NZ617454B2 - Molecule for treating an inflammatory disorder - Google Patents

Molecule for treating an inflammatory disorder Download PDF

Info

Publication number
NZ617454B2
NZ617454B2 NZ617454A NZ61745412A NZ617454B2 NZ 617454 B2 NZ617454 B2 NZ 617454B2 NZ 617454 A NZ617454 A NZ 617454A NZ 61745412 A NZ61745412 A NZ 61745412A NZ 617454 B2 NZ617454 B2 NZ 617454B2
Authority
NZ
New Zealand
Prior art keywords
seq
nucleic acid
acid molecule
sequence
inflammatory
Prior art date
Application number
NZ617454A
Other versions
NZ617454A (en
Inventor
Bedate Carlos Alonso
Sanchez Jeronimo Carnes
Garcia Laura Ramirez
Escutia Marta Roman
Alvarez Manuel Soto
Original Assignee
Laboratorios Leti Sl
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 Laboratorios Leti Sl filed Critical Laboratorios Leti Sl
Priority claimed from PCT/EP2012/058453 external-priority patent/WO2012152792A1/en
Publication of NZ617454A publication Critical patent/NZ617454A/en
Publication of NZ617454B2 publication Critical patent/NZ617454B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/002Protozoa antigens
    • A61K39/008Leishmania antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • 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
    • C07K14/44Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from protozoa
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/24Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a MBP (maltose binding protein)-tag
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

Discloses a nucleic acid molecule for use as a medicament, wherein said nucleic acid molecule is represented by a nucleotide sequence selected from the group consisting of: i. nucleotide sequences encoding a polypeptide comprising an amino acid sequence that has at least 50% sequence identity with the amino acid sequence of SEQ ID NO:1, ii. nucleotide sequences comprising a nucleotide sequence that has at least 50% sequence identity with the nucleotide sequence of SEQ ID NO:2, iii. nucleotide sequences the complementary strand of which hybridises to a nucleic acid molecule of sequence of (i) or (ii) and iv. nucleotide sequences the sequences of which differs from the sequence of a nucleic acid molecule of (iii) due to the degeneracy of the genetic code; wherein the sequences are as defined in the complete specification. the amino acid sequence of SEQ ID NO:1, ii. nucleotide sequences comprising a nucleotide sequence that has at least 50% sequence identity with the nucleotide sequence of SEQ ID NO:2, iii. nucleotide sequences the complementary strand of which hybridises to a nucleic acid molecule of sequence of (i) or (ii) and iv. nucleotide sequences the sequences of which differs from the sequence of a nucleic acid molecule of (iii) due to the degeneracy of the genetic code; wherein the sequences are as defined in the complete specification.

Description

Molecule for treating an inflammatory disorder Field of the invention The invention provides a L19 source as a medicament, preferably for ting or treating an inflammatory disorder in an individual.
Background of the invention Immune and related inflammatory diseases are a manifestation of complex, frequently interconnected biological pathways which in normal physiology respond to insult or injury by initiating repair of the insult or injury, and mount an innate and acquired response. Disease or pathology occurs when these logical pathways cause further insult or injury, either by an exaggerated response caused by an abnormal regulation or overstimulation, or a combination ofthe two. Despite the advent of new nflammatory drugs such as anti-TNF agents, inflammatory diseases continue to ent an important unmet medical need, often due to a lack of responsiveness and resistance to these drugs.
Immune and related inflammatory es that may be modulated by the use of anti- inflammatory agents include Autoimmune Diabetes (any others similar), diabetes mellitus, uveitis, (1) Multiple Sclerosis, Rheumatoid Arthritis (RA), Irritable Bowel Disease (IBD), Irritable Bowel syndrome, ulcerative colitis, s disease, lling Allograft Rejection after organ transplantation, graft versus host disease (GVHD), inflammatory lung diseases including asthma and chronic obstructive pulmonary disease (COPD) (2), cancer (4) systemic lupus erythematosus, SLE, sarcoidosis, cancer and Psoriasis.
RA is considered a systemic autoimmune disease, managed by treatment with Disease- modifying anti-rheumatic drugs (DMARDS), typically in combination, to minimize the side s associated with systemic drugs. Side effects of these drugs include ulcerative stomatitis, reduced white blood count IBD is a term that describes c inflammation disorder of the small and/0r large intestine.
Included within the area of IBD is ulcerative colitis and Crohn's e. While the exact causes are not firmly established, IBD is ered to be an autoimmune disease. Currently no cure is ble, and treatments are focused on suppressing the abnormal or exaggerated inflammatory response. Treatments e corticosteroids (such as methotrexate, WO 52792 azathioprine, and mercaptopurine) and aminosalicylates. Long term use of corticosteroids are associated with ng of the bones, infection, cataracts, and love and bone marrow effects.
Aminosalicylates tend to be better tolerated, since they are poorly absorbed, and act on the affected area topically. Side effects include headache, and rarely more serious conditions, such as atitis.
Psoriasis is treated in different ways. Use of corticosteroids topically is a common method of treatment, but drawbacks include ineffectiveness and development of resistance. Use of phototherapy is effective in treating psoriasis by increasing apoptosis, implicated in reduced inflammation. Short term cks are increased discomfort, and itching, with long term effects being an increased risk of squamous cell and melanoma skin cancers. Systemic drugs are utilised to treat psoriasis, which have a variety of other, often red systemic effects and must be used under close supervision and monitoring by a dermatologist.
Therefore there is still a need to design new treatments for an inflammatory disease such as RA, IBD, and psoriasis which do not have all the drawbacks of existing treatments.
Description of the invention L19 source In a first aspect, there is provided a L19 source for use as a medicament.
L19 is a ribosomal protein. Ribosomal ns are well conserved cytosolic proteins.
Therefore, a L19 source may be prepared from any eukaryotic organism, be it plant or , be it from mammals, es, fish, insects, or any other chromosome g organism, such as protozoa. The invention is not limited to a specific L19 source as long as the encoded L19 n product is able to induce an anti-inflammatory response as later defined herein. Preferred protozoans include plasmodium and in particular members of the trypanosomatid family, more in particular different species of the trypanosomatical protozoan Leishmam'a. There are over 20 known species of Leishmania, ing species of the subgenus Leishmam'a, comprising the x L. major, including L. major, the complex L.
Donovam', including L. chagasz', L. donovani and L. infantum, the complex L. Mexicana, including L. amazonensz's and L. mexz'cana, as well as the subspecies Vianm'a, comprising the x L. brazilz'ensz's, ing L. brazilz'ensz's and L. peruvz’ana and the complex L. guyanensz's, including L. guyanensz’s and L. panamensz’s. Plasmodz'um species of particular interest are Plasmodz'um falcz'parum and Plasmodium vivax. Alternatively a L19 source may be obtained from a Trypanosoma species. A Trypanosoma species may be a Trypanosoma cruzz', a Trypanosoma brucez’. In a preferred embodiment, a L19 source is ed from or d from or originated from a Leishmam’a species, preferably Leishmania major, Leishmam'a infantum Leishmania donovani, Leishmam'a chagasz' and/or Leishmam'a brazilz'ensz's. More preferred is a L19 source which is obtained from or derived from or ated from ania major. The skilled person will understand that a source of L19 may also be prepared by mixing two or more L19 sources derived from the same organism or from several distinct organisms as identified herein. The use of an L19 source has been demonstrated herein to have attractive properties since it has been shown that the encoded L19 n product is able to induce the production of an nflammatory response in a treated subject.
A preferred L19 source is a nucleic acid molecule, an oligonucleotide, a protein, a protein fragment and/or a peptide each being derived from a L19 protein or polypeptide or nucleic acid molecule as defined herein. A L19 source preferably comprises or consists of a L19 protein, a L19 ptide, a L19 derived peptide or a L19 n fragment and/or a c acid le encoding a L19 protein or L19 polypeptide or L19 derived peptide or L19 protein fragment, each has defined herein. A preferred L19 protein is represented by SEQ ID NO: 1. This preferred L19 protein is preferably encoded by SEQ ID NO:2. Another preferred L19 protein is represented by SEQ ID NO: 5, 7, 3,15, 17,19, 21, 23, 25, 27 or 29.
Each of these other L19 proteins is preferably d by SEQ ID NO: 6, 8, 10, l2, l4, 16, 18, 20, 22, 24, 26, 28, 30 respectively.
In a first embodiment, a preferred L19 source is a nucleic acid molecule represented by a nucleotide sequence selected from the group consisting of: i. nucleotide ces encoding a polypeptide comprising an amino acid sequence that has at least 50% sequence identity or similarity with the amino acid sequence of SEQ ID NO:1, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23 ,25, 27 or 29, ii. nucleotide sequences comprising a nucleotide sequence that has at least 50% sequence identity or similarity with the nucleotide sequence of SEQ ID N02, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or 30, iii. nucleotide ces the complementary strand of which hybridizes to a nucleic acid molecule of sequence of (i) or (ii) and iv. nucleotide sequences the sequences of which differs from the sequence of a nucleic acid molecule of (iii) due to the degeneracy of the genetic code.
In a second embodiment, a preferred L19 source is a polypeptide encoded by a nucleic acid molecule ofthe first embodiment as fied above. In a more preferred embodiment, a L19 source is a polypeptide whose amino acid sequence has at least 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity or similarity with a polypeptide having amino acid sequence SEQ ID NO:1, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23 ,25, 27, 29 or 31.
We identified several L19 proteins and corresponding encoding nucleic acid molecules. Each of these L19 proteins comprises an amino acid sequence having at least 60%, 70%, 80%, 90%, 95% or more with SEQ ID NO:1. Each of the nucleic acid molecule encoding each of these L19 proteins ses a nucleotide sequence having at least 60%, 70%, 80%, 90%, 95% or more with SEQ ID NO:2. Each of these L19 proteins represents a homologue of Leishmania major L19 protein as represented by SEQ ID NO:1 Briefly, we identified three L19 proteins from Leishmam’a brazilz'ensz's, being represented by SEQ ID NO: 5, 7 or 9. Each of these proteins is preferably encoded by the following nucleotide sequence SEQ ID NO: 6, 8 or 10 respectively.
We also identified two L19 proteins from am’a infantum, being represented by SEQ ID NO: 11 or 13. Each of these proteins is preferably d by the following nucleotide sequence SEQ ID NO: 12 or 14 tively.
We also identified two L19 proteins from Leishmam’a mexz'cana, being represented by SEQ ID NO: 15 or 17. Each of these proteins is preferably d by the following nucleotide sequence SEQ ID NO: 16 or 18 tively.
We also identified one L19 protein from Leishmania donovani, being represented by SEQ ID NO: 19. This n is preferably d by the following nucleotide sequence SEQ ID NO:20.
In addition, we identified four L19 proteins from Trypanosoma cruzz', being represented by SEQ ID NO: 21, 23, 25 or 27. Each of these proteins is ably encoded by the following nucleotide sequence SEQ ID NO: 22, 24, 26 or 28 respectively.
We also identified one L19 protein from Trypanosoma brucez’, being represented by SEQ ID NO: 29. This protein is preferably encoded by the following nucleotide sequence SEQ ID NO:30.
Preferably, said amino acid sequence or nucleotide sequence as defined herein having at least 50% identity or similarity with a specific identified amino acid or nucleotide sequence are encompassed by the present invention and are said to be fianctional when the encoded protein polypeptide, protein nt or peptide is capable of inducing an nflammatory response as obtainable by the L19 protein represented by SEQ ID NO:1, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23 ,25, 27 or 29 to at least some extent. To at least some extent preferably means that at least 50%, at least 60%, 70%, 80%, at least 90% or 100% of the anti-inflammatory response induced by SEQ ID NO:1, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27 or 29.
Inducing an anti-inflammatory response is or is preferably defined as the ability to induce a detectable production of an anti-inflammatory compound and/or the ability to induce a decrease of the tion of an inflammatory compound in a treated t or individual.
An anti-inflammatory compound is preferably a cytokine. More red cytokine is IL-10.
An inflammatory compound is preferably a cytokine. More preferred cytokine IFNy and/or TNFOL. The tion of IL-10 or IFNy or TNFOL is preferably assessed at the mRNA level using PCR or at the protein level using ELISA., an ELISPOT or FACS. All these techniques are known to the skilled person. Many ations have implicated the elevation of IL-10 with a reduction in inflammation, as a result of disease. The same holds with the elevation of IFNy or TNFOL and the presence of inflammation. The production of an nflammatory nd may be assessed on a treated subject or on a sample obtained from said subject. In this context, a sample may be a tissue or a fluid or a cell. Preferred tissue includes spleen or skin or intestine or lung. Preferred fluid includes blood. Preferred cells include a PBMC (Peripheral Blood Mononuclear Cell) or skin cells or intestinal cells or lung cells. An anti- inflammatory response may be induced after at least 1, 2, 3, 4, 5, 6, 7 days of treatment with a L19 source. Preferred L19 sources are a L19 polypeptide, n, protein fragment or peptide. The induction of an anti-inflammatory response may also be an increase of the induction of an nflammatory response. In this context, an “increase” may mean an increase of at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%. The ion of an anti-inflammatory response may also be the decrease of the amount or quantity of IFNy and/or TNFOL. In this context, a “decrease” may mean a decrease of at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, %, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%.
In a preferred embodiment, an anti-inflammatory compound is produced and no detectable inflammatory compound (i.e. IFNy and/or TNFOL) are detected. In this t, no IFN— y and/or no TNFOL is ed. The e of TNFOL and/or IFN—y is ably assessed using PCR or an ELISA. The absence of an inflammatory compound may be assessed on a treated subject or on a sample obtained from said subject as for the anti-inflammatory compound.
In a preferred assay, an anti-inflammatory response, more preferably the production of IL-10 or an increase of IL-10 is detected after at least 24 hours or 48 hours or 72 hours of incubation of a L19 source, preferably a L19 polypeptide or a L19 peptide with a PBMC. In this preferred assay, a decreased amount of IFN—gamma and/or a decreased amount of TNFOL, or no detectable IFN—y and/or no TNFOL is detected after at least 24 hours or 48 hours or 72 hours of incubation of a L19 source, preferably a L19 polypeptide or a L19 peptide with a PBMC. More preferably, IL-10, INFy and/or TNFOL is assessed by ELISA as described in the experimental part. In a fiarther red embodiment, a L19 source which is able to induce an anti-inflammatory response is also able to prevent and/or delay the development of an inflammatory disorder or condition or disease and/or is able to alleviate one or more symptom(s) and/or one or more characteristic(s) or parameter(s) of a cell or tissue from a treated subject as later defined herein.
A red L19 source is a nucleic acid molecule of the first ment as identified above. This preferred nucleic acid molecule is represented by a nucleotide sequence which is derived from SEQ ID N02, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or 30, or a sequence having at least 50% identity or similarity with SEQ ID N02, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or 30 or with a part thereof and that may comprise substitutions, insertions, deletions and additional or 3’ terminal nucleotides or chemical es to increase stability, solubility or targeting. In a red embodiment, a L19 source is a nucleic acid molecule whose nucleotide sequence has at least 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity or WO 52792 similarity with SEQ ID NO:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or 30 or with a part thereof A L19 nucleic acid le as defined herein is preferably an oligonucleotide. A preferred oligonucleotide has a length of at least 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 nucleotides and is derived from SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18, , 22, 24, 26, 28 or 30. More preferred oligonucleotides comprise at least 8, 10, 15, 20, 25, , 35, 40,45, 50, 55, 60,65, 70, 75, 80, 85, 90, 95, 100 or more contiguous nucleotides ofa corresponding L19 nucleic acid molecule as identified above, preferably represented by SEQ ID N02, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or 30 and whose encoded product is able to induce an anti-inflammatory response as earlier defined herein. In a preferred embodiment, therefore, a L19 nucleic acid molecule as defined herein is ably an oligonucleotide comprising at least 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more contiguous nucleotides of SEQ ID N02, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or 30.
Accordingly a preferred L19 source is an oligonucleotide comprising at least 8, 10, 15, 20, , 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more contiguous nucleotides of SEQ ID NO:2.
Another preferred L19 source is a polypeptide encoded by a nucleic acid molecule of the first ment as fied above and/or is a polypeptide whose amino acid sequence has at least 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity or similarity with a polypeptide having amino acid sequence SEQ ID NO:1, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23 ,25, 27 or 29 or with a part thereof A preferred polypeptide is represented by an amino acid ce which is derived from SEQ ID NO:1, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23 ,25, 27 or 29 or from a part thereofor a sequence having at least 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity or similarity with SEQ ID NO:1, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23 ,25, 27 or 29 or with a part thereof and that may comprise substitutions, ions, deletions and additional N— or C- terminal amino acids or chemical moieties to increase stability, solubility.
A L19 protein nt or a L19 derived peptide or a L19 polypeptide or a L19 protein as defined herein is preferably a fragment comprising at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 265, or 267 contiguous amino acids of a corresponding L19 protein, preferably represented by SEQ ID N021, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23 ,25, 27 or 29 and which is able to induce an anti- inflammatory response as earlier defined herein. In a preferred embodiment, therefore, a L19 protein fragment or a L19 d e as defined herein is ably a fragment comprising at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 265, or 267 contiguous amino acids of SEQ ID N021, 5, 7, 9,11,13,15,17,19, 21, 23 ,25, 27 or 29. A L19 source may also comprise a full length L19 protein such as the one represented by SEQ ID N021, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23 ,25, 27 or 29 and comprises additional amino acids at the N— and/or inus of the L19 protein. In another preferred embodiment, a L19 source comprises or consists of a protein or a polypeptide comprising at least one protein fragment of a L19 protein. A preferred L19 source is a protein fragment comprising at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 265, or 267 contiguous amino acids of SEQ ID NO:1.
In an embodiment, a source of L19 is a peptide derived from SEQ ID NO:1 or a fragment of SEQ ID NO:1. A red fragment or peptide comprises at least 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42 uous amino acids of SEQ ID NO:1. In example 3, three regions of L19 and specific peptides derived from L19 have been identified as being able of inducing the tion of IL-10. The preferred regions of L19 are the following: Region 1 comprises peptides having SEQ ID NO: 31, 32 and/or 55, Region 2 comprises peptides having SEQ ID NO: 42, 43, 44 and/or 56, Region 3 comprises peptides having SEQ ID NO: 53, 54 and/or 57 Below we define in more details these peptides or fragments of SEQ ID NO:1. A protein fragment of SEQ ID NO:l comprising at least 14 contiguous amino acids of SEQ ID NO:l and comprising SEQ ID NO: 31, 32, 55, 42, 43, 44, 56, 53, 54 and/or 57.
A more preferred fragment of SEQ ID NO:l comprises SEQ ID NO:3l or 32 or 42 or 43 or 44 or 53 and comprises up to 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21 or 20 contiguous amino acids from SEQ ID NO:l. Said fragment may se SEQ ID NO:3l or 32 or 42 or 43 or 44 or 53 and may have a length of up to 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21 or 20 amino acids. Said fragment preferably consists of SEQ ID NO:3l or 32 or 42 or 43 or 44 or 53.
Another more preferred fragment of SEQ ID NO:l comprises SEQ ID NO: 54 and comprises up to 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28,27, 26, 25, 24, 23, 22, 21, 20, l9, l8, l7, l6, 15 or 14 uous amino acids from SEQ ID NO:l. Said fragment may comprise SEQ ID N054 and may have a length of up to 40, 39, 38,37,36, 35, 34,33, 32, 31, 30,29, 28, 27, 26, 25, 24, 23, 22, 21, 20, l9, l8, l7, l6, 15 or 14 amino acids. Said nt preferably ts of SEQ ID NO:54.
A more preferred fragment of SEQ ID NO:l ses SEQ ID NO: 57 and ses up to 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25 contiguous amino acids from SEQ ID NO:l. Said fragment may comprise SEQ ID NO: 57 and may have a length of up to 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25 amino acids. Said fragment preferably consists of SEQ ID NO: 57.
A more preferred fragment of SEQ ID NO:l comprises SEQ ID NO: 55 and comprises up to 40, 39, 38, 37, 36, 35, 34, 33, 32, 31 contiguous amino acids from SEQ ID NO:1. Said fragment may comprise SEQ ID NO: 55 and may have a length of up to 40, 39, 38, 37, 36, , 34, 33, 32, 31 amino acids. Said fragment preferably consists of SEQ ID NO: 55.
A more preferred fragment of SEQ ID NO:l comprises SEQ ID NO: 56 and comprises up to 50, 49, 48, 47, 46, 45, 44, 43, 42 contiguous amino acids from SEQ ID NO:l. Said fragment may comprise SEQ ID NO: 56 and may have a length of up to 50, 49, 48, 47, 46, 45, 44, 43, 42 amino acids. Said fragment preferably consists of SEQ ID NO: 56.
Each of the red fragments of SEQ ID NO:l as identified herein is preferably able to induce an anti-inflammatory response as earlier defined herein.
The source of L19 may be a protein, a digest of the protein and/or a fragment thereof, which may be in a purified form or may be comprised within a crude composition, preferably of biological origin, such as a bacterial lysate, yeast lysate, fiangal lysate, bacterial supernatant, yeast supernatant, flingal supernatant, sonicate or fixate. Alternatively, a L19 source may be chemically synthesized or tically produced in vitro in a cell free system or in a cellular system. The source of a L19 protein, or fragment thereof, may also be a nucleic acid encoding said, or fragment thereof, from an RNA or DNA template. The RNA or DNA molecules may be ‘naked’ DNA, preferably comprised in vesicles or mes, or they may be comprised in a vector. The vector may be any binant) DNA or RNA vector known in the art, and preferably is a plasmid; wherein genes encoding latency antigens are operably linked to regulatory sequences ring expression and translation of the encoded messengers. The vector may also be any DNA or RNA virus, such as, but not limited to, Adenovirus, Associated Virus (AAV), a retrovirus, a lentivirus, modified ia Ankara virus (MVA) or Fowl Pox virus, or any other viral vector capable of ring expression of said polypeptide into a chosen subject. DNA vectors may be non-integrating, such as episomally replicating s, or may be vectors integrating in the host genome by random integration or by homologous recombination.
A L19 source or a composition as defined herein for use ing to the invention may be suitable for in vitro administration to a cell, a tissue and/or an organ of individuals affected by or at risk of developing an inflammatory er, and/or may be suitable for in vivo or ex vivo administration to a cell, a tissue and/or an organ of such individuals and/or may be suitable for in viva administration to such individuals. Depending on the type of source used (protein-based or nucleic acid-based), the skilled person will know which type of formulation is suited. A L19 source may be administered as such (naked n or nucleic-acid).
Alternatively, a nucleic acid-based source may be administered using a nucleic acid construct as defined herein. Said L19 source or a composition as defined herein may be directly or indirectly in vivo, in vitro or ex vivo stered to a cell, tissue and/or an organ of an individual affected by or at risk of developing an inflammatory disorder or in vivo to such individual. Preferably said cells are cells of an individual suffering from an inflammatory disorder. Preferably said tissue is a tissue of an individual ing from an inflammatory disorder. Depending on the inflammatory disorder, a given type of cell or tissue may be more suited to be treated with a L19 source or a composition of the invention. For example a tissue may be skin, blood, ine, lung and suitable cells may be derived from these tissues.
A L19 source or a composition ofthe invention may be indirectly administered using suitable means known in the art. A nucleic acid molecule as defined in a first embodiment may for example be provided to an individual or a cell, tissue or organ of said individual in the form of an expression vector wherein the expression vector encodes a transcript comprising said nucleic acid molecule. The expression vector is preferably introduced into a cell, , organ or individual via a gene delivery vehicle. In a preferred embodiment, there is provided a viral-based expression vector sing an expression cassette or a ription te that drives expression or transcription of a molecule as fied herein. A preferred delivery vehicle is a viral vector such as an adeno-associated virus vector (AAV), or a retroviral vector such as a lentivirus vector and the like. Also plasmids, artificial chromosomes, plasmids suitable for targeted homologous recombination and integration in the human genome of cells may be ly d for delivery of nucleic acid le as defined in a first embodiment.
Improvements in means for providing an dual or a cell, tissue, organ of said individual with a L19 source or a ition as defined herein, are anticipated considering the progress that has already thus far been achieved. When administering a L19 source or a composition, it is preferred that said L19 source or composition is dissolved in a on that is compatible with the delivery method. For intravenous, subcutaneous, intramuscular, intradermal, intrathecal and/or intraventricular administration it is preferred that the solution is a physiological salt solution.
In the t of the invention, a subject or an individual or a patient or an animal means a human or an animal. An animal which is encompassed within the scope of the invention includes a mammal. Preferred mammals include a dog and a cat.
In a preferred embodiment, at least lug of an L19 source is used to induce an anti- inflammatory response. The ranges of dose of L19 source as given above are preferred doses for in vitro or ex vivo uses. The skilled person will understand that depending on the L19 source used, the cell, tissue, organ or subject to be treated, the medium used and the transfection and incubation conditions, the dose of L19 source used may further vary and may need to be optimised any further.
A L19 source is preferably a medicament or for use as a medicament. More preferably, said medicament is for ting, delaying, and/ or treating an inflammatory disorder to a subject 2012/058453 in the need thereof Within the t of the invention, an inflammatory disorder is any inflammatory disease or condition or any condition wherein inflammation will occur at a given stage. Examples of inflammatory diseases or conditions e, but are not d to, rheumatoid arthritis (RA), juvenile toid arthritis, psoriasis, psoriatic arthritis, ankylosing spondylitis, inflammatory bowel disease (IBD) (including Crohn's disease or ulcerative colitis), irritable bowel syndrome, hepatitis, sepsis, alcoholic liver disease, and non-alcoholic sis, nephritis, such as glomerular nephritis, asthma, endocarditis, myasthenia gravis, multiple sclerosis, autoimmune diabetes (any others similar), diabetes mellitus, uveitis, (l) controlling allograft rejection after organ transplantation, graft versus host disease (GVHD), inflammatory lung diseases including asthma and chronic ctive pulmonary disease (COPD) (2), cancer (4) systemic lupus erythematosus, SLE, UV-induced skin inflammation, atopic itis and sarcoidosis.
As used herein, the term "hepatitis" refers to a gastroenterological disease, condition, or disorder that is characterized, at least in part, by ation of the liver. Examples of hepatitis include, but are not limited to, hepatitis associated with hepatitis A virus, hepatitis B virus, tis C virus, or liver inflammation associated with ischemia/reperfiJsion.
In a more preferred embodiment, said medicament is able to alleviate one or more symptom(s) from a treated patient and/or one or more characteristic(s) or parameter(s) of a cell or tissue or organ from a treated patient is/are improved using a L19 source or a composition of the invention. For each inflammatory disease, the d person knows at least one symptom, parameter or characteristic, values of said parameter or characteristic associated with said e and how to assess each of them. If a medicament of the invention is able to induce an anti-inflammatory response as earlier defined herein, said medicament is said to be able to prevent and/or delay the development of an inflammatory disorder or condition or disease and/or to improve one or more characteristic(s) or parameter(s) of a cell or tissue from a d subject as later defined herein.
Below, we give a parameter specific for Rheumatoid arthritis, psoriasis and inflammatory bowel disease respectively.
Rheumatoid arthritis is a ic e and is one of the most common forms of arthritis.
It is characterised by inflammation of the membrane lining the joint, causing pain, stiffness, warmth, redness and swelling.
There are several animal models for RA known in the art. One example is the collagen- induced arthritis (CIA) model, in which mice develop chronic inflammatory arthritis that closely resembles human rheumatoid arthritis. Since CIA shares similar immunological and pathological features with RA, this makes it a suitable model for screening potential treatments for RA. In this model, the basic mechanisms of pathogenesis are known with the various immunological and inflammatory parameters relating to immune-mediated arthritis having been determined. These parameters can be used to assess compound efficacy in the CIA model (5).
RA is preferably diagnosed after having assessed the index of Disease Activity Score (DAS) or the related DAS28 (6) including the measurements of several ters and symptoms on a t. The ment of said s may be carried out by a clinician examining a subject. In a more preferred embodiment, said medicament is able to alleviate one or more symptom(s) from a treated patient and/or one or more characteristic(s) or parameter(s) of a cell or tissue or organ from a treated patient is/are improved using a L19 source or a composition of the invention when said medicament is able to induce a significant change in DAS or DASZS. Other ways of assessing toid arthritis are also described in (6) and in (7). A medicament as defined herein is able to improve one parameter if after at least one week, one month, six month, one year or more of treatment using a L19 source or a composition of the invention. Preferably, the value of said parameter has been improved of at least 1%, 2%, 5%, 10% or more by comparison of the value of said ter before the onset of the treatment.
A ment as defined herein is able to alleviate one symptom or one characteristic of a patient or of a cell, tissue or organ or said t if after at least one week, one month, six month, one year or more of treatment using a L19 source or a composition of the invention, said symptom or characteristic is no longer able.
Inflammatory Bowel Disease (IBD) is a group of inflammatory conditions of the colon and the small ine ing ulcerative colitis and Crohn's disease. Ulcerative colitis is characterized by ation of the colon, resulting in the colon emptying frequently, resulting in diarrhea and ated cramps, fever and weight loss. The lining of the colon becomes damaged, forming ulcers that release mucous, pus and blood. Repeated episodes can result in the formation of scar tissue, and death of colon tissue, or sepsis with severe disease. Current treatments focus on suppressing the abnormal atory process in the colon lining.
A well-characterized animal model for human IBD, ulcerative colitis and especially s e is the 2,4,6-trinitrobenzenesulphonic acid/ethanol (TNBS) induced colitis model.
Colitis induced by intra-rectal administration of TNBS. This induces a T-cell mediated immune response in the c mucosa, leading to a e mucosal inflammation, characterized by the infiltration of T-cells and macrophages throughout the entire wall of the large bowel. The histopathological nature is accompanied by progressive weight loss, bloody diarrhea, large bowel wall thickening (8). The current animal models of colon inflammation do not fillly reflect the complexity of the disease in humans, however, they are valuable tools to evaluate efficacy of therapeutic compounds.
Psoriasis is a common, c skin disease, in which new skin cells grow abnormally resulting in inflamed, swollen and scaly patches of skin, where the old skin has not shed quickly . The most common form is plaque sis, characterised by lesions topped with silvery white scales. sis may be limited to a few lesions, or may involve extensive areas of skin, most ly appearing on the elbows, knees, scalp and trunk. Mild cases of psoriasis are managed by topical applications. However, more severe cases e ultraviolet therapy, which is inconvenient or the use of systemic immunosuppressive therapies, which, due to toxic side effects, are often of limited value in long term use. In addition, psoriasis frequently , including shortly after stopping immunosuppressive therapy.
Several disease models have been developed for the evaluation of potential disease tors. One such model is an in viva xenograft model for psoriasis with human psoriatic skin implanted into a severe immune deficient (SCID) mouse. Therapies that abolish, or reduce the inflammation can be tested by administration to the SCID mice, baring human inflammatory tissue. Efficacy of treatment can be assessed by a range of indices. Psoriasis is a disease that is preferably diagnosed after having assessed the index of Psoriasis Area and Severity Index (PASI), physician global assessment (PGA) (9) or NPF Psoriasis Score (NPFPS ), including the measurements of several parameters and symptoms on a subject. The assessment of said indexes may be carried out by a clinician examining a subject. In a more preferred embodiment, said medicament is able to alleviate one or more symptom(s) from a treated patient and/or one or more characteristic(s) or parameter(s) of a cell or tissue or organ from a d patient is/are improved using a L19 source or a composition of the invention when said medicament is able to induce a significant change in PASI, PGA or NPF-PS. Other ways of assessing psoriasis include the Dermatology Life Quality Index (DLQI) (10) and the Salford sis Index (SPI) also described in (11) A ment as defined herein is able to e one parameter if after at least one week, one month, six month, one year or more of treatment using a L19 source or a composition of the invention. Preferably, the value of said parameter has been improved of at least 1%, 2%, 5%, 10% or more by comparison of the value of said parameter before the onset of the treatment.
A medicament as defined herein is able to alleviate one symptom or one characteristic of a patient or of a cell, tissue or organ or said patient if after at least one week, one month, six month, one year or more of treatment using a L19 source or a composition of the invention, said symptom or characteristic is no longer detectable.
A preferred L19 source as defined herein is for preventing or treating an inflammatory disorder in an individual. An individual which may be treated using such L19 source may already have been diagnosed as having an atory disorder. atively an dual which may be d using such L19 source may not have yet been diagnosed as having an inflammatory er but may be an individual having an increased risk of developing an inflammatory disorder in the future given his or her c background. A preferred individual is a human being. ition In a further aspect, there is provided a composition comprising a L19 source as defined herein. In a preferred ment, said composition being preferably a pharmaceutical composition said pharmaceutical composition comprising a pharmaceutically acceptable carrier, salt, diluent and/or excipient.
Such a pharmaceutical composition may comprise any pharmaceutically acceptable carrier, filler, salt, preservative, solubilizer, diluent and/or excipient is also provided. Such pharmaceutically acceptable carrier, filler, salt, preservative, solubilizer, diluent and/or excipient may for instance be found in (12). Each feature of said composition has earlier been defined herein.
If several L19 sources are used, dose as defined herein may refer to the total dose of all L19 sources used or the dose of each L19 source used or added. Therefore in one embodiment, there is provided a composition wherein each or the total amount of L19 source used is dosed in an amount from 0.1mg/kg and lOOmg/kg.
Particularly preferred in the invention is the use of an excipient that will aid in delivery of each of the constituents as defined herein to a cell and/or into a cell. Preferred are excipients capable of forming xes, rticles, es, vesicles, liposomes, proteoliposomes, and/or virus like les (VLP) that deliver each constituent as defined herein, complexed or trapped in a vesicle or liposome h a cell membrane. Many of these excipients are known in the art. Suitable ents comprise polyethylenimine (PEI), or similar cationic polymers, including polypropyleneimine or polyethylenimine mers (PECs) and derivatives, synthetic amphiphils (SAINT-18), lipofectinTM, DOTAP and/or viral capsid proteins that are capable of self assembly into particles that can deliver each constituent as defined herein to a cell.
Depending on their identity, the skilled person will know which type of formulation is the most appropriate for each constituent as defined herein. In a preferred embodiment, the invention es a composition or a preparation which is in the form of a kit of parts comprising a L19 source as defined herein.
A medicine or medicament or pharmaceutical composition as defined herein may be locally or systemically administered. A medicament is preferably administered parenterally, e. g. by injection or infilsion by intravenous, subcutaneous, intraperitoneal, intramuscular, intradermal, intraarterial or intralesional route. A preferred stration mode is subcutaneous or transdermal. An example of transdermal administration is a cream. The invention is not limited to a specific mode of administration of a medicament or a L19 source or a composition as defined herein. A preferred mode of administration is oral administration using a capsule or a tablet. atively a medicament or a L19 source or a composition as defined herein may be locally administered via a catheter or a pump, or a suppository or a cream. Alternatively, a medicament or a L19 source or a ition as defined herein may be topically administered. The formulation of a ment or a L19 source or a composition as defined herein depends on the intended mode of administration and (therapeutic) application. A pharmaceutical carrier can be any compatible, non toxic substance suitable to deliver said compound to a subject. E.g. sterile water, or inert solids or excipients may be used as the carrier, usually complemented with ceutically acceptable adjuvants, buffering agents, dispersing agents, and the like. Compositions will either be in liquid, e.g. a stabilized suspension of said compound, or a composition comprising said compound, or in solid and/or dry forms: e.g. powder. For oral and rectal administration, said compound can be administered in solid dosage forms, such as es, tablets, suppositories, and powders, or in liquid dosage forms, such as elixirs, syrups, cream, ointment and suspensions. Another form may be a semi-solid or semi-liquid form wherein said compound is present as a liquid form in or on a solid support such as a patch.
A composition may be in the , solid or semi-liquid or semi-solid form as already defined herein.
In a preferred embodiment, other compounds are used sequentially or simultaneously with a L19 source or a composition in order to improve the specificity of the therapeutic or prophylactic ent. It is advantageous for example to use other compounds that will r e the anti-inflammatory response of the treated subject. More preferably, such compounds are not present in a single composition together with a L19 source or composition. Such compound may be an antibody, a DMARD (disease-modifying anti- rheumatic drugs), a NSAID (Non-steroidal Anti-inflammatory Agents) and/or an IL-lO r such as those described in table 1 of (13). An IL-lO inducer includes a nd selected fiom the group consisting of: cordycepin, a gold salt, a corticosteroid, cyclosporine A, STl959 3-(2-ethylphenyl)(3-methoxyphenyl)-lH-l,2,4-triazole, SR 3l747A, SSR 125329A, aprotinin, linomide, monomethylfumarate, cAMP-elevating agents such as rolipram or cicaprost, a olamine, vitamin D3, a fish oil comprising a n- 3polyunsaturated fatty acid, an estriol sex e, KM 2210 or bestrabucil, a type I IFN such as IFN-r IFN-0t or IFN-B, a mimic ntigen as glatiramer acetate (copolymer I), a pyrimidylpiperazine or a tive thereof, l-ethyl(3-dimethyl aminopropyl) urea dihydrochloride, 5’-methylthioadenosine and a pirfenidone such as 5-methyl-l-phenyl-lH- pyridine-one.
In a r aspect, there is provided the use of a L19 source or of a composition as defined herein for the manufacture of a medicament for preventing or treating an inflammatory disorder in an individual. Each feature of said use has earlier been defined herein.
A treatment in a use or in a method according to the invention is at least one week, at least one month, at least several months, at least one year, at least 2, 3, 4, 5, 6 years or more. Each L19 source as defined herein for use according to the invention may be suitable for direct in vivo, in vitro or ex vivo administration to a cell, tissue and/or an organ of individuals affected by or at risk of developing an inflammatory disorder, and may be stered directly in vivo to said individuals. The frequency of administration of a L19 source or composition of the invention may depend on several parameters such as the age of the patient, the number of molecules (i.e. dose), the formulation of said molecule. The frequency may be daily, weekly or ranged between at least once in two weeks, or three weeks or four weeks or five weeks or a longer time period.
Method In a further , there is provided a method for alleviating one or more m(s) of an inflammatory disorder in an individual, in a cell, tissue or organ of said individual or alleviate one or more characteristic(s) or symptom(s) of an individual or a cell, tissue or organ of said individual, the method comprising administering to said dual a L19 source or a composition as defined herein.
In one embodiment said method is performed in vitro, for instance using a cell culture or a tissue culture. Said method may also be ex vivo. Preferably, said method is in viva. Each feature of these methods has already been defined herein. In a method of the invention, a L19 source may be combined with an additional compound known to be used for ng an inflammatory er in an individual. Such compound may be an antibody, a DMARD (disease-modifying anti-rheumatic drugs), a NSAID (Non-steroidal Anti-inflammatory Agents) and/or an IL-lO inducer as described in (13). Preferred IL-lO inducers have already been identified earlier herein.
Definitions c acid le A nucleic acid molecule may be a cDNA or synthetic DNA. The DNA may be double- stranded or single-stranded and if single-stranded may be the coding strand or non-coding (anti-sense) . DNA or RNA with a backbone modified for stability or for other reasons are a further part of the invention. A nucleic acid le is represented by a nucleotide sequence. A nucleotide ce may be an allelic variant of the nucleotide sequence according to the invention. If desired, the nucleotide sequence can be prepared or altered synthetically so the known codon preferences of the intended expression host can advantageously be used. Depending on the size of the nucleic acid molecule, it could be identify as being an oligonucleotide. An oligonucleotide may comprise at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 nucleotides.
Polypeptide “Polypeptide” as used herein refers to any peptide, oligopeptide, polypeptide, gene product, expression product, or protein. A polypeptide is represented by an amino acid sequence. It may comprise from 2 to 267 (i.e. length of SEQ ID NO:1, 5, 7, 9,11,13,15,17,19, 21, 23 ,25, 27 or 29) or 5 to 265 or 8 to 260 or 10 to 250 amino acids. It may comprise more than 267 amino acids. The term "polypeptide" encompasses naturally occurring or synthetic molecules. An oligopeptide may comprise 2 to 20 amino acids. A e may comprise 5 to or 5 to 20 or 5 to 30 or 5 to 50 amino acids.
Identity/similarity "Sequence identity" is herein defined as a relationship between two or more amino acid (polypeptide or protein or peptide or protein nt) sequences or two or more nucleic acid (polynucleotide, nucleic acid or nucleotide or oligonucleotide) ces, as ined by comparing the sequences. In a preferred embodiment, sequence identity is calculated based on the filll length of two given SEQ ID NO or on part thereof Part thereof preferably means at least 50%, 60%, 70%, 80%, 90%, or 100% of both SEQ ID NO. In the art, "identity" also means the degree of sequence dness between amino acid or nucleic acid sequences, as the case may be, as determined by the match between strings of such sequences.
"Similarity" between two amino acid sequences is determined by comparing the amino acid sequence and its conserved amino acid substitutes of one ptide to the sequence of a second polypeptide. "Identity" and "similarity" can be readily calculated by known s, including but not limited to those described in (Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; er Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heine, G., ic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; and Carillo, H., and , D., SIAM J. Applied Math., 48:1073 (1988).
Preferred methods to determine ty are designed to give the largest match between the sequences tested. Methods to determine identity and similarity are codified in publicly ble er ms. Preferred computer program s to determine identity and rity between two sequences include e.g. the GCG program e (Devereux, J ., et al., Nucleic Acids Research 12 (1): 387 ), BestFit, BLASTP, BLASTN, and FASTA (Altschul, S. F. et al., J. Mol. Biol. 215:403-410 (1990). The BLAST X program is publicly available from NCBI and other sources (BLAST Manual, ul, S., et al., NCBI NLM NIH Bethesda, MD 20894; Altschul, S., et al., J. Mol. Biol. 215:403-410 (1990). The well- known Smith Waterman algorithm may also be used to determine identity.
Preferred parameters for polypeptide sequence comparison include the following: Algorithm: Needleman and Wunsch, J. Mol. Biol. 48:443-453 (1970); ison matrix: BLOSSUM62 from Hentikoff and Hentikoff, Proc. Natl. Acad. Sci. USA. 15-10919 (1992); Gap Penalty: 12; and Gap Length Penalty: 4. A program usefill with these parameters is publicly available as the "Ogap" program from Genetics er Group, located in Madison, WI. The aforementioned parameters are the t parameters for amino acid comparisons (along with no penalty for end gaps).
Preferred parameters for nucleic acid comparison include the following: Algorithm: Needleman and Wunsch, J. Mol. Biol. 48:443-453 (1970); Comparison matrix: matches=+10, mismatch=0; Gap Penalty: 50; Gap Length y: 3. Available as the Gap program from Genetics Computer Group, located in n, Wis. Given above are the default parameters for nucleic acid comparisons.
Optionally, in determining the degree of amino acid similarity, the skilled person may also take into account so-called "conservative" amino acid substitutions, as will be clear to the skilled person. Conservative amino acid substitutions refer to the interchangeability of residues having similar side chains. For example, a group of amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having tic-hydroxyl side chains is serine and threonine; a group of amino acids having amide- containing side chains is asparagine and glutamine; a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan; a group of amino acids having basic side chains is lysine, arginine, and histidine; and a group of amino acids having sulphur- containing side chains is cysteine and methionine. Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, and asparagine-glutamine. Substitutional variants of the amino acid sequence disclosed herein are those in which at least one residue in the disclosed sequences has been removed and a different residue ed in its place. Preferably, the amino acid change is conservative. Preferred conservative substitutions for each of the naturally occurring amino acids are as s: Ala to ser; Arg to lys; Asn to gln or his; Asp to glu; Cys to ser or ala; Gln to asn; Glu to asp; Gly to pro; His to asn or gln; Ile to leu or val; Leu to ile or val; Lys to arg; gln or glu; Met to leu or ile; Phe to met, leu or tyr; Ser to thr; Thr to ser; Trp to tyr; Tyr to trp or phe; and, Val to ile or leu.
Hybridization conditions Hybridization conditions for a nucleic acid molecule may have low or medium or high stringency (southern blotting procedures). Low or medium or high stringency conditions means pre-hybridization and hybridization at 420C in 5x SSPE, 0.3% SDS, 200pg/ml sheared and denatured salmon sperm DNA, and either 25% or 35% or 50% formamide for low or medium or high stringencies respectively. Subsequently, the hybridization reaction is washed three times for 30 minutes each using 2x ssc, 0.2% SDS and either 55 0C or 65 0C, or 75 0C for low or medium or high stringencies respectively.
Nucleic acid construct/expression/control sequences A nucleic acid construct comprises a tide sequence encoding a protein or a protein fragment as d herein. A c acid construct comprising a nucleic acid molecule coding for a given protein or protein fragment as defined herein will ensure expression ofthe given nucleic acid molecule, and of the corresponding protein or n fragment in a d subject. In a more preferred embodiment, a nucleic acid construct comprises more than one nucleic acid molecule, each nucleic acid molecule coding for a given protein or protein fragment. In an even more preferred embodiment, a nucleic acid construct comprises two, three, four c acid molecules, each nucleic acid molecule coding for a given protein or n fragment. In a preferred ment, a nucleic acid construct comprises an expression cassette, said expression cassette comprising each needed nucleic acid molecule.
Each nucleic acid molecule is ly linked with other c acid molecule present. Most preferably, a suitable promoter is operably linked with the expression cassette to ensure expression of the nucleic acid molecule in a subject.
“Operably linked” is defined herein as a uration in which a control sequence is appropriately placed at a position relative to the nucleotide sequence coding for the polypeptide of the ion such that the l sequence directs the production/expression ofthe polypeptide of the invention in a cell and/or in a subject. sion will be tood to e any step involved in the production of the polypeptide including, but not limited to transcription, post-transcriptional ation, translation, post-translational modification and secretion.
Control ce is defined herein to include all components, which are necessary or advantageous for the expression of a polypeptide. At a minimum, the control sequences include a promoter and riptional and translational stop signals. Optionally, a promoter represented by a nucleotide sequence present in a nucleic acid construct is operably linked to another nucleotide sequence ng a nucleic acid molecule as identified herein.
An expression vector may be any vector which can be conveniently subjected to recombinant DNA procedures and can bring about the expression of a nucleotide sequence encoding a ptide of the invention in a cell and/or in a subject. As used herein, the term "promoter" refers to a nucleic acid fragment that filnctions to l the transcription of one or more genes or nucleic acids, located upstream with respect to the direction of transcription of the transcription initiation site of the gene, and is related to the binding site identified by the presence of a binding site for DNA-dependent RNA polymerase, transcription initiation sites and any other DNA sequences, including, but not limited to transcription factor binding sites, repressor and activator protein binding sites, and any other sequences of nucleotides known to one skilled in the art to act directly or indirectly to regulate the amount of transcription from the promoter. Within the context of the invention, a promoter preferably ends at nucleotide -l ofthe ription start site (TSS).
Unless otherwise indicated each embodiment as described herein may be combined with another embodiment as described herein.
In this document and in its , the verb "to comprise" and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded. In addition the verb “to consist” may be replaced by “to consist essentially of’ meaning that a L19 source or a composition as defined herein may comprise additional component(s) than the ones specifically identified, said additional component(s) not altering the unique characteristic of the invention.
In on, reference to an element by the indefinite article a or "an" does not exclude the ility that more than one of the element is present, unless the context clearly requires that there be one and only one of the elements. The indefinite article "a" or "an" thus usually means "at least one".
The word “approximately” or “about” when used in association with a numerical value (approximately 10, about 10) preferably means that the value may be the given value of more or less 1% of the value.
All patent and literature references cited in the present cation are hereby incorporated by reference in their entirety. Each ment as identified herein may be combined together unless otherwise indicated.
The invention is further explained in the following examples. These es do not limit the scope of the invention, but merely serve to clarify the invention.
Brief description of the drawings Figure 1. Purification of the his-LmL19. SDS-PAGE gel showing the different steps in the purification of the erLl9 protein. (1) Molecular weight standard. Total bacterial extracts after (2) and before (3) column passage. (4) Purified protein.
Figure 2. IFN- gamma and IL-10_ production by splenocytes of naive BALB/c mice (n=6) stimulated in vitro with Lle9. The P value obtained after the statistical analysis performed by a Student's t-test is shown. Differences in the tion of the cytokines between the Lle9 or the Concanavalin A (ConA) stimulated cells were considered significant when P< 0.05 *).
Figure 3. IFN- gamma and IL-10_ production by PBMCa of healthy human donorse (n=3) stimulated in vitro with Lle9. The P value obtained after the statistical analysis med by a Student's t-test is shown. Differences in the production of the nes between the Lle9 or the ConA stimulated cells were considered significant when P< 0.05 Figure 4. Schematic entation of the MBP-LmL19 inant protein.
Figure 5. Concentration of TNFd in the different study groups after 24 hours of incubation (pg/ml). The values are presented as average :: standard ion. #; p<0.005, test groups without irradiation compared to the Control group.*; p<0.005, ated test groups compared to the Control / UV Group.
Figure 6. Concentration of IL-lO in the different study groups after 24 hours of incubation (pg/ml). The values are presented as average :: standard deviation.#; p<0.005, test groups without ation compared to the Control group.*; p<0.005, irradiated test groups compared to the Control / UV Group.
Figure 7. Lle9 peptide 11-10 production. Cells were obtained and es as indicated in the text. The level of IL-lO in culture supematants were analyzed by ELISA.
The assay has been performed once using pooled cells obtained from three naive mice (Assayed by duplicate). * P< 0.05 when each group was compared with the non-stimulated cells.
Examples Example 1 Cloning and expression.
The gene was characterized after in silico search in the L. major genome database. On the basis of the sequence two ucleotides were synthesized (see below) and ed as primer for a PCR using DNA extracted from L. major [clone Vl (MHOM/IL/80(Friedlin)] parasites. The obtained DNA was digested with BamHI/HindIII, cloned in the corresponding sites of pBluescript plasmid and sequenced. The obtained DNA sequence and the d aminoacid sequence is shown below, respectively.
A. Oligonucleotides employed.
LmL19D: CATGACCCCTCTCTCCCTCTC (SEQ ID NO:3) (Underlined a BamHI cut site was included for cloning purposes).
LmL19R: cccAAGCTTTTACTTCTTCGACTTCTTCAC (SEQ ID NO:4) (Underlined a HindIII cut site was included for cloning purposes).
B. Nucleotide sequence.(The ction enzymes cut sites are included and marked in italics and underlined: these sites do not belong to the nucleic acid molecule ng L19 from Leishamnia major or Lm) Sequence analysis of the Lle9 (SEQ ID NO:2 is the sequence below without the underlined sequence added for cloning purposes) GGATCCATGA CCCCTCTCTC CCTCTCTTCC TCCCGCCACA GTTTTAAGCA AACG CAGAACATGG TGTCTCTGAA GCTGCAGGCT CGCCTTGCGT CGAGCATCCT CGGCTGCGGC CGCGCCCGCG TGTGGCTGGA CCCCAACGAG GCGGTGGAGA TCCAGAACGC GCGC AAGAGCGTGC GCAAGCTGAT CAAGGATGGC TTCATCATCC GCAAGCCGGT GAAGGTGCAC TCGCGCGCGC GTAA AATGAAGGAG GCGAAGGACA TGGGGCGCCA CAACGGCGTT CGCG AGGGTAGCCG CGAGGCCCGC ATGCCGAGCA AGGAGTTGTG GATGCGCCGC CTGCGCATTC TGCGCCGCCT GCTGCGCAAG TACCGCGCGG ACAAGAAGAT TGACCGCCAC GTGTACCGCG ACCTGTACAT GCGCGCGAAG GGTAACGTGT ACAA GCGCAACCTT GTGGAGCACA TCCACAAGAT CAAGAATGAG AAGAAGAAGG AGCGCCAGCT GGCGGAGCAG CTCGCGGCGA AGCACCTGCG CGACGAGCAG AACCGCAACA AGGCTCGCAA GCAGGAGCTG AAGAAGCGCG AGAAGGAGCG CGAGCGCGCG AGGCGCGACG ACGCTGCTGC CGCTGCGCAG AAGAAGAAGG CGGACGCCGC GAAGAAGTCC GCCGCGCCTG CTGCGAAGTC GCCT GCCGCGAAGG CTGCTGCCCC GAAG GCCGCTGCTG CTGCCCCCGC CACGAAGGGT GCTGCGCCGG TGAAGAAGTC GAAGAAGTAA AAGCTT C. Deduced amino acid sequence (SEQ ID NO: 1) MTPLSLSSSR HSFKQNETQN MVSLKLQARL ASSILGCGRA NEAV EIQNANSRKS VRKLIKDGFI IRKPVKVHSR ARWRKMKEAK DMGRHNGVGR REGSREARMP SKELWMRRLR ILRRLLRKYR ADKKIDRHVY RDLYMRAKGN VFRNKRNLVE HIHKIKNEKK KERQLAEQLA AKHLRDEQNR NKARKQELKK REKERERARR DDAAAAAQKK KADAAKKSAA PAAKSAAPAA KAAAPATKAA KGAA PVKKSKK* The DNA encoding Lle9 was subcloned in the BamHI/HindIII sites of the pQE-30 prokaryotic expression d that allow the obtention of the recombinant protein fiJsed to 6xhistidines.
Escherichia 6011' (strain M15) cultures ormed with the recombinant plasmid was employed for the expression of the recombinant protein. The first assays were done at 37°C, but we observed that the protein was degraded inside the bacteria. For that reason, cultures were induced at 30°C in order to se protein degradation. At these conditions we ed a low production of the intact recombinant protein. Thus, erLl9 was purified by affinity chromatography under denaturing conditions. The purified n obtained presents some ation bands with lower lar weight (Fig. l). The recombinant proteins were passed through a polymyxin—agarose column to remove endotoxins. ation ofmice spleen cells with the recombinant erL19.
Single-cell preparations from spleen tissue were plated in duplicate in l plates at 5 x 106 cells/ml. Cells were incubated in complete RPMI medium supplemented with 2 mM L- glutamine, penicilin (100 U/ml), streptomycin (100 ug/ml) and 10% heat inactivated foetal bovine sera alone (background control; medium) or stimulated with erLl9 (l2ug/ml) or ConA l) at 37°C in 5% C02 for 72 h. IFN-gamma and IL-lO release in the culture supematants was assessed by sandwich ELISA (Fig. 2). It can be concluded that the recombinant Lle9 protein induced an c production of IL-lO, without the production ofIFN—gamma by spleen cells obtained from naive mice.
Stimulation of human peripheral mononuclear blood cells (PBMCs) from humans with the recombinant erL19.
PBMC were obtained from heparinized venous blood by passage over a Ficoll Hypaque gradient. PBMC were washed three times and resuspended at a concentration of 5 X 106 cells/ml in RPMI supplemented with 2 mM L- glutamine, penicilin (100 U/ml), streptomicin (100 ug/ml) (Gibco, NY) and 10% heat inactivated human AB serum). Cells were plated in 24 well tissue culture plates at a concentration of 5 X 106 cells/ml and ted at 37°C, 5% CO2. Stimulation was performed by addition of erLl9 (20 ug/ml and 5 ug/ml) and ConA (l ug/ml) for 72 h. As above, IFN—gamma and IL-lO release in the culture supematants was assessed by sandwich ELISA (Fig. 3).
As occurred with mice spleen cells, PBMC form healthy human donors produced IL-lO after in vitro stimulation with the recombinant Lle9. The production of this cytokine was dose dependent.
Expression ofthe erL19 as afusion protein with the maltose bindingprotein.
These preliminary results were indicating that the erLl9 protein was able to induce the IL- release from human and mice white cells. The level of production of the recombinant protein was not yet optimal. .
In order to e protein production the DNA encoding the LmL9 protein was cloned in the pMal-c2 yotic expression . This vector allows the production of heterologous proteins in E. 0012' fused to the bacterial MBP protein. As indicated in Fig. 4 the fiasion protein and the heterologous protein can be separated using an specific protease (Xa factor) due to the ce of an Xa factor cut site between both ns.
E. colz’ (strain XLl-blue) cultures transformed with the recombinant plasmid was ed for the expression of the recombinant protein. Protein was overexpressed as a e product that was purified by affinity chromatography under native conditions in amilose columns.
Using this system higher level ofrecombinant protein was obtained (not shown).
Example 2: skin study The objective of this study was to evaluate the anti-inflammatory capacity of a protein named, L19, in human skin explants. This was done by a screening method to study the protective efficacy of this protein versus radiation of iolet (UV) light on the skin.
The basis of this work is the performance of a single inflammatory study on skin explants to assess the anti-inflammatory capacity of said product versus ion with UV light. The study was divided into the following tasks in order to achieve the proposed objective.
Basic cytotoxicity screening A single assay was performed using the MTT technique to determine the maximum concentration of the product to be assayed in efficacy screening. This is a colorimetric assay based on the metabolic reduction oftetrazolium salts (MTT) due to cell metabolism (cellular respiration) of mouse fibroblasts (BALB/3T3. This metabolic reduction of the MTT is caused by the mitochondrial enzyme ate dehydrogenase, which produces a blue compound zan) and determines the mitochondrial fianctionality of the treated cells. The number of live cells is proportional to the resulting blue color. The product was incubated at 8 different trations (6 replicas of each concentration) for 24 hours.
Anti-inflammatory efficacy screening Efficacy was assessed by an nflammatory study ting in a single assay on skin explants that analyzed two interleukins present in the inflammation caused by UV radiation using the ELISA que.
The study groups were as follows: 0 Healthy control group. 3 skin explants. These did not receive UV light radiation. 0 Damaged group. 3 skin explants irradiated with UV light. 0 Test group. 9 skin explants irradiated with UV light and then incubated with recombinant Lm L19 (SEQ ID NO:1) expressed in E. Cali. 0 Test group 2. 9 skin explants not irradiated and then incubated with Lm L19 (SEQ ID NO:1) expressed in E. 0012'.
Three different product concentrations, 3 replicas per concentration (the highest product tration was determined by product toxicity screening).
After irradiation of the explants with ultraviolet light, Lm L19 (i.e. SEQ ID N021) expressed in E. Coli was incubated; 24 hours later, we measured two interleukins involved in the inflammatory effect caused by ultraviolet light on the skin, IL-10 and TNFu.
MATERIALS AND METHOD Cell cultures As the experimental system for xicity screening, the study used a cell culture of the immortalized line of mouse fibroblasts from the cell line BALB/3T3 from the European Collection of Cell es (ECACC) Cat. No. 01.
The immortalized fibroblasts from the ECACC grew in a DMEM medium with 10% FCS (Fetal Calf Serum). After thawing, the cells were cultivated in a monolayer in a humid atmosphere with 5% C02 and at a temperature of 37°C. During this time, the culture medium WO 52792 was changed every 2-3 days, according to the instructions of the supplier. After this period, when the culture flasks reached a confluence of 80%, the cells were distributed on 96-well plates at a concentration of 5,000 cells per well.
Cytotoxicity study with endpoint, MTT During the cytotoxicity assay, the cells were treated with ent concentrations of the study product known as L19.After 24 hours of treatment, MTT staining was performed. This assay is based on the lic reduction of 3-(4,5-dimethyl thiazoleyl)-2,5-diphenyl tetrazol bromide (MTT) or tetrazolium salts (yellow and soluble) produced by the mitochondrial enzyme succinate-dehydrogenase that generates a compound with a blue color (formazan) that allows determination of the ondrial flinctionality of the treated cells. This method is widely used to measure cell survival and proliferation. The number of live cells is proportional to the amount of formazan produced. Since dead cells do not breath, they do not present the enzyme and therefore cannot reduce it since they do not present succinate- dehydrogenase. The r the reduction in MTT, the bluer the color and the greater cell viability.
The experiment was performed on 96-well plates with 3T3 cells grown on a yer with 80% confluence. These cytotoxicity studies allowed us to determine LC80, LC50 and LC20 values (product concentrations that reduce cell viability by 80%, 50% and 20%, respectively).
During this task, a plate of immortalized flbroplasts was ted with eight distinct concentrations of L19 protein for 24 hours (1 product plate with 6 replicas per concentration assayed) and a second MTT assay control plate with sodium dodecyl e (SDS) at eight distinct concentrations (1 plate with MTT, with 6 replicas per concentration assayed) as the toxicity reference product. This was used to establish a standard curve for cell death. All the study plates were also seeded with fibroblasts in at least 12 wells that were used as healthy controls, and 3T3 cultures with only the culture medium, where L19 was not added to the study plate nor SDS to the cell death control plate (SDS Data: LC20: 0,124 mg/m1; LC50: 0,142 mg/m1; LC80: 0,163 mg/ml).
According to the tration of protein L19 supplied by LETI, the highest concentration assayed was 200 ug/ml at ons of 1:2. The final trations used are detailed below: C2: C3: C4: C8:l.56 ug/ml.
After incubation, the plates were developed with MTT and absorbance was ed at 540 nm with an ELISA plate reader. The results obtained were used to ate the lethal concentration values LC80, LCSO and LC20, in the fibroblast cultures for the products under study.
Interleukin-10 and TNFa determination assay The interleukin-lO and TNFu quantitation assay was performed with the supernatant of the skin explant culture mediums. Quantitative determination of both IL-lO and TNFOL was performed with BD OptEIATM ELISA kits manufactured by Becton Drive, Franklin Lakes, NJ, USA): Human TNF ELISA Kit II L19 efficacy was assessed by quantitation of the eukins induced by UV radiation on human skin explants.
Exposure to solar simulator light: The plate with the skin samples was exposed to UV/vis light emitted by a SOL 500 (Dr.
Honle) solar tor. Light intensity was measured throughout the exposure process by a UV light meter. Radiation doses can be adjusted with these values, considering that during 5 minutes of exposure, the cells receive approximately 1 J/cm2 at that intensity. The final time of exposure of the skin explants was 50 minutes, implying radiation of 10 J/cmz.
The groups included in the study were: 0 Healthy control group. 3 skin explants. These did not receive UV light radiation. 0 Damage group. 3 skin explants ated with UV light. 0 Test group. 9 skin explants irradiated with UV light and then incubated with recombinant Lle9 (i.e. SEQ ID NO:l) expressed in E. 0012'. 0 Test group 2. 9 skin ts not irradiated and then incubated with Lle9 (i.e. SEQ ID NO: 1).
Three different product trations, 3 replicas per concentration (the highest product concentration was determined by product toxicity screening). After irradiation of the ts with iolet light (10 J/cmz) they were incubated with product L19. After 24 hours, we measured two interleukins involved in the inflammatory effect caused by ultraviolet light on skin, IL-10 and TNFOL.
IL-10 and TNFOL were quantified by adding 100 ul of each of the IL-10 and TNFu standards included in the kits manufactured by BD Biosciencies (BD OptEIATM ELISA Sets: IL-10 Cat- No. 555157 and TNF Cat. No. ), as well as 100 ul ofthe culture medium used to incubate each of the samples, all in duplicate and incubating on ELISA plates at ambient temperature for 2 hours. The wells were then washed with PBS, 200 ul of the conjugated solution was added and incubation was performed for 1 hour. The wells were then washed again, 200 ul of the substrate solution was added and incubation was performed for 20 minutes at ambient temperature.Lastly, 50 ul of the stopper solution was added and ance was read at 450 nm with a reference of 540 nm. The absorbance results were extrapolated to the amount of IL-10 and TNFu, using the curve obtained with the standards of both cytokines as reference.
RESULTS AND DISCUSSION Basic cytotoxicity screening Cytotoxicity study of n L19.
To determine the maximum concentration of the study product in efficacy screening, a single assay was performed using the MTT technique on an immortalized line of fibroblasts, BALE/3T3, which were seeded on 96-well plates with an approximate density of 5,000 cells per well. The product was incubated at 8 different concentrations (6 replicas of each concentration) for 24 hours.
The results of this assay were used to define the concentrations necessary to determine LCgo, LC50 and LCZO. LC is the lethal concentration of the substance. LCgo is the concentration of the substance at which 80% of the cell tion dies, LC50 is the concentration of the substance at which 50% of the cell population dies and LCZO is the toxic concentration ofthe substance at which 20% of the cell culture population dies.
In these assays, each fibroblast plate was incubated with six distinct active trations for 24 hours: C2: C3: C4: C5:C6:C8:1.56 ug/ml.
After incubation, the plates were developed for analysis with MTT and absorbance was measured at 540 nm with an ELISA plate reader.The following table shows LC80, LC50 and LC20, obtained for each product (Table 1) without statistical significance: Table 1. s for LC20, LC50 and LC80 of n L19 from LETI obtained by cytotoxicity assays, MTT. 34 228.357 22.03021 The values are expressed as mean (ug/ml) The results of this cytotoxicity assay were obtained taking into consideration the absorbance values obtained in the healthy l cultures that were not incubated with the L19 t, as a reference of 100% viability. As well as occurred with the cell death values, viability was 0% with the SDS t at a concentration > 175 ug/ml (SDS Data: LC20: 0,124 mg/m1; LC50: 0,142 mg/m1; LC80: 0,163 mg/ml).
Due to the results obtained in the cytotoxicity assay, it was decided that for the next task, anti-inflammatory efficacy screening of protein L19, the highest concentration to test of the product would be 25 ug/ml, as well as 12.5 ug/ml and 6.25 ug/ml.
The choice of the highest concentration, 25 ug/ml, refers to LC20.This value is adequate for the assay of the product on skin since it is at the minimum toxicity limit for the product. It should also be noted that toxicity screening was performed in a single culture of fibroblasts.
Monocultures are always more sensitive to the toxicity of a product than an organotypic culture such as skin explant.
Anti-inflammatory efficacy screening Study on the anti-inflammatory ty of protein L19 after irradiation of UV light on human skin explants.
Treatment of human skin explants with protein L19 (25 ug/ml, 12.5 and 6.25 ug/ml for 24 hours) (Lm L19 or SEQ ID NO: 1) was performed to study the anti-inflammatory effect ofthe protein. The study groups were divided into explants that were irradiated with UV light and later incubated with protein L19 and explants that were exposed to the product but not ated with UV light.
The study was performed on organotypic cultures of human skin explants from cosmetic surgery. The assay used two control groups; e without protein L19 or solar radiation and a damage control group, culture without the protein but irradiated with solar light. The concentrations and conditions can be found below: 0 Control Group: Skin ts in normal culture conditions. 0 L19-25 u g/ml Group; Skin explants incubated with 25 ug/ml ofprotein for 24 hours. 0 L19-125;; g/ml Group; Skin explants incubated with 125 ug/ml of protein for 24 hours. 0 L19-6.25ug/ml Group; Skin explants incubated with 6.25 ug/ml of protein for 24 hours. 0 l / UV Group: Skin explants in normal culture conditions and irradiated with UV light, 10 J/cmz. o L19-25 ug/ml / UV Group; Skin explants irradiated with UV light (10 J/cmz) and then ted with 25 ug/ml of protein for 24 hours. 0 .5 ug/ml / UV Group; Skin explants ated with UV light (10 J/cmz) and then incubated with 125 ug/ml of protein for 24 hours. 0 L19-6.25ug/ml / UV Group; Skin explants irradiated with UV light (10 J/cmz) and then incubated with 6.25 ug/ml of protein for 24 hours.
At the end of the incubation period (24 hours) fication was performed on both cytokines, IL-10 and TNFu, with the ELISA technique to determine the quantity of these cytokines. A single assay was performed in this task, where protein L19 from LETI Laboratories was analyzed. This assay used 3 as of each condition and each replica was ed by ELISA que in duplicate.
The concentration of TNFu (Figure 5) and the concentration of IL-10 (Figure 6) for these study groups are shown below, both for skin explants irradiated with UV light and those that were not irradiated. Figure 5 shows the production of TNFu in the skin explants in the conditions to which they were submitted.
The inflammatory reaction ed by UV radiation in the l / UV group (153.9 :: 44 pg/ml) is significantly higher than in the control group without radiation (22.9 :: 7.3 pg/ml).
This indicates that radiation with 10 J/cm2 ofUV light (in monoculture, the radiation used is normally 1 J/cmz) on skin explants triggers an inflammatory reaction in this case. This reaction is sufficient to see whether protein L19 has anti-inflammatory effects after incubation in skin samples exposed to this level of radiation.
As shown in the graph, the statistical study of this protein shows that the production ofTNFu after UV light radiation ses significantly in the explants that were incubated ards (for 24 hours) with protein L19 at concentrations of 25 and 12.5 ug/ml (23.3 :: 2.8 and 25.9 :: 4.8 pg/ml, respectively). The L19-6.25ug/ml / UV group showed a non-significant reduction in the production of said cytokine.
On the other hand, the graph also shows a significant increase in the production of TNFu in the L19-25 ug/ml, L19-12.5 ug/ml and L19-6.25 ug/ml groups (that were not irradiated) in comparison with the Control group. All of the above leads us to consider that protein L19 could affect the skin in some way, ting an inflammatory reaction. This last point should be corroborated by further studies.
The significant decrease in the production of TNFu in the cultures with n L19 d ards with solar radiation versus the Control / UV group leads us to think that this protein may have an anti-inflammatory effect.
Figure 6 shows the production of IL-10 in the skin explants in the conditions to which they were submitted. The analysis of IL-10 in both irradiated and non-irradiated groups incubated with protein L19 indicated a significant increase in the production of IL-10 at all the concentrations of the protein under study versus the non-irradiated Control group (#; In the statistical study, this increase was ed to a lesser degree versus the Control group irradiated with UV light, where only the L19-12.5ug/ml / UV group presents a significant increase (*; p<0.005), 363.7 :: 22 pg/ml, versus the Control / UV group : 77). This is due to the standard deviations in the other two study groups, L19-6.25ug/ml / UV and L19- ug/ml / UV (366 :: 94 and 724.8 :: 288 pg/ml), that cause the statistical study to determine differences higher than a statistical significance of 0.005. An se in the number of replicas would improve the statistical significance.
The data obtained indicate an se in the production of IL-10, an anti-inflammatory cytokine, which corroborates the results obtained for TNFu, leading to the possibility that protein L19 triggers an anti-inflammatory cascade in skin explants, with an increased tion of IL- 1 0.
CONCLUSIONS In the cytotoxicity study, toxicity of protein L19 was very low under values of 25 ug/ml.
The choice of 25, 12.5 and 6.25 ug/ml to perform task 2 was based on cytotoxicity screening UV light radiation at 10 J/cm2 produces a significant inflammatory effect in skin explant samples not incubated with n L19 and significantly increases both TNFu levels and IL- levels (intrinsic anti-inflammatory reaction in skin).
Protein L19 causes a detectable reduction in the production of TNFu cytokine in the skin samples irradiated with UV light.
Protein L19 produces a general se in IL-10 levels in all the groups incubated with the protein, both as regards samples that undergo UV light radiation and those that do not.
According to the results obtained, the capacity of protein L19 to activate an anti- inflammatory effect is very high.
Example 3: IL-10 ed production of Lle9 and Lle9 derived es Objective of the study To determine which regions of L19 (linear epitopes) are capable to induce the production of 11-10 in spleen cultures.
Material Twenty four different peptides were designed and synthesized chemically. Each of them corresponds to a linear region of the ania major n L19 (i.e. SEQ ID NO: 1). Each peptides comprises 20 amino acids derived from Leishmania major L19 (i.e. SEQ ID NO: 1), except peptide 24 which only comprises 14 amino acids. Each peptide overlaps 9 amino acids with the previous peptide. 1: Peptide 1: SSSRHSFKQNETQN (SEQ ID NO: 31) : Peptide 2: SFKQNETQNMVSLKLQARLA (SEQ ID NO: 32) : Peptide 3: SLKLQARLASSILGCGRARV (SEQ ID NO: 33) : Peptide 4: ILGCGRARVWLDPNEAVEIQ (SEQ ID NO: 34) : Peptide 5: DPNEAVEIQNANSRKSVRKL (SEQ ID NO: 35) : Peptide 6: NSRKSVRKLIKDGFIIRKPV (SEQ ID NO: 36) : Peptide 7: DGFIIRKPVKVHSRARWRKM (SEQ ID NO: 37) QOONOKI‘I-hUJN : Peptide 8: HSRARWRKMKEAKDMGRHNG (SEQ ID NO: 38): Peptide 9: AKDMGRHNGVGRREGSREAR (SEQ ID NO: 39) : Peptide 10: RREGSREARMPSKELWMRRL (SEQ ID NO: 40) 11: Peptide 11: SKELWMRRLRILRRLLRKYR (SEQ ID NO: 41) 12: Peptide 12: LRRLLRKYRADKKIDRHVYR (SEQ ID NO: 42) 13: e 13: KKIDRHVYRDLYMRAKGNVF (SEQ ID NO: 43) 14: e 14: YMRAKGNVFRNKRNLVEHIH (SEQ ID NO: 44) : Peptide 15: KRNLVEHIHKIKNEKKKERQ (SEQ ID NO: 45) 16: Peptide 16: KNEKKKERQLAEQLAAKHLR (SEQ ID NO: 46) 17: Peptide 17: EQLAAKHLRDEQNRNKARKQ (SEQ ID NO: 47) 18: Peptide 18: QNRNKARKQELKKREKERER (SEQ ID NO: 48) 19: Peptide l9: KKREKERERARRDDAAAAAQ (SEQ ID NO: 49) : Peptide 20: RDDAAAAAQKKKADAAKKSA (SEQ ID NO: 50) 21: Peptide 2l: KADAAKKSAAPAAKSAAPAA (SEQ ID NO: 51) 22: Peptide 22: AAKSAAPAAKAAAPATKAAA (SEQ ID NO:52) 23: Peptide 23: AAPATKAAAAAPATKGAAPV (SEQ ID NO: 53) 24: e 24: PATKGAAPVKKSKK (SEQ ID NO: 54) Whole recombinant Leishmania major L19 (Lm L19) n (i.e. SEQ ID NO: l)expressed in Escherichia coli was used as control Spleen Cells from naive BALB/c mice (5 x 106 cells/ml) were stimulated (final volume 200 ul) with each dual peptide (100 , with a mixture of the 24 peptides (100 ug/ml), with the recombinant Leishmania major protein obtained from bacteria (12 ug/ml), and non- stimulated. After 48 h and 72 h supematants were collected and the amount of IL-lO was measured by ELISA as earlier described herein.
The recombinant Lle9 ed in bacteria was able to induce IL-lO synthesis in spleen cells from naive mice. None of the peptides were able to induce the same amount of IL-10 d by the protein, However, statistically significant amounts of 11-10 were induced by peptides l, 2, l2, l3, 14, 23 and 24.
Level of induced IL-lO was higher 72 h after stimulation in all cases.
Current experiments with peptides We are repeating the experiments with new synthetized es in order to confirm the results obtained. According to these results, we are using the same es but we have included 3 new peptides containing the sequences of the peptides which induce the highest Il- levels. The composition ofthese peptides are: : Peptide l-2: MTPLSLSSSRHSFKQNETQNMVSLKLQARLA (SEQ ID NO:55) 26: Peptide l2-l3-l4: LRRLLRKYRADKKIDRHVYRDLYMRAKGNVFRNKRNLVEHIH (SEQ ID NO:56) 27: Peptide: 23-24: AAPATKAAAAAPATKGAAPVKKSKK (SEQ ID NO:57) e 4: Animal model for the study of the anti-inflammatory effect of L19 Obj ective The ives of this study are: to test the anti-inflammatory effect of our active substance derived from Lrn L19 (i.e. SEQ ID NO:1) expressed in E. Coli and tested in ex Viyo cultures of intestinal tissue and; to investigate the anti-inflammatory effect in an animal model of Chron's disease.
Methodology To perform these objectives, two different steps will be taken: 1. In Vitro assays with mucosal explants from patients of Crohns disease, healthy controls and healthy samples where inflammation has been induced in Vitro with PMA-ionomycine. After 6 hours of incubation of samples with the active nce, supernatants will be ed for presence of Pro-inflammatory cytokines, regulatory cytokines and chemokines. such as TNFu, IL-lO, etc.
RNA will be extracted from the different tissues in order to analyse the expression of genes coding for Cytokines, Chemokines, Transcription factors and Inflammatory signals.
Tissues will be digested to obtain mucosal mononuclear cells, where the expression of some cell markers will be d in order to study the ent states of the dendritic cells after incubation. The lymphocyte population will be also investigated in order to determine the specificity of the response to the active substance.
In Viyo study of the anti-inflammatory effect of the active substance using murine inflammatory models in which chronic colitis is induced in the animals by the intake of drinking water with dextran sodium sulphate (DSS) for several days. Active substance will be ated subcutaneously in the animals before and after treatment with DSS. Different parameters such as quantitative evaluation of intestinal inflammation will be ed to te the intestinal lesion (14) to (25).
References 1. Zhou, X., et al, Current Drug Targets - Immune, Endocrine & Metabolic Disorders. (4), 2005, 5 2. Toshiyuki Y., et al, European Journal of Pharmacology. 533, 2006, 289-301 3. Weiss E., et al, Journal of the American Academy of ology, 50(5), 2004, 657-675 4. Wan-Wan L., et al, The l of Clinical Investigation 117(5), 2007, 1175-83.
. Wooley, Curr.Opin. Rheum. 3:407-20, 1999. 6. van Riel P.L.C.M., (2001), Best Practice & Research Clinical Rheumatology, 15: 67-76. 7. Gester A.M., (1999), Bailliere’s Clinical logy, 13: 629-644. 8. h et al. Intern. Rev. Immunol. 19:51-62, 2000. 9. Ann Rheum Dis 2005;64(Suppl II):ii65—ii68. doi: 10.1136/ard.2004.031237) 10. Hongbo Y., et al, Journal of Investigative Dermatology (2005) 125, 659—664. 11. Kirby B., et al, Br J Dermatol 2000;142:728—32. 12. Remington: The Science and Practice of Pharmacy, 20th n. ore, MD: Lippincott Williams & Wilkins, 2000). 13. Zhou X. Et al, (2005), Curr. Drug Targets Immune Endocr. Metabol. Disord., 5(4): 465-475. 14. Sartor R.B., Gastroenterology 2008; 134(2):577-94.
. BorruelN., et al, Gut 2002; 51: 659-664. 16. BorruelN., et al, Am J Gastroenterol 2003; 98: 865-870. 17. Carol M., et al, J Leukocyte Biol 2006; 79: 917-922. 18. Mufioz-Provencio D., et al, Arch Microbiol. 2008 Oct 31. 19. Llopis M., et al, Inflamm Bowel Dis. 2009; 15: 275-283.
. Lugea A., et al, Gut 2000; 46: 515-521. 21. Videla S., et al, Am J Gastroenterol 2001; 96: 1486-1493. 22. Medina C., et al, Scand J enterol 2001; 36: 1314-1319. 23. Medina C., et al, Am J Physiol 2003; 284: G116-G122. 24. Videla S., et al, J Pharmacol Exp Ther. 2006; 316: 940-945.
. Videla S., et al, Dig Dis Sci 2007; 52: 45-51.

Claims (13)

What we claim is:
1. Use of a nucleic acid molecule for the manufacture of a medicament for the prevention or treatment of an inflammatory disorder in an individual, wherein said nucleic acid molecule encodes a product that is able to induce the production of an anti-inflammatory response in a treated subject and comprises a nucleotide sequence selected from the group consisting of: i. nucleotide sequences encoding a polypeptide comprising an amino acid sequence that has at least 50% sequence identity with the amino acid sequence of SEQ ID NO:1, ii. nucleotide sequences comprising a nucleotide sequence that has at least 50% sequence identity with the nucleotide sequence of SEQ ID NO:2, iii. nucleotide sequences the complementary strand of which hybridizes to a nucleic acid molecule of ce of (i) or (ii) and iv. nucleotide sequences the sequences of which s from the sequence of a nucleic acid molecule of (iii) due to the degeneracy of the genetic code.
2. Use of a nucleic acid molecule according to claim 1, wherein said nucleic acid molecule encodes a t that is able to induce the tion of an antiinflammatory response in a treated subject and comprises a nucleotide sequence ed from the group consisting of: i. nucleotide sequences ng a ptide comprising an amino acid sequence that has at least 70% sequence identity with the amino acid sequence of SEQ ID NO:1, ii. nucleotide sequences comprising a nucleotide sequence that has at least 70% sequence ty with the nucleotide sequence of SEQ ID NO:2, iii. nucleotide sequences the complementary strand of which hybridizes to a nucleic acid molecule of ce of (i) or (ii) and iv. nucleotide sequences the sequences of which differs from the sequence of a nucleic acid molecule of (iii) due to the degeneracy of the genetic code.
3. Use of a ptide encoded by a nucleic acid molecule as identified in claim 1 or 2 for the manufacture of a medicament for the prevention or ent of an inflammatory disorder in an individual.
4. Use of a nucleic acid molecule or a polypeptide according to any one of claims 1 or 3, wherein said nucleic acid molecule or polypeptide derives from or originates from Leishmania major, Leishmania braziliensis, Leishmania infantum, Leishmania Mexicana or Leishmania donovani..
5. Use of a nucleic acid molecule according to claim 1, 2, or 4, wherein the c acid molecule is an oligonucleotide comprising at least 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more contiguous nucleotides of SEQ ID NO:2.
6. Use of a polypeptide according to claim 3 or 4, wherein the ptide is a protein fragment comprising at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 265, or 267 contiguous amino acids of SEQ ID NO:1.
7. Use of a protein fragment according to claim 6, comprising at least 14 contiguous amino acids of SEQ ID NO:1 and comprising SEQ ID NO: 31, 32, 55, 42, 43, 44, 56, 53, 54 and/or 57.
8. Use of a nucleic acid molecule or a polypeptide according to any one of claims 1 to 5, wherein the inflammatory er is toid arthritis (RA), juvenile rheumatoid arthritis, psoriasis, psoriatic arthritis, ankylosing spondylitis, inflammatory bowel disease including Crohn's disease or tive colitis, hepatitis, sepsis, alcoholic liver e, and coholic steatosis, sarcoidosis, autoimmune diabetes, diabetes mellitus, uveitis, multiple sclerosis, Controlling Allograft Rejection after organ transplantation, graft versus host disease (GVHD), inflammatory lung diseases including asthma and chronic ctive ary disease (COPD) (2), cancer (4) systemic lupus erythematosus, SLE, sarcoidosis, atopic dermatitis and cancer.
9. Use of a composition comprising at least a nucleic acid molecule and/or a polypeptide as defined in any one of preceding claims for the manufacture of a medicament for the prevention or treatment of an inflammatory disorder in an individual.
10. Use of a composition according to claim 9, wherein the composition is a pharmaceutical composition, said pharmaceutical composition comprising a pharmaceutically acceptable carrier, adjuvant, salt, diluent and/or excipient.
11. A use according to claim 1, ntially as herein described or exemplified.
12. A use according to claim 3, substantially as herein described or exemplified.
13. A use according to claim 9, substantially as herein described or exemplified.
NZ617454A 2011-05-09 2012-05-08 Molecule for treating an inflammatory disorder NZ617454B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161484167P 2011-05-09 2011-05-09
US61/484,167 2011-05-09
EP11165248 2011-05-09
EP11165248.3 2011-05-09
PCT/EP2012/058453 WO2012152792A1 (en) 2011-05-09 2012-05-08 Molecule for treating an inflammatory disorder

Publications (2)

Publication Number Publication Date
NZ617454A NZ617454A (en) 2015-09-25
NZ617454B2 true NZ617454B2 (en) 2016-01-06

Family

ID=

Similar Documents

Publication Publication Date Title
JP2000060580A (en) Homolog tl5 of tnf
JPH10327880A (en) Human sodium-dependent phosphate carrier (ipt-1)
JP2001509372A (en) Human F11 antigen, a cell surface receptor involved in platelet aggregation
JP2003024085A (en) Tumor necrosis-related receptor tr7
JPH11225774A (en) Member of immunoglobulin gene superfamily, pigr-1
JPH11177A (en) New members il1delta of il-1 family
JPH1142092A (en) Human cardiac/cerebral toroidal protein
JPH1118786A (en) Tumor necrosis related receptor tr7
JPH10313874A (en) Protein tr5 referred to tumor necrosis factor receptor
CA2835258C (en) Molecule for treating an inflammatory disorder
JPH1156376A (en) Human i kappa b beta
NZ617454B2 (en) Molecule for treating an inflammatory disorder
JPH11151094A (en) Member pigrl-1 of immunoglobulin gene super family
EP1002865A1 (en) Adipose specific protein
JP2002527037A (en) Cytokine family member EF-7
JP2002507413A (en) Members of the cytokine family, 2-21
JPH11206391A (en) Human lig-1 homologue (hlig-1)
JP2002513548A (en) Cytokine family members 2-19
JPH10327881A (en) New human gene similar to secretory mouse protein sfrp-1
JPH11215989A (en) Member of immunoglobulin gene super family, pigr-2
JPH11164693A (en) New smad3 splice mutant as target for chronic renal failure, atherosclerosis and fibrosis
WO1999022006A1 (en) CBLAFC02: A SUBUNIT OF VACUOLAR H(+)-ATPase
JPH11215988A (en) Human sdr2 cdna clone
JPH1132783A (en) Hfizg 53 polynucleotide and polypeptide
JP2002330793A (en) Hnbc1a being member of sodium carbonate co-transporter family