled directly or indirectly by any known method in the art.
  • the detection or quantification of CXCL5 in a biological sample may be achieved by a competitive immunoassay.
  • competitive immunoassays include enzyme immunoassay or enzyme-linked immunoassay (EIA or ELISA), fluorescent immunoassay, radiometric or radioimmunoassay (RIA), magnetic separation assay (MSA), lateral flow assay, diffusion immunoassay, immunoprecipitation immunoassay.
  • the quantification of CXCL5 in a biological sample may be achieved by a non-competitive immunoassay referred as immunomethc, "two-site” or “sandwich” immunoassays, wherein the CXCL5 may be bound to or sandwiched between two antibodies that bind to CXCL5.
  • immunomethc non-competitive immunoassay
  • two-site or “sandwich” immunoassays
  • non-competitive immunoassays include enzyme immunoassay or enzyme-linked immunoassay (EIA or ELISA), fluorescent immunoassay, radiometric or radioimmunoassay (RIA), magnetic separation assay (MSA), lateral flow assay, diffusion immunoassay, immunoprecipitation immunoassay, immunosorbent or "antigen-down" assay using antibodies that bind to CXCL5 bound to a solid support, or agglutination assay.
  • EIA or ELISA enzyme immunoassay or enzyme-linked immunoassay
  • fluorescent immunoassay fluorescent immunoassay
  • RIA radiometric or radioimmunoassay
  • MSA magnetic separation assay
  • lateral flow assay diffusion immunoassay
  • immunoprecipitation immunoassay immunosorbent or "antigen-down” assay using antibodies that bind to CXCL5 bound to a solid support, or agglutination assay.
  • Examples of said assays are the human CXCL5 /ENA-78 DuoSet kits DY254 and DY443 purchased by R1 D Systems.
  • the quantification of CXCL5 in a biological sample is achieved by
  • the first antibody that binds to CXCL5 is an antibody that binds specifically to a first domain of CXCL5 and the second antibody is an antibody directed to an epitope in a second domain of CXCL5.
  • a one-step assay (simultaneous incubation of the two antibodies that bind to CXCL5) is useful.
  • a two-step assay (sequential incubation of the two antibodies that bind to CXCL5) is useful.
  • a two-step assay is preferred in the case where other molecules could compete for binding to the antibodies that bind to CXCL5.
  • Antibodies useful in the various embodiments of the invention encompass commercially available antibodies and antibody fragments, as well as any novel antibodies generated to bind to a suitable epitope on CXCL5.
  • the antibodies used in various embodiments exemplified herein are monoclonal or polyclonal in nature.
  • Other antibodies and antibody fragments, such as recombinant antibodies, chimeric antibodies, humanised antibodies, Fab or Fv fragments are also useful.
  • the amount of CXCL5 quantified may thus be compared with the corresponding amount detected in the samples of control subjects, in previous samples obtained from the subject or with normal reference values.
  • control subjects are for example subjects that have not been diagnosed for said insulin resistance.
  • Normal reference values refer to the amount of CXCL5 that can be determined by the method of the invention in a subject that has not been diagnosed for insulin resistance.
  • said control value or reference value is determined by using the average values obtained from at least 10, preferably from at least 100 control subjects.
  • said method for diagnosing and/or prognosing an insulin resistance in a subject in need thereof comprises the steps of:
  • FIGURES are a diagrammatic representation of FIGURES.
  • Quantification of mRNA was normalized by the expression level of rS9, here and in the following figures.
  • D-E Quantification of relative CXCL5 mRNA levels at the indicated times of 3T3-L1 (D) or human preadipocyte (E) differentiation.
  • F aP2 mRNA expression was analyzed by quantitative RT-PCR on 3T3L1 adipocytes differentiated with or without recombinant mouse CXCL5 (50ng/ml).
  • FIG. 2 Obesity increases CXCL5 release from WAT and CXCL5 systemic level:
  • C Circulating CXCL5 levels were measured by ELISA methods in serum from control C57BI/6 mice and mice fed with a high fat diet (HFD) for 12 weeks D.
  • HFD high fat diet
  • FIG. 3 CXCL5 is involved in insulin resistance: A. Comparative expression of CXCR2 mRNA in WAT, liver and muscle from 6-month-old C57BI/6 mice. B. 2-deoxy-glucose uptake in isolated soleus muscles from mice. Muscle strips were incubated with or without recombinant CXCL5 (100ng/ml) for 1 h before stimulation with insulin (10 "7 M). C. Protein expression of phospho-Akt, and total Akt in whole cells lysates from isolated muscles (upper panel) described in (B) and from primary MEFs (lower panel) as analyzed by Western blotting with anti-phospho- Ser473-Akt and anti-Akt antibodies. D.
  • E-H Glucose clearance after intraperitoneal injection of insulin (0.75U/kg) (E-F)) or glucose (G-H) in animals treated with anti-CXCL5 or control IgG. For the graph E, F and G, glucose values are relative to initial glucose levels. Area under the curve was analyzed (AUC).
  • Figure 5 Inhibition of CXCR2 signaling improves insulin sensitivity: A-B.
  • FIG. 6 CXCR2 -I- mice improves insulin sensitivity: A-B. Fasting glucose (A) and insulin (B) in 10-weeks-old CXCR2 -/- and control mice before
  • C Body weight in control and CXCR2 -/- mice in chow and high fat diet.
  • D Food intake in control and CXCR2 -/- mice.
  • E-F Glucose clearance after intraperitoneal injection of insulin (E) or glucose (F) in control and CXCR2 -/- mice under chow or high fat diet. Area under the curve was analyzed (AUC).
  • FIG. 7 CXCL5 expression is regulated by TNF ⁇ and rosiglitazone: A.
  • CXCL5 level in conditioned medium of human subcutaneous adipose tissue explants after treatment with vehicle (control) or TNF ⁇ (10ng/ml) for 24h (n 3 per group).
  • B. Quantification of relative CXCL5 mRNA levels in WAT from obese ob/ob TNF ⁇ receptor -/- mice compared to the control obese mice (n 3 per group) .
  • CXCL5 in medium was analyzed by ELISA and normalized by DNA tissue content.
  • E Q-PCR analysis of CXCL5 mRNA in THP- 1 macrophages treated as in (C).
  • F Luciferase activity expressed as relative luciferase units of a 1422 bp DNA fragment of the CXCL5 promoter coupled to the luciferase gene in the absence or in the presence of a PPARY expression vector in 293T cells treated with vehicle, TNF ⁇ (10ng/ml), rosiglitazone (10 "6 M) or both TNF ⁇ /rosiglitazone. Results were normalized to ⁇ galactosidase activity.
  • G
  • Figure 8 Comparison of mRNA levels in adipocytes, stromavascular fraction
  • Figure 9 Quantification of relative PPARY mRNA levels at the indicated times of 3T3-L1 preadipocyte differentiation.
  • FIG. 11 Rosiglitazone effect on diabetes: Fasting glucose level in db/db mice after 12 days treatment with 20mg/Kg rosiglitazone (rosi) compared to non- treated mice (control).
  • mice Male ob/ob mice, male db/db mice and male C57BL/6 mice were purchased at 8-9 weeks of age from Charles River laboratory. Male CXCR2 -/- mice were purchased at 9 weeks of age from Jackson Laboratory. Animals were maintained according to European Union guidelines for use of laboratory animals. In vivo experiments were performed in compliance with the French guidelines for experimental animal studies (Agreement No. B-34-172-27). For the diet-induced obesity model, 6-weeks-old C57BL/6 mice were fed ad libitum with a diet including 45% from fat (TestDiet) for 12 weeks. For antibody treatment, the neutralizing CXCL5 antibody we purchased from Dr.
  • mice were collected. The blood let to clot for at least 30 minutes before centhfugation for 10 minutes at 1000xg.
  • glucose tolerance test 18h-fasted mice were injected i.p. with glucose (1.7g/kg) and glycemia were measured at time 0, 30, 60 and 90 min after injection.
  • insulin tolerance test 8h- fasted mice were injected i.p. with insulin (0.75U/kg) and glucose level was measured at time 0, 20 and 40 min after injection.
  • Human studies Serum samples and subcutaneous abdominal WAT were obtained from subjects in agreement with French, Czech and English laws on biomedical research. Human adipocytes in primary culture were differentiated as described (Tiraby et al., 2003).
  • WAT explants culture Fat pads from male C57BL/6 mice and/or db/db mice were dissected under sterile conditions, washed in Krebs Ringer Bicarbonate Hepes buffer (KRBH), minced finely, and incubated in 12-well tissue culture plates containing KRBH supplemented with 1 % bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • CXCL5 concentration in cellular extracts, conditioned medium and serum was determined by ELISA with Duoset kit (DY254 and DY443, R&D Systems) as recommended by the manufacturer.
  • CXCL1 and CXCL2 concentrations in serum were evaluated by Bio-Plex Cytokine Assay (Bio- Rad) as recommended by the manufacturer.
  • RNA Extraction and Reverse Transcriptase Quantitative PCR: Total RNA from cells and tissues were isolated with TRIzol reagent (Invitrogen) as described by the manufacturer. Reverse transcription was performed using 5 ⁇ g total RNA, random primers and MMLV enzyme (Invitrogen). Quantitative PCR was performed with FastStart DNA Master SYBR Green I kit (Roche, Manheim, Germany) on a Light Cycler instrument (Roche) as specified by the manufacturer. Ribosomal protein S9 (rS9) was used as an internal control. The sequence of the primers used is available upon request.
  • Preadipocytes 3T3-L1 cells were grown in DMEM supplemented with 10% fcetal bovine serum (FBS), 100U/ml penicillin and 100 g/ml streptomycin. 3T3-L1 cells were differentiated as previously described (Sarruf et al., 2005). To analyze CXCL5 effect on adipocyte differentiation, 3T3-L1 preadipocytes were differentiated with mix supplemented with recombinant mouse CXCL5 (50ng/ml). Glucose transport experiments were performed as previously described (Abella et al., 2005).
  • Human monocytic cell line Thp-1 were grown in RPMI 1640 supplemented with 10% FBS, 100U/ml penicillin and 100 g/ml streptomycin. Macrophage differentiation was induced by stimulating cells with TPA 10OnM for 48h. Then, macrophages Thp-1 were maintained in DMEM 10% FBS and stimulated or not with TNF ⁇ (10ng/ml), rosiglitazone (10-6M) or co-treated with TNF ⁇ / rosiglitazone for 18h. CXCL5 mRNA level was analyzed by Q-PCR. Secreted CXCL5 was measured by ELISA assay and normalized by DNA content.
  • Fibroblastic 293T cells were grown in Dulbecco's Modified Eagle's Medium (DMEM) supplemented with 10% FBS, 100U/ml penicillin and 100 g/ml streptomycin. Transient transfections were performed as described previously (Annicotte et al., 2005) and luciferase activity measurements were normalized for ⁇ -galactosidase activity to correct for differences in transfection efficiency. Graph values represent the mean of three independent experiments.
  • DMEM Dulbecco's Modified Eagle's Medium
  • MEFs Primary MEFs were obtained from embryos at day 13.5 as previously described (Abella et al., 2005) and maintained in DMEM supplemented with 25mM glucose and 10% FBS. MEFs were starved 1 h in DMEM containing 0.2% BSA and then treated 30min with Jak2 inhibitor AG490 (50 ⁇ M) or CXCR2 antagonist SB225002 (10-6M) before to stimulate 1 h with CXCL5 (300ng/ml). For insulin signaling, cells were stimulated with insulin (10-9M) for 10min.
  • Jak2 inhibitor AG490 50 ⁇ M
  • CXCR2 antagonist SB225002 10-6M
  • Chromatin immunoprecipitation ChIP assays were performed as described previously (Annicotte et al., 2006). Briefly, proteins from Thp-1 cells were formaldehyde cross-linked to DNA. After homogenization, lysis and DNA sonication, proteins were then immunoprecipitated using anti-p65 antibody. After washing, DNA- protein-complexes were subsequently eluted and cross-linking was reversed by heating the samples at 65°C for 16 h. DNA was then purified using Qiagen PCR purification kit (Qiagen, Courtabceuf, France) and PCR amplification was performed using cxcl ⁇ promoter-specific oligonuleotide primers.
  • Qiagen PCR purification kit Qiagen, Courtabceuf, France
  • CXCL5 promoter corresponding to -1359/+63 was cloned from breast cancer cell line MCF-7 using 5'-TCA GAA CCA GCC AGA AGA GGA-3' and 5'-AGC GGA GAT TGG AGG AGC GAA GAT-3' and subcloned in xp-2 luciferase reporter vector.
  • CMV-GAL corresponds to the ⁇ -galactosidase gene under the control of the cytomegalovirus (CMV) promoter.
  • CXCL5 is expressed by WAT resident macrophages: Global analysis of cytokine expression in insulin-sensitive tissues highlighted the presence of high levels of CXCL5 protein expression in WAT in mice, compared to liver or muscle ( Figure 1A), suggesting that CXCL5 could be a new adipocytokine. Consistent with this hypothesis, CXCL5 was found to be secreted by WAT explants in culture ( Figure 1 B). Furthermore, fractionation of human subcutaneous WAT by collagenase digestion showed that the stromal vascular fraction (SVF) expressed higher levels of CXCL5 than did mature adipocytes, as assessed by real-time PCR analysis (Figure 1 C).
  • SVF stromal vascular fraction
  • CXC L5 expression and secretion is increased in obesity:
  • the inflammatory state associated with obesity includes macrophage infiltration in WAT, and increased cytokine secretion by these macrophages. Since it is expressed in WAT macrophages ( Figure 1 C), we hypothesized that CXCL5 expression would be increased in obesity. Consistent with this hypothesis CXCL5 secretion in WAT explants from obese db/db mice was markedly increased compared to non-obese control mice (2.5-fold, Figure 2A). Most interestingly, circulating serum CXCL5 concentration was increased 2-fold in obese ob/ob and db/db and high fat diet-fed mice compared to lean mice ( Figures 2B and 2C).
  • CXCL5 mediates insulin resistance: Some of the insulin desensitizing effects observed during obesity development are mediated by adipokines. The positive association of CXCL5 concentration with obesity suggested that this adipokine could be a mediator of insulin resistance. The biological effects of CXCL5 are mediated through interaction with the chemokine receptor CXCR2. To investigate the role of CXCL5 in insulin resistance, we first aimed to identify tissues that are potentially responsive to CXCL5. Expression analysis in insulin-sensitive tissues showed that CXCR2 mRNA was expressed at higher levels in muscle than in liver or WAT ( Figure 3A). The physiological effects of CXCL5 in muscle were next analyzed.
  • Obese db/db mice were treated with the insulin- sensitizing drug rosiglitazone. This resulted, as expected in a decrease in glycemia ( Figure 11 ). Decreased circulating glucose concentration was significantly associated with a 30% decrease in serum CXCL5 concentration in rosiglitazone-treated mice ( Figure 3F).
  • Anti-CXCL5-treated obese mice were more sensitive to insulin as measured by fasted insulin level ( Figures 4C and 4D) and by insulin tolerance test ( Figures 4E and 4F). Moreover, treatment with anti-CXCL5 improved glucose tolerance during an intraperitoneal glucose tolerance test in both mouse models of obesity ( Figure 4G and 4H).
  • CXCL5 is regulated by TNF ⁇ :
  • TNF ⁇ is a key factor regulating CXCL5 level in WAT since we demonstrated that CXL5 mRNA expression is strongly decreased in WAT from obese ob/ob TNF ⁇ receptor -/- mice compared to the ob/ob mice ( Figure 7B).
  • chromatin immunoprecipitation (ChIP) studies were performed using a specific anti-p65 antibody or preimmune serum in THP-1 cells. No amplification of the CXCL5 promoter was observed when non-specific antibodies were used to immunoprecipitate the chromatin (IgG, Figure 7G), whereas a specific fragment corresponding to the NFKB binding site of the CXCL5 promoter was amplified when chromatin of TNF ⁇ -treated cells was immunoprecipitated with an anti- NFkB antibody ( Figure 7G).
  • the nuclear receptor liver receptor homolog-1 is an estrogen receptor target gene. Oncogene 24, 8167- 8175.
  • Epithelial-neutrophil activating peptide (ENA-78) is an important angiogenic factor in non-small cell lung cancer. J Clin Invest 102, 465-472.
  • Cyclin D3 promotes adipogenesis through activation of peroxisome proliferator- activated receptor gamma. MoI Cell Biol 25, 9985-9995.

Abstract

La présente invention porte sur des antagonistes du récepteur CXCR2 ou des inhibiteurs de l'expression génique du récepteur CXCR2 pour le traitement ou la prévention de la résistance à l'insuline. La présente invention porte également sur un procédé destiné à diagnostiquer et/ou à pronostiquer une résistance à l'insuline chez un sujet, comprenant la détection de la présence et/ou la quantification du CXCL5 dans un échantillon biologique obtenu à partir dudit sujet.
PCT/IB2009/052312 2009-02-16 2009-02-16 Antagonistes du récepteur cxcr2 pour le traitement ou la prévention de la résistance à l'insuline WO2010092440A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/IB2009/052312 WO2010092440A1 (fr) 2009-02-16 2009-02-16 Antagonistes du récepteur cxcr2 pour le traitement ou la prévention de la résistance à l'insuline

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/IB2009/052312 WO2010092440A1 (fr) 2009-02-16 2009-02-16 Antagonistes du récepteur cxcr2 pour le traitement ou la prévention de la résistance à l'insuline

Publications (1)

Publication Number Publication Date
WO2010092440A1 true WO2010092440A1 (fr) 2010-08-19

Family

ID=41020972

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2009/052312 WO2010092440A1 (fr) 2009-02-16 2009-02-16 Antagonistes du récepteur cxcr2 pour le traitement ou la prévention de la résistance à l'insuline

Country Status (1)

Country Link
WO (1) WO2010092440A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014138452A1 (fr) * 2013-03-08 2014-09-12 Allergan, Inc. Inhibiteurs des kinases
WO2018010856A1 (fr) * 2016-07-13 2018-01-18 Universitat De Barcelona Activateurs de hri utiles pour le traitement de maladies cardiométaboliques
CN114042158A (zh) * 2021-10-11 2022-02-15 中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所) Cxcl5抑制剂在制备治疗肿瘤的药物中的应用
TWI774059B (zh) * 2020-09-14 2022-08-11 國立陽明大學 Cxcl5中和抗體用於製備預防或治療周邊動脈阻塞疾病的藥物的用途

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997029743A1 (fr) * 1995-02-17 1997-08-21 Smithkline Beecham Corporation Antagonistes de recepteurs de l'interleukine-8
EP1088819A2 (fr) * 1999-09-30 2001-04-04 Pfizer Products Inc. Dérivés de 6-azauracil utiles en tant que ligands du récepteur thyroid
WO2004058683A2 (fr) * 2002-12-20 2004-07-15 Migenix Corp. Ligands d'adenine nucleotide translocase (ant) et compositions et methodes associees
WO2005019220A2 (fr) * 2003-08-11 2005-03-03 Cellular Genomics Inc. Imidazo[1,2-a]pyrazines substituees utilisees comme modulateurs de l'activite d'une kinase
WO2005023238A1 (fr) * 2003-09-04 2005-03-17 Poseidon Pharmaceuticals A/S Ouvreurs du canal erg destines au traitement de maladies neuronales liees a l'hyperexcitabilite
WO2006052722A1 (fr) * 2004-11-09 2006-05-18 Smithkline Beecham Corporation Composes inhibiteurs de la phosphorylase du glycogene et compositions pharmaceutiques les comprenant
EP1676573A1 (fr) * 2004-12-30 2006-07-05 Laboratorios Del Dr. Esteve, S.A. Composition pharmaceutique comprenant un composé 2,5-dihydroxybenzensulfonique, un modulateur des canaux de potassium et un inhibiteur de la phosphodiesterase 5
WO2008074756A1 (fr) * 2006-12-18 2008-06-26 Neurosearch A/S Nouveaux dérivés thio-urée biphényliques convenant comme modulateurs du canal potassium

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997029743A1 (fr) * 1995-02-17 1997-08-21 Smithkline Beecham Corporation Antagonistes de recepteurs de l'interleukine-8
EP1088819A2 (fr) * 1999-09-30 2001-04-04 Pfizer Products Inc. Dérivés de 6-azauracil utiles en tant que ligands du récepteur thyroid
WO2004058683A2 (fr) * 2002-12-20 2004-07-15 Migenix Corp. Ligands d'adenine nucleotide translocase (ant) et compositions et methodes associees
WO2005019220A2 (fr) * 2003-08-11 2005-03-03 Cellular Genomics Inc. Imidazo[1,2-a]pyrazines substituees utilisees comme modulateurs de l'activite d'une kinase
WO2005023238A1 (fr) * 2003-09-04 2005-03-17 Poseidon Pharmaceuticals A/S Ouvreurs du canal erg destines au traitement de maladies neuronales liees a l'hyperexcitabilite
WO2006052722A1 (fr) * 2004-11-09 2006-05-18 Smithkline Beecham Corporation Composes inhibiteurs de la phosphorylase du glycogene et compositions pharmaceutiques les comprenant
EP1676573A1 (fr) * 2004-12-30 2006-07-05 Laboratorios Del Dr. Esteve, S.A. Composition pharmaceutique comprenant un composé 2,5-dihydroxybenzensulfonique, un modulateur des canaux de potassium et un inhibiteur de la phosphodiesterase 5
WO2008074756A1 (fr) * 2006-12-18 2008-06-26 Neurosearch A/S Nouveaux dérivés thio-urée biphényliques convenant comme modulateurs du canal potassium

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHAVEY C ET AL: "CXC Ligand 5 Is an Adipose-Tissue Derived Factor that Links Obesity to Insulin Resistance", CELL METABOLISM, CELL PRESS, CAMBRIDGE, MA, US, vol. 9, no. 4, 8 April 2009 (2009-04-08), pages 339 - 349, XP007909843, ISSN: 1550-4131 *
LIAO ET AL: "Inhibition of CXCR2 Expression in HL60 Cells by CXCR2 Antisense RNA", CLINICAL IMMUNOLOGY, ACADEMIC PRESS, US, vol. 123, 1 January 2007 (2007-01-01), pages S181 - S182, XP022077144, ISSN: 1521-6616 *
NEELS JAAP G ET AL: "KERATINOCYTE-DERIVED CHEMOKINE IN OBESITY: EXPRESSION, REGULATION, AND ROLE IN ADIPOSE MACROPHAGE INFILTRATION AND GLUCOSE HOMEOSTASIS", JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY OF BIOLOCHEMICAL BIOLOGISTS, BIRMINGHAM, US, vol. 284, no. 31, 3 June 2009 (2009-06-03), pages 20692 - 20698, XP007909844, ISSN: 0021-9258 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014138452A1 (fr) * 2013-03-08 2014-09-12 Allergan, Inc. Inhibiteurs des kinases
US9321721B2 (en) 2013-03-08 2016-04-26 Allergan, Inc. Kinase inhibitors
WO2018010856A1 (fr) * 2016-07-13 2018-01-18 Universitat De Barcelona Activateurs de hri utiles pour le traitement de maladies cardiométaboliques
TWI774059B (zh) * 2020-09-14 2022-08-11 國立陽明大學 Cxcl5中和抗體用於製備預防或治療周邊動脈阻塞疾病的藥物的用途
CN114042158A (zh) * 2021-10-11 2022-02-15 中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所) Cxcl5抑制剂在制备治疗肿瘤的药物中的应用
CN114042158B (zh) * 2021-10-11 2023-03-03 中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所) Cxcl5抑制剂在制备治疗肿瘤的药物中的应用

Similar Documents

Publication Publication Date Title
Chavey et al. CXC ligand 5 is an adipose-tissue derived factor that links obesity to insulin resistance
Pedroza et al. STAT-3 contributes to pulmonary fibrosis through epithelial injury and fibroblast-myofibroblast differentiation
Hong et al. Hepatic stellate cells express functional CXCR4: Role in stromal cell–derived factor‐1α–mediated stellate cell activation
Chen et al. The novel role of IL-7 ligation to IL-7 receptor in myeloid cells of rheumatoid arthritis and collagen-induced arthritis
Zhou et al. Interleukin-1β and tumor necrosis factor-α augment acidosis-induced rat articular chondrocyte apoptosis via nuclear factor-kappaB-dependent upregulation of ASIC1a channel
Song et al. DEK‐targeting aptamer DTA‐64 attenuates bronchial EMT‐mediated airway remodelling by suppressing TGF‐β1/Smad, MAPK and PI3K signalling pathway in asthma
MXPA06013483A (es) Modulacion de produccion de inmunoglobulina y trastornos atopicos.
JP6538138B2 (ja) Herv−wエンベロープタンパク質発現関連疾患において再ミエリン化遮断を治療するための化合物
Warren et al. Tumor necrosis factor deficiency inhibits mammary tumorigenesis and a tumor necrosis factor neutralizing antibody decreases mammary tumor growth in neu/erbB2 transgenic mice
Qiu et al. Identification and characterization of a novel adiponectin receptor agonist adipo anti‐inflammation agonist and its anti‐inflammatory effects in vitro and in vivo
Yoon et al. Activation of the adipocyte CREB/CRTC pathway in obesity
JP5274458B2 (ja) 黒色腫における治療応答のための早期指標としての黒色腫阻害性活性(mia)タンパク質の使用
Hsu et al. IL‐20 is involved in obesity by modulation of adipogenesis and macrophage dysregulation
WO2010092440A1 (fr) Antagonistes du récepteur cxcr2 pour le traitement ou la prévention de la résistance à l&#39;insuline
KR101539138B1 (ko) 병용 요법 및 치료에 대한 내성 평가 방법
Jin et al. Babam2 negatively regulates osteoclastogenesis by interacting with Hey1 to inhibit Nfatc1 transcription
Zhang et al. IGFBP3 induced by the TGF-β/EGFRvIII transactivation contributes to the malignant phenotype of glioblastoma
Raices et al. A novel role for IκBζ in the regulation of IFNγ production
EP3452512B1 (fr) Procédés et compositions pharmaceutiques pour le traitement de lésion de tissu
US8748493B2 (en) Inhibitors of cathepsin S for prevention or treatment of obesity-associated disorders
WO2023080157A1 (fr) Agent thérapeutique ou prophylactique pour myélopathie associée à htlv-1 (ham), procédé de détermination d&#39;efficacité thérapeutique, et procédé d&#39;évaluation d&#39;activité ou procédé de diagnostic
US20230075368A1 (en) Identification of an egfr-bin3 pathway that actively suppresses invasion and reduces tumor size in glioblastoma
JP2010502985A (ja) インスリン感受性の制御におけるリポカリン2の使用
US20130236480A1 (en) Transglutaminase 2 inhibitors for use in the prevention or treatment of rapidly progressive glomerulonephritis
Zhihua et al. CXCL14 Accelerates Fibroblast Ferroptosis in Inflammatory Bowel Disease by Regulating Lipid Metabolism via SCD1

Legal Events

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

Ref document number: 09786412

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09786412

Country of ref document: EP

Kind code of ref document: A1