WO2005089080A2 - Method and kit for screening, pharmaceutical compositions and method for treatment of autoimmune disorders - Google Patents

Method and kit for screening, pharmaceutical compositions and method for treatment of autoimmune disorders Download PDF

Info

Publication number
WO2005089080A2
WO2005089080A2 PCT/SE2005/000414 SE2005000414W WO2005089080A2 WO 2005089080 A2 WO2005089080 A2 WO 2005089080A2 SE 2005000414 W SE2005000414 W SE 2005000414W WO 2005089080 A2 WO2005089080 A2 WO 2005089080A2
Authority
WO
WIPO (PCT)
Prior art keywords
expression
runx2
runx3
runxl
cells
Prior art date
Application number
PCT/SE2005/000414
Other languages
French (fr)
Other versions
WO2005089080A3 (en
Inventor
Marta E. ALARCÓN-RIQUELME
Ludmila Prokunina
Sergey Kozyrev
Original Assignee
Everygene Ab
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 Everygene Ab filed Critical Everygene Ab
Publication of WO2005089080A2 publication Critical patent/WO2005089080A2/en
Publication of WO2005089080A3 publication Critical patent/WO2005089080A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present invention relates to a method for screening and/or diagnosis of autoimmune disorders.
  • the invention also relates to a kit, a method for identifying compounds, that modulate PDCDl, RUNXl, RUNX2 or RUNX3 expression, a pharmaceutical composition comprising such a compound, the use of such a composition for the preparation of a pharmaceutical composition for the treatment of autoimmune diseases and a method for the treatment of a patient suffering from an autoimmune disease.
  • the invention especially relates to systemic lupus erythematosus (SLE) and rheumatoid arthritis.
  • SLE systemic lupus erythematosus
  • genes if not all have regulatory sequences located within non-coding regions, including promoters, 5' regulatory sequences, 3' regulatory sequences and introns. Within such sequences, there are binding sites for various transcription factors and repressors. Through those binding sites, trancription factors or repressor regulate expression of genes, which are therefore known as targets.
  • Three transcription factors in the human belong to the RUNX family sharing the runt domain, involved in regulation of a large number of targets, many known, but many as yet completely unknown (Choi et al. 2001; Cohen 2001; Javed et al. 2001; Backstrom et al. 2002; Burns et al. 2002; Eggers et al. 2002; Kundu et al.
  • RUNXl also called AML1, CBF ⁇ 2 or PEBP2 ⁇ B
  • RUNX2 also called AML3, CBF ⁇ 1 or PEBP2 ⁇ A
  • RUNX3 also called AML2, CBF ⁇ 3 or PEBP2 ⁇ C
  • RUNXl can be inactivated by translocations or point mutations, leading to acute myeloid and other types of leukemia and familial thrombocytopenia, functional insufficiency of RUNXl is also associated with the inability of myeloid cells to enter apoptosis (Lutterbach et al. 2000; Okuda et al. 2001; Michaud et al. 2002; Walker et al. 2002); mutations in RUNX2 (AML3) lead to arrest of osteoblast maturation and bone formation and have been found associated with cleidocranial dysplasia (CCD) (Leboy et al. 2001; Kundu et al. 2002; Otto et al.
  • CCD cleidocranial dysplasia
  • autoimmune diseases individuals have in their serum large quantities of antibodies (i.e. autoantibodies) reacting against such self-components.
  • autoimmune diseases are systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, psoriasis, Sj ⁇ gren's syndrome, insulin-dependent diabetes (also known as type 1 diabates mellitus or IDDM), myasthenia gravis, and primary biliary cirrhosis.
  • IDDM insulin-dependent diabetes
  • SLE patients The classification of SLE patients is based on the presence, at any time and in a cumulative fashion of at least 4 out of 11 criteria. Recently, manifestations related with the antiphospholipid syndrome were also added to the criteria to classify lupus, in 1997 (Hochberg 1997).
  • Systemic lupus erythematosus is mainly characterized by the presence of skin rashes (butterfly rash), or discoid lesions, arthralgias and arthritis, typically non-destructive but at times similar to that of rheumatoid arthritis, oral ulcers, alopecia (hair fall), inflammation of small blood vessels, inflammation of the kidney glomeruli (glomerulonephritis), lymphopenia and leucopenia, hemolytic anemia and thrombocytopenia.
  • Patients present with high levels of immunoglobulin and antibodies in their sera and with abnormal behaviour of their peripheral blood mononuclear cells. Lymphocytes of the B class are hyperactive and produce the high levels of antibodies, while Tcells are hyperreactive and produce various cytokines and interleukins.
  • Cytokines are soluble factors that can be produced by various differenTcell types depending on their function that stimulate or inhibit other cells activity at a distance or within localized compartments in secondary lymphoid organs.
  • cells of myeloid origin such as monocytes, macrophages or dendritic cells, also produce various substances that normally stimulate lymphoid cells.
  • These soluble factors are produced abnormally in systemic lupus erythematosus as well as in other autoimmune diseases (Sebbag et al. 1997; Feldmann et al. 1998; Magnusson et al. 2001; Prud'Appel et al. 2001).
  • the normal immune reaction includes first the appearance of the non-adaptive, also called innate immune system where monocytes and dendritic cells first encounter antigen.
  • the adaptive immune system is engaged, as the monocytes and dendritic cells initiate the interaction between the Tcell receptor in CD4+ or CD8+ cells with antigen presenting cells, showing small peptides within the peptide groove of the major histocompatibility complex (HLA) class I or class II molecules depending on the nature of the antigen.
  • HLA major histocompatibility complex
  • other molecules are accessory and help to amplify or inhibit the activating effect, proliferation or cell cycle entry of the cells as well as the production of the various cytokines (or lymphokines) is initiated.
  • There are several accessory molecules such as CD28 or CTLA-4.
  • CD28 is an amplifier of activation
  • CTLA-4 is an inhibitor of activation.
  • fii ⁇ RSl ⁇ TUTE SHEET (RULE 26) molecules may be involved.
  • PD-1 coded by a gene in chromosome 2q37.3 known as PDCDl.
  • the complete gene sequence (including previously unknown introns) of PDCDl is part of a previous patent application (US Application Serial No. 10/219,446).
  • PD-1 50-55kDa type I transmembrane receptor
  • PD-1 is an iinmuno-receptor tyrosine inhibitory motif-containing molecule expressed on early lymphocytes in thymus and bone marrow and in activated T and B cells.
  • PD-1 appears to inhibit lymphocyte activation by inhibiting proliferation at the Gl phase of the cell cycle and cytokine production without causing cell death, though an activating role has also been described (Shinohara et al. 1994; Agata et al. 1996; Finger et al. 1997; Nishimura and Honjo 2001; Carreno and Collins 2002; Greenwald et al. 2002).
  • PD-1 belongs to the immunoglobulin super-family, containing a single Ig-like variable domain in the extra-cellular region, while the cytoplasmic region contains an inhibitory motif (ITIM) known to recruiTcellular phosphatases and transmit in this fashion negative signals.
  • ITIM inhibitory motif
  • PD-1 has been shown to recruit the src homology 2-domain- containing tyrosine phosphatase 2 (SHP-2 or PTPN11) during inhibition of B cell receptor signalling (Okazaki et al. 2001). As all other proteins, the expression of PD-1 is dependent on the presence of transcription factors not only within the promoter region, but also through enhancers and silencers.
  • RUNXl As the binding site for the transcription factors RUNXl, RUNX2 and RUNX3 is shared between the three, any of these can be involved in the regulation of the PDCDl gene, it is required to know which of the RUNX genes regulates PDCDl and also their expression in autoimmune diseases. Not all RUNX proteins can be found in the same cell at the same time and if they do, there is strong competition for the binding site dependent on affinity, dose and quantity of each in the cellular milieu. Also, it is important to know if there are abnormalities in the tissue expression of these genes including also PD-1 or not in patients with SLE and in whaTcells this abnormal expression takes place.
  • PD-1, Runxl, Runx2 and Runx3 are differently expressed in Tcells from SLE patients than in healthy individuals.
  • PD1 is more expressed in CD4+ cells from SLE patients
  • Runxl more expressed in Tcells and Runx2 and Runx3 more expressed in Tcells and CD 4+ cells than in the corresponding cells from healthy persons (Example 3, tables 1 and 2).
  • Runxl and Runx2 are expressed in f ⁇ broblast synovial cells from patients with rheumatoid arthritis (Example 4).
  • the inventors have detected the cellular subpopulation where the gene expression is to be analyzed and the patterns of expression in various tissues for comparison.
  • the present invention relates to a method for screening and/or diagnosis of autoimmune diseases, such as rheumatic disorders and SLE based on the detection of levels of expression of PDCDl, RUNXl, RUNX2 and RUNX3. Further, the method detects the cellular subpopulation where the gene expression is to be analyzed and the patterns of expression in various tissues for comparison.
  • the invention also includes a method for the preparation of nucleic acids such as cellular RNA and DNA from specific cellular populations. Further, the invention includes a kit to detect cDNA in cells of patients with SLE or any other autoimmune disease. Further, the invention includes the detection of the cDNA expression of the gene alone or in combinations for the diagnosis of SLE or other related autoimmune conditions.
  • the invention includes a method for the follow-up of patients with SLE with various treatments, where levels of PDCDl, RUNXl, RUNX2 and/or RUNX3 can be detected and followed longitudinally. Further, the invention describes a method where the levels of PDCDl, RUNXl, RUNX2 and RUNX3 can be used for screening of therapeutic agents to treat or cure SLE that modify the expression of PDCDl, RUNXl, RUNX2 or RUNX3 or all of these combined or in variable combinations.
  • the invention also relates to a pharmaceutical composition, comprising at least one compound that modulate PDCDl, RUNXl, RUNX2 or RUNX3, the use of such a compound for the preparation of a pharmaceutical composition for the treatment of autoimmune diseases and a method for the treatment of a patient suffering from an autoimmune disease.
  • FIG. 1 Expression of PD-1 (PDCDl) and RUNX genes in tissues and blood fractions of normal controls. Prepared cDNAs from the cell subpopulations and tissues were obtained from CLONTECH.
  • RNA was prepared from the cells after harvesting at the points in time shown in the figure. As shown, TGFbeta induces strong expression of RUNXl and RUNX2, while hypoxia tends to reduce the levels, in particular after 4, 6 and 24 hours.
  • FIG. 7 a. Endogenous expression of the RUNX proteins in Jurkat Tcells. Interestingly RUNXl expression decreases when JurkaTcells are stimulated, b. PD-1 expression is conversely increased during Tcell stimulation. Over- expression of RUNXl reduces endogenous PD-1 expression in stimulated cells.
  • the invention relates to a method for screening and/or diagnosis of autoimmune disorders comprising the step of: a) obtaining biological sample from a test subject b) contacting the biological sample with at least one agent capable of detecting at least one of PDCDl, RUNXl, RUNX2 and/or RUNX3 nucleic acid or the corresponding expression product coded for by one or more of these genes or a sub fragment thereof, c) comparing the expression rate of at least one expression product from PDCD 1 , RUNXl , RUNX2 and/or RUNX3 nucleic acid or a subfragment thereof from a healthy test subject with the expression rate thereof from another test subject.
  • RUNXl, RUNX2 and/or RUNX3 and/or PDCDl or the expression products thereof may be analysed in the biological sample.
  • One or a combination of several of the above mentioned genes or their expression products may be analysed.
  • the biological sample may be chosen from peripheral blood mononuclear cells such as lymphocytes, especially T-lymphocytes or CD4+ cells.
  • lymphocytes are activated. This may be done e.g. with anti-CD3 antibodies, PMA (phorbol-12- myristate- 13 -acetate) and Ionomycin (I), PWM (pokeweed mitogen) or a combination thereof.
  • the nucleic acid which is detected may be RNA, such as cellular RNA, mRNA, or
  • the at least one agent may be a nucleic acid probe capable of hybridizing to any of
  • PDCD PDCD, RUNXl, RUNX2 or RUNX3 mRNA, cDNA or genomic DNA.
  • Suitable probes for use in the diagnostic assays of the invention are described herein.
  • the biological sample contains mRNA molecules from the test subject or genomic DNA.
  • the methods further involve obtaining control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting mRNA, cDNA or genomic
  • DNA in the biological sample and comparing the presence or absence or increased or decreased levels of mRNA in the test sample.
  • Usable probes may be a nucleic acid that comprises at least 20, at least 30, preferably at least 40 and most preferred at lest 50 nucleotides capable of hybridizing to any sequence chosen from sequences that differ between RUNXl, RUNX2 or RUNX3, or chosen from the PDCDl sequence.
  • RUNXl differentiates from the sequence of RUNX2 and RUNX3 at sequence SEQ
  • sequence of 135 bp is used covering nucleotides 1186-
  • No.4 primer RUNXlR In a still preferred embodiment, detection is improved using a probe described in SEQ.
  • the minimal sequence that can be detected comprises the minimal sequence of 1231-1281 bp, a nucleotide sequence of 50bp (SEQ No. 6) or also the nucleotide sequence comprising as minimal sequence 1391-1441 bp, as a contiguous sequence of 48 bp (SEQ No. 7).
  • the invention also describes a method for detection of variation in expression of the cDNA of the RUNX2 gene.
  • RUNX2 differentiates from the sequence of RUNXl and RUNX3 at SEQ No. 8, covering base pairs 721-1140 bp (based on Genebank sequence NM_004348).
  • sequence of bp 757-858, corresponding to amino acids 253-286, a total of 102 contiguous nucleotides is used (SEQ No. 9). This sequence is recognized and hybridizes the nucleotides from the kit known as primer RUNX2F (SEQ No. 10) and SEQ No. 11, known as Primer RUNX2R. In a preferred embodiment, detection is improved using a probe described in SEQ. No. 12.
  • the minimal sequence that can be detected comprises the minimal sequence of 791-841 bp (SEQ 13) comprised of 50 contiguous nucleotides or the sequence bp 951-1061 (junction over two exons) comprised by a contiguous sequence of 110 nucleotides (SEQ No. 14).
  • the invention also describes a method for detection of variation in expression of the cDNA of the RUNX3 gene specifically at a region covering a 340 bp sequence, SEQ no. 15 unique for RUNX3 gene from bp 581-921, a contiguous nucleotide sequence of 340bp (based on Genebank sequence: NM_004350).
  • the method detects and produces an amplicon of 125 contiguous nucleotides at the borders of exons 4 and 5, corresponding to bp 220-260, corresponding to amino acids 220-260 of the RUNX 3 gene (SEQ No.16).
  • detection is improved using a probe described in SEQ. No.
  • the minimal sequence that can be detected comprises the minimal sequence of 681-741 bp or a contiguous nucleotide sequence of 60 bp (SEQ No. 20).
  • the invention also describes a method for detection of variation in expression of the cDNA of the PDCDl gene specifically at a region that hybridizes any of the consecutive nucleotide sequences, SEQ no. 21, of 50 bp, SEQ NO. 22, of 50 bp, SEQ NO: 23, of 50 bp, SEQ NO. 24 of 50 bpbased and SEQ NO. 25 of 65 consecutive nucleotides (numbering is based on the Genbank sequence NM_005018) being these also the minimal sequences that can be detected.
  • This sequence is recognized and hybridizes the nucleotides from the kit known as SEQ NO. 26 PDlforw and SEQ NO: 27 PDlrev.
  • detection is improved using a probe described in SEQ. No. 28.
  • detection can be done using the polymerase chain reaction and detection using gel electrophoresis and staining with ethidium bromide.
  • detection can be performed using real-time quantitative PCR. Detection can be performed by labelling of the probes with SYBRgreen and the use of a laser based detector or with radioactive isotopes and the use of autoradigraphic films or a phosphoimager (Hernandez et al. 2003; Maeda et al. 2003; Von Samson- Himmelstjerna et al. 2003; Luthra et al. 2004).
  • the oligonucleotides are used for hybridization in microarrays.
  • the probe can be detected through hybridization and autoradiography.
  • the detection can be done using fluorescent labels such as FAM and TAMRA, or other amidites such as ROX, or other labels such as VIC, JOE, etc or other labels that can use fluorescent detection.
  • Preferred nucleic acid probes are, for detection of RUNXl those set forth in SEQ ID NO. 3, 4, 5; for detection of RUNX2 10, 11, 12; for detection of RUNX3, 17, 18, 19; and for PDCDl, SEQ NO. 26, 27 and 28 or a portion thereof.
  • the probe is labelled.
  • the autoimmune disorder that may be screened and /or diagnosed may be chosen from: a) multiple sclerosis, b) myasthenia gravis, c) Type 1 diabetes, d) rheumatoid arthritis, e) Sj ⁇ grens syndrome f) atopy, g) allergy, or
  • systemic lupus erythematosus where systemic lupus erythematosus (SLE) is characterised by conditions selected from the group including any one or more; fatigue, fever, loss of appetite, nausea, weight loss, hives, loss of scalp hair, red “butterfly rash” and raised rash, sensitivity to sun, ulcers in mouth, nose, or vagina, arthritis, joint pain, loss of blood supply to bone, pain, infections within joints, decrease in kidney function including, blood, aberrant amounts of protein or white blood cells in urine, intracerebral haemorrhage, headaches, loss of coordination, memory loss, seizures, strokes, anaemia, low white blood cell or low platelet count, pericardial effusion, heart attack, inflammation in the heart, infection in the heart, inflammation of the lining of the heart, infection of the lining of the heart, heart valve problems, shortness of breath, cough, inflammation of the lungs, inflammation of the lining of the lungs, abdominal distress, diarrho
  • SLE
  • the invention especially relates to the screening or diagnosing of SLE and rheumatoid arthritis.
  • SLE may be diagnosed when the rate of expression of PDCDl in CD4+ cells is higher in the tested subject that in a healthy test subject; when the rate of expression of RUNXl in T - cells is higher in the tested subject that in a healthy test subject ; when the rate of expression of RUNXl and/or RUNX3 in T - cells and/or CD4+cells is higher in the tested subject that in a healthy test subject.
  • Rheumatoid arthritis may be diagnosed when the rate of expression of RUNXl and 2 in synovial fibroblasTcells is modulated in the tested subject compared to a healthy test subject when subjected to hypoxia or to TGFbeta.
  • SLE may be diagnosed when PD-1 is at least 2 fold higher, especially at least 3 fold higher, preferably at least 4 fold higher and most preferred at least 4,5 fold higher in lymphocytes of SLE patients compared to controls.
  • lymphocytes such as Tcells, preferably CD4+cells are used.
  • the cells are preferably activated.
  • SLE may be diagnosed when RUNX2 is at least 2 fold higher, especially at least 3 fold higher, preferably at least 4 fold higher in lymphocytes of SLE patients compared to controls.
  • lymphocytes, such as Tcells, preferably CD4+cells are used.
  • the cells are preferably activated.
  • SLE may be diagnosed when RUNX3 is at least 2 fold higher, especially at least 3 fold higher in lymphocytes of SLE patients compared to controls.
  • lymphocytes such as Tcells, preferably CD4+cells are used.
  • the cells are preferably activated.
  • rheumatoid arthritis may be diagnosed when RUNXl and/or RUNX2 are at least 2 fold higher, especially at least 3 fold higher, preferably at least 4 fold higher in fibroblast synovial cells from patients with rheumatoid arthritis compared to controls.
  • kits for detecting the presence of PDCDl, RUNXl, RUNX2 and/or RUNX3 in a biological sample can comprise a labelled or unlabeled compound or agent capable of detecting PDCDl, RUNXl,
  • RUNX2 and/or RUNX3 mRNA cDNA or genomic DNA in a biological sample means for determining the amount in the sample, means of comparing the amounts in the sample with a standard and instructions for the use of the kit. Examples of how this may be done are to be found in the examples below.
  • the compound or agent can be packaged in a suitable container.
  • a kit comprising at least one, nucleic acid probe capable of hybridizing to mRNA, cDNA or genomic DNA from RUNXl, RUNX2,
  • the kit may comprise at least 20, preferably at least 30, such as at least 40, most preferred at least 50 nucleotides capable of hybridizing to any of SEQ ID NO 1, 2, 6,
  • the kit comprises at least one nucleic nucleic acid probe chosen from SEQ ID NO. 3, 4, 5; 10, 11, 12; 17, 18, 19; 26, 27 and 28 or a portion thereof.
  • the at least one probe is labelled, preferably as described above.
  • the kit can further comprise instructions for using the kit to detect PDCD 1 , RUNXl , RUNX2 and/or RUNX3 nucleic acid, preferably mRNA and instruction for the statistical comparison with a set standard as well as preparation of the standard.
  • the invention provides a method (also referred herein as screening assay) for identifying modulators, i.e. candidate or test compounds or agents that modify PDCDl, RUNXl, RUNX2 and/or RUNX3 expression, have an induction or reduction effect on their expression.
  • Such a screening assay comprises the step of: a) obtaining biological sample from a test subject b) contacting the biological sample with at least one agent capable of detecting the PDCDl, RUNXl, RUNX2 and/or RUNX3 nucleic acid or the corresponding expression product coded for by these gene or a subfragment thereof, c) optionally adding a compound to be tested for modulation d) determining the rate of expression of at least one of the PDCDl, RUNXl, RUNX2 and/or RUNX3 expression products e) determining if the compound has induction or reduction effect on their expression.
  • the invention provides assays for screening candidate or test compounds which alter or modulate the expression of the RUNX genes.
  • the compounds normalize the expression of the genes, making them candidates for treatment of systemic lupus erythematosus.
  • the test compounds of the present invention can be obtained using any of numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries, synthetic library methods using affinity chromatography selection, and other methods to peptide libraries, non-peptide oligomer or small molecule libraries of compounds (DeWitt et al. 1993; Lam et al. 1997).
  • Monitoring the ability of compounds to modulate the RUNX genes can be accomplished by monitoring in a cell-based assay the effect of the compound on the expression of the genes, by adding the compound in the cellular culture.
  • the compound can be tested in transfection assays, where any compound is added and expression of the RUNX genes is tested.
  • compounds that modulate Tcell activation can be identified by determining the ability of the compound to modulate expression of PDCDl, RUNXl, RUNX2 and RUNX3.
  • compounds that modulate myeloid activation can also be tested for their ability to modulate expression of PDCDl, RUNXl, RUNX2 and RUNX3.
  • the diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk of developing systemic lupus erythematosus or any disorder associated with aberrant or unwanted PDCDl, RUNXl, RUNX2, or RUNX3 activity, alone or in combination, known to be an autoimmune disease.
  • "aberrant” includes a PDCDl, RUNXl, RUNX2 and/or RUNX3 expression or activity which deviates from the wild-type PDCDl, RUNXl, RUNX2 and/or RUNX3 expression.
  • Aberrant expression includes increased, decreased expression or activity alone for each of the genes or in combination.
  • the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g. agonist, antagonist, peptidomimetic, peptide, nucleic acid, small molecule or other drug or candidate) to treat an autoimmune disease or disorder with aberrant expression of PDCDl, RUNXl, RUNX2 and/or RUNX3.
  • agent e.g. agonist, antagonist, peptidomimetic, peptide, nucleic acid, small molecule or other drug or candidate
  • PDCDl e.g. agonist, antagonist, peptidomimetic, peptide, nucleic acid, small molecule or other drug or candidate
  • an agent e.g. agonist, antagonist, peptidomimetic, peptide, nucleic acid, small molecule or other drug or candidate
  • autoimmune disorder including systemic lupus erythematosus, rheumatoid arthritis, or other disorder known in the art to be of autoimmune nature.
  • the methods of the invention can also be used to detect genetic alterations in the RUNXl, RUNX2 and/or RUNX3 gene, thereby determining if a subject with the altered gene is at risk for a disorder characterized by missregulation of RUNXl, RUNX2 and or RUNX3 in particular if this is an autoimmune disorder.
  • the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic alteration characterized by mis-expression of PDCDl, RUNXl, RUNX2 and/or RUNX3, in particular in autoimmune disorders.
  • Monitoring the influence of agents (e.g. drugs) on the expression or activity of PDCDl, RUNXl, RUNX2 and/or RUNX3 can be applied not only to basic drug screening, but also in clinical trials.
  • agents e.g. drugs
  • the effectiveness of an agent determined by a screening assay as described herein to increase or decrease PDCDl, RUNXl, RUNX2 and/or RUNX3 expression can be monitored in clinical trials for subjects with systemic lupus erythematosus or other autoimmune disease exhibiting increased, decreased or aberrant levels or pattern of expression of the RUNXl, RUNX2 and/or RUNX3 genes alone or in combination with or without the expression of PDCDl.
  • genes including PDCDl, RUNXl, RUNX2 and RUNX3, that are modulated in cells by treatment with an agent (e.g. compound, drug or small molecule) which modulate PDCDl, RUNXl, RUNX2 and/or RUNX3 activity can be identified.
  • an agent e.g. compound, drug or small molecule
  • cells can be isolated and RNA prepared and analyzed for the levels of expression of PDCD 1 , RUNXl , RUNX2 and/or RUNX3 alone or in combination.
  • the levels of gene expression e.g.
  • a gene expression pattern can be quantified by Northern blot analysis or RT-PCR as described herein, by one of the methods described herein.
  • the gene expression can serve as a marker, indicative of the physiological response of the cells to the agent. Accordingly, this response state may be determined before, and at various points during treatment of the individual with the agent.
  • the present invention provides a method for monitoring the effectiveness of treatment of a subject with an agent including a pre-administration sample from a subject prior to administration of the agent; detecting the expression of PDCDl, RUNXl, RUNX2 and/or RUNX3 in the preadministration sample; obtaining one or more postadministration samples and; measuring expression of the genes in the postadministration samples; comparing the levels of expression in the preadministration sample or postadministration sample; and altering the administration of the agent to the subject accordingly.
  • PDCDl, RUNXl, RUNX2 and/or RUNX3 expression alone or in combination may be used as an indicator of the effectiveness of the agent, even in the absence of an observable phenotypic response.
  • compositions and treatment are provided.
  • the invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound that modulates PDCD 1 , RUNXl , RUNX2 or RUNX3 expression identified by the step of: a) obtaining biological sample from a test subject b) contacting the biological sample with at least one agent capable of detecting the PDCDl, RUNXl, RUNX2 and/or RUNX3 nucleic acid or the corresponding expression product coded for by these gene or a subfragment thereof, c) optionally adding a compound to be tested for modulation d) determining the rate of expression of at least one of the PDCD 1 , RUNXl, RUNX2 and/or RUNX3 expression products e) determining if the compound has induction or reduction effect on their expression.
  • the compound is an agonist, antagonist to the expression products of PDCDl, RUNXl, RUNX2 or RUNX3, such as peptidomimetics, peptides, nucleic acids such as anti sense DNA, antisense RNA, such
  • SUBSTITUTE feT (RULE 2 as siRNA or ribozyme, small molecules or drugs, antibodies, including mouse or humanized antibodies.
  • the invention includes the construction of a pharmaceutical compound used to over express the cDNA and the protein for PDCDl, RUNXl, RUNX2 or RUNX3 in cases where the expression of these genes is deficient. They may be pair of nucleotide primers that over express the cDNA of the genes. The compounds may be used as gene therapy. Especially the invention relates to compounds, such as nucleotides that over express RUNXl and RUNX3 such as the primers SEQ ID NO 29, 30 and 31, 32.
  • the oligonucleotides from SEQ. 29 and 30 are used to amplify the full-length cDNA that produces the peptide known as the AML-lc isoform of RUNXl shown in SEQ. NO. 31 of 1464bp including a 4bp sequence CACCT at the beginning used to clone in the vector.
  • the oligonucleotides from SEQ. 32 and 33 are used to amplify the full-length cDNA that produces the peptide for RUNX3 shown in SEQ. NO. 34 of 1258bp including a 4bp sequence CACCT at the beginning used to clone in the vector.
  • the nucleotides may be used with a vector that is introduced onto cells.
  • the term "vector” refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • One type of preferred vector is an episome, i.e., a nucleic acid capable of extra-chromosomal replication.
  • Preferred vectors are those capable of autonomous replication and/or expression of nucleic acids to which they are linked.
  • Vectors capable of directing the expression of genes to which they are operatively linked are referred to herein as "expression vectors".
  • expression vectors of utility in recombinant DNA techniques are often in the form of "plasmids" which refer generally to circular double stranded DNA loops which, in their vector form are not bound to the chromosome.
  • plasmid and “vector” are used interchangeably as the plasmid is the most commonly used form of vector.
  • the invention is intended to include such other forms of expression vectors which serve equivalent functions and which become known in the art subsequently hereto.
  • the invention is intended to include all forms of expression vectors including replication defective retroviruses, adenoviruses and adeno-associated viruses and lentiviruses.
  • One type of vector used is a plasmid, which refers to a circle double stranded DNA loop into which additional DNA segments can be ligated. Certain vectors are capable of directing the expression of genes.
  • the invention further includes pharmaceutical compositions, which comprise an expression vector comprising at least one DNA molecule of PDCDl, RUNXl, RUNX2 or RUNX3 cloned into the expression vector in an antisense orientation.
  • the DNA molecule may be operatively linked to a regulatory sequence in a manner, which allows for expression of an RNA molecule, which is antisense to PDCDl, RUNXl, RUNX2 or RUNX3 mRNA.
  • the system includes the use of RNA interference technology where persistent expression of a silencer RNA molecule known as siRNA, is allowed in order to silence the gene desired. This siRNA is an antisense of PDCDl, RUNXl, RUNX2 or RUNX3 mRNA or a subfragment of PDCDl, RUNXl, RUNX2 or RUNX.
  • the invention especially relates to the nucelotides SEQ ID 35, 36 and 37 that target the positions of the RUNXl gene at 505-525 (SEQ. NO. 35), 959-978 (SEQ. NO. 36) and 1242-1263 (SEQ. NO. 37).
  • a lentiviral vector may be used.
  • the ideal vector for gene therapy should be first of all safe, non-immunogenic, non-toxic, non-allergic, enabling ease, efficient and stable gene transfer to targeTcells, which are often non-replicating and terminally differentiated. Such vector should have a satisfactory cloning capacity.
  • LW Longer-based Vector
  • Lentiviruses are a class of retroviruses that can
  • Self-inactivating lentiviruses with all deleted viral genes can accommodate about 8.5kb of DNA fragment. This is highly desirable, as some therapeutic genes together with regulatory elements may be long.
  • Viral particles are packed in a special cell line with help of three independent non-overlapping split- genome packaging constructs, two of them expressing HIV proteins necessary for vector propagation and a third expressing heterologous envelope (vesicular stomatitis virus glycoprotein, VSVG) protein.
  • the major feature of self-inactivating viral vectors is a deletion in the U3 region of the 3'- LTR, making them incapable to produce packageble viral genomes after integration into the host genomic DNA increasing biosafety.
  • lupus Since lupus is characterized by reiterated periods of flares and remissions, it is important to have systems allowing regulation of expression of a therapeutic gene at the proper time. Up-regulation of a gene caused by a single drug administration will ameliorate disease and improve the health of the patient. First of all such regulated system should not influence the transcription of endogenous genes and provide low- level (ideally undetectable) basal activity and rapid induction of a drug dose-response. The system must consist of artificial promoters and regulator proteins to prevent interference with cellular processes.
  • the important part of the regulated system is the inducer that should not be toxic, do not cause adverse effects (i.e. be fully physiologically inert compound) and be preferably orally bio-viable to ease treatment.
  • adverse effects i.e. be fully physiologically inert compound
  • Rapamycin in the dimerization-based regulation system interferes with cellular signaling through inhibition of evolutionary conserved signal transduction pathways and cause immunosuppressive effects (Fruman et el. 1994).
  • Ecdysone is quite complicated and overexpression of one of the regulator protein RXRa may exert pleiotropic effects because of its human origin (No et el. 1996, Bugge et al.
  • Tet-system has serious drawbacks such as high basal level activity of the therapeutic gene (tet-on system, Freundlich et al. 1999, Urlinger et al. 2000, Lamartina et al. 2002).
  • the tet-off system is not suitable for long-term applications since un-induced state is maintained by continuous treatment with Tet or Dox.
  • the expression of the gene is activated only after withdrawal of drug. Best suited for testing different gene therapy approaches is the last of the modifications of the RU486-dependent regulated system, called GeneSwitch (Abruzzese et al. 2000).
  • the system includes a chimeric regulator protein and an autoinducible promoter consisting of four copies of Gal4 binding sites linked to a minimal thymidine kinase promoter.
  • the promoter/enhancer for the therapeutic gene is composed of six copies of Gal4 sites followed by the TATA sequence from the Adenoviral Elb promoter.
  • the regulatory GeneSwitch protein contains the following functional domains: a) Mutated human progesterone receptor ligand binding domain (LPR-LBDDdelta) capable of changing its conformation upon binding to the progesterone antagonist, mifepristone (RU486); b) Yeast Gal4 DNA binding domain (Gal4-DBD); c) Transcription activation domain from the human NF-kB transcription factor p65 (p65AD). Expression of the regulator as well as the transgene is ligand-dependent and inducible. In normal, un-induced states the system provides weak expression of the inactive regulator. After a single oral or i.p.
  • mifepristone RU486
  • the concentration of mifepristone needed for maximal induction of gene expression 250-350 mg/kg is far below the levels at which mifepristone has antiprogesterone and antiglucocorticoid activity (Abruzzese et al. 200).
  • the advantage is that the recombinant regulatory protein is synthesized at high levels only when mifepristone is administered.
  • the maximum level of expression varies in different models and for different transgenes ranging from 25% to 80% of the CMV-driven expression but it is high enough to achieve therapeutic effect.
  • the GeneSwitch system is preferred.
  • the pharmaceutical compounds described herein can be used to screen cells from various individuals or cell lines and study how the overexpression or silencing of the RUNX genes modulate the expression of other molecules, genes or peptides. This is also done using stable or temporary transfection for which an example is provided.
  • the invention also relates to the use of such a compound for the preparation of a pharmaceutical composition for the treatment of the above mentioned diseases or disorders.
  • an effective amount means a therapeutically effective amount of a compound or composition large enough to modify the symptoms and/or condition to be treated, but small enough to avoid serious side effects (at a reasonable benefit/risk ratio), within the scope of sound medical judgment.
  • the effective amount of active ingredient for use in the pharmaceutical compositions and the methods of the invention herein will vary depending upon the severity of the diseases, the duration of the treatment, the nature of concurrent therapy, the particular active ingredient being employed, the particular pharmaceutically-acceptable excipients utilized, and like factors within the knowledge and expertise of the attending physician.
  • the dosage may range from about 0.0001 to lg/kg, such as 0,01 mg to 500 mg/kg and more usually 0.1 to 5 lOOmg/kg, e.g. 0,5-50 mg of the host body weight.
  • Doses for nucleic acids may range from about 10 ng to lg, 100 ng to 100 mg, 1. ⁇ g to
  • pharmaceutically-acceptable excipients includes any physiologically inert, pharmacologically inactive material known to one skilled in the art, which is compatible with the physical and chemical characteristics of the particular azapeptide compound active ingredient selected for use.
  • excipients include, but are not limited to, polymers, resins, plasticizers, fillers, binders, lubricants, glidants, disintegrates, solvents, co-solvents, buffer systems, surfactants, preservatives, sweetening agents, flavouring agents, pharmaceutical grade dyes or pigments, and viscosity agents.
  • the invention relates to a method for the treatment of a patient suffering from an autoimmune disease wherein a compound that is an agonist, antagonist to the expression products of PDCDl, RUNXl, RUNX2 or RUNX3, such as peptidomimetic, peptide, nucleic acid such as anti sense DNA or RNA is administrated.
  • a compound that is an agonist, antagonist to the expression products of PDCDl, RUNXl, RUNX2 or RUNX3, such as peptidomimetic, peptide, nucleic acid such as anti sense DNA or RNA is administrated.
  • the present invention is used to detect variable levels of PDCD 1 , RUNXl , RUNX2 and/or RUNX3 in normal individuals and in patients with SLE, rheumatoid arthritis or other diseases of the immune system including autoimmune diseases.
  • the kit described here allows for the detection of levels of each gene individually or in combination forming a pattern characteristic of patients with SLE as compared to normal individuals.
  • Peripheral blood mononuclear cells are purified from heparin-treated blood samples by Fycoll-hypaque gradient centrifugation (Bouyant density gradients). For further separation of cell types, T and non-Tcells are separated using sheep red blood cells and 95% purity of separated Tcells is obtained, as determined with anti-CD3 antibodies, a specific peripheral Tcell marker.
  • total PBMC are fractionated using cell sorting (FACS) where PBMC are incubated with antibodies to CD14, CD19, CD4 and CD8, conjugated with fluorescent dyes (antiCD14-PE, antiCD19-FITC, antiCD4-APC and antiCD8-PC5) for 20 min on ice, washed and subjected to FACS separation. The purity of each population is confirmed by FACS analysis and is not less than 95%.
  • FACS cell sorting
  • cells are activated by a combination of PMA (phorbol-12-myristate- 13 -acetate, 20ng/ml) and Ionomycin (I) (0.5mM) (Sigma) and incubated for 2, 4 and 6 hours.
  • PMA phorbol-12-myristate- 13 -acetate, 20ng/ml
  • I Ionomycin
  • non-Tcells or CD 19+ cells are activated by PMA/I for 4 hours or PWM (pokeweed mitogen) for 3 days.
  • RNA is extracted using Trizol reagent (Life Technologies) and cDNAs is prepared with TaqMan Reverse Transriptase Kit (PE Biosystem). The amount of cDNA generated is measured using the TaqMan assays with a 7700 ABI PRISM sequence detector system (PE Biosystem). TaqMan primers and probes are designed with Primer Express Software (PE Biosystem) so that they covered exon junctions to assure for the sole amplification of cDNA and none of genomic DNA. All samples are run at least twice and mean values are used for calculations. Standard curves are constructed for all assays from serial dilutions of cDNA from the JurkaTcell line. Standard curves are used to calculate the level of expression of all the genes according to the manufacturers instructions.
  • Absolute values are calculated as the proportion of the amount of transcript of the query genes normalized to the amount of transcript of endogenous control.
  • Two endogenous controls: glyceraldehydephosphate dehydrogenase (GAPDH) and B2-microglobulin (B2M) are run for each sample in duplicates and Ct values are calculated.
  • GPDH glyceraldehydephosphate dehydrogenase
  • B2M B2-microglobulin
  • RNA extracted from non-treated non-Tcells and activated Tcells (30 control samples in total) and cDNAs were diluted so that 100 ng of total RNA is used for each RT- PCR reaction.
  • the mean Ct value for GAPDH is 23.7 +/- 2.17 with coefficient of variation of 9.2%.
  • the mean Ct value is 21.0+/- 0.82 and coefficient of variation of 3.9%. Based on these calculations B2M is a more stable endogenous control for the study and used to calculate the results from the various experiments.
  • the invention provides a method to detect mRNA from PDCDl, RUNXl, RUNX2 and RUNX3 alone or in combination in cells cultured in vitro and stimulated with various substances.
  • PBMC of 5 controls were treated by PMA/I for 2, 4 and 6 hours and compared with non-treated samples.
  • PD-1 expression is not detected in resting B-cells (cell line Daudi, peripheral blood B cells, bone marrow, tonsils) or myeloid cells (cell lines THP-1, U937, K562, KM3 and peripheral CD 14+ cells).
  • Activation of B cells by PMA/I for 4 hours or by PWM for 4 days and activation of monocytes by PMA/I for 4 hours does not induce PD-1 expression.
  • PD-1 expression is observed in the Molt 4 cell line (representing CD4+CD8+ thymic cells) and in cDNA from the thymus, spleen and lymph nodes.
  • the low expression is increased by 3-4 fold by PMA/I activation in peripheral blood CD4+ cells and in the Jurkat CD4+ cell line. Expression is not detected in either resting or activated peripheral blood CD8+ cells and the HSB2 cell line (CD8+).
  • RUNX3 was also detected in resting and activated CD4+ cells and B cells, but was not present in myeloid cells ( Figure l,d).
  • Figure 2 We decided to clarify further which blood fractions contribute to this difference.
  • There was no difference of the PD-1 expression in non-Tcells (resting and activated B cells and monocytes) of patients compared to controls, while expression was 4.5 fold higher in activated Tcells of patients (P 0.00004) as compared with activated Tcells from controls.
  • the difference in expression was due to increased PD-1 expression in activated CD4+ cells of patients, while expression in resting and activated CD8+ cells of both patients and controls was not detectable.
  • patients with SLE have an increased expression of PD-1 in activated CD4+ cells ( Figure 2).
  • the levels of PDCDl, RUNXl, RUNX2 and/or RUNX3 can be compared to those of normal controls using statistical methods such as non-paired Student' s T test and determination of a p value.
  • synovial fibroblast-like cells were obtained through arthroscopy from the knee joint of patients with rheumatoid arthritis.
  • the cells were grown in culture in standard conditions or were subjected to hypoxa or to tumor growth factor beta (TGFbeta). After 2, 4, 6 or 24 hours the cells were harvested and RNA was prepared using described methodology.
  • TGFbeta tumor growth factor beta
  • the kit described herein for RUNXl, RUNX2 and RUNX3 the expression of these genes was determined.
  • RUNX3 and PDCDl were not detected, while changes were observed with RUNXl and RUNX2. Overall the levels of RUNX2 were higher than for RUNXl.
  • the full-length cDNA sequence of RUNXl is amplified using the oligonucleotides described in SEQ. 29 and 30 (that recognize nucleotides 425-447 and 1868-1887 in the sequence NM_001754 and amplify the portion known as AML-lc described in SEQ. 33). That for RUNX3 is obtained by PCR using the oligonucleotides described in SEQ. No. 31 and 32 (and recognize positions 1-24 and 1237-1258 in the sequence NM_004350 and amplify the portion described in SEQ. 34). Both and cloned into ⁇ cDNA3.1D/V5-His-TOPO vector (Invitrogen, K 4900-01).
  • JurkaTcells were transfected with LipofectamineTM 2000 (Invitrogen) in 24-well plates according to the manufacturer's protocol. Briefly, 0.35xl0 6 cells were seeded in each well in antibiotic-free RPMI medium supplemented with 10% FBS. 0.4 ⁇ g of the pcDNA vector carrying either of the Runx genes or empty vector for negative control, were incubated in 50 ⁇ l of Opti-MEM medium (GIBCO) for 5 min. Then diluted DNA was combined with diluted in 50 ⁇ l of Opti-MEM Lipofectamine 2000, incubated at room temperature for 20 min and added to the cells. Cells were incubated at 37°C in a C0 2 incubator for 48 hours.
  • Opti-MEM medium GIBCO
  • THP-1 and Daudi cells were washed once in PBS and once in RPMI medium and resuspended in 750 ⁇ L of RPMI medium.
  • 2.5 ⁇ g of plasmid DNA were mixed with 50 ⁇ g DEAE/Dextran in 250 ⁇ l RPMI medium and incubated at RT for 10 min before it was added to the cells.
  • THP-1 Human monocytic leukemia cells
  • Jurkat human Tcell leukemia cells
  • Daudi Burkitt's lymphoma cells
  • GBCO RPMI 1640 media
  • FBS fetal bovine serum
  • Peptomycin 100 ⁇ g/mL
  • the medium was replaced every third day.
  • THP-1 cells, JurkaTcells and Daudi can be obtained from The American Type Culture Collection (ATCC).
  • Nishimura H, Honjo T (2001) PD-1 an inhibitory immunoreceptor involved in peripheral tolerance. Trends Immunol 22:265-268 No D, Yao TP, Evans RM. Proc Natl Acad Sci U S A 93:3346-51, 1996 Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T (2001) PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine.
  • Prokunina L Castillejo-Lopez C, Oberg F, Gunnarsson I, Berg L, Magnusson V, Brookes AJ, Tentler D, Kristjansdottir H, Grondal G, Bolstad Al, Svenungsson E, Lundberg I, Sturfelt G, Jonssen A, Truedsson L, Lima G, Alcocer-Varela J, Jonsson R, Gyllensten UB, Harley JB, Alarcon-Segovia D, Steinsson K, Alarcon-Riquelme ME (2002) A regulatory polymorphism in PDCD 1 is associated with susceptibility to systemic lupus erythematosus in humans.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Pathology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The present invention relates to a method for screening and/or diagnosis of autoimmune disorders comprising the step of : a) obtaining biological sample from a test subject b) contacting the biological sample with at least one agent capable of detecting at least one of PDCD1, RUNX1, RUNX2 and/or RUNX3 nucleic acid or the corresponding expression product coded for by one or more of these genes or a subfragment thereof. d) comparing the expression rate of at least one expression product from PDCD1, RUNX1, RUNX2 and/or RUNX3 nucleic acid or a subfragment thereof from a healthy test subject with the expression rate thereof from another test subject. The invention also relates to a kit, a method for identifying compounds, that modulate PDCD1, RUNX1, RUNX2 or RUNX3 expression, a pharmaceutical composition comprising such a compound, the use of such a composition for the preparation of a pharmaceutical composition for the treatment of autoimmune diseases and a method for the treatment of a patient suffering from an autoimmune disease. The invention especially relates to systemic lupus erythematosus (SLE) and rheumatoid arthritis.

Description

Method and kit for screening, pharmaceutical compositions and method for treatment of autoimmune disorders
The present invention relates to a method for screening and/or diagnosis of autoimmune disorders. The invention also relates to a kit, a method for identifying compounds, that modulate PDCDl, RUNXl, RUNX2 or RUNX3 expression, a pharmaceutical composition comprising such a compound, the use of such a composition for the preparation of a pharmaceutical composition for the treatment of autoimmune diseases and a method for the treatment of a patient suffering from an autoimmune disease. The invention especially relates to systemic lupus erythematosus (SLE) and rheumatoid arthritis.
Background of the Invention
Many genes if not all have regulatory sequences located within non-coding regions, including promoters, 5' regulatory sequences, 3' regulatory sequences and introns. Within such sequences, there are binding sites for various transcription factors and repressors. Through those binding sites, trancription factors or repressor regulate expression of genes, which are therefore known as targets. Three transcription factors in the human belong to the RUNX family sharing the runt domain, involved in regulation of a large number of targets, many known, but many as yet completely unknown (Choi et al. 2001; Cohen 2001; Javed et al. 2001; Backstrom et al. 2002; Burns et al. 2002; Eggers et al. 2002; Kundu et al. 2002; Lund and van Lohuizen 2002; Perry et al. 2002; Strieker et al. 2002; Barnes et al. 2003; Bristow and Shore 2003; Cameron et al. 2003; Coffman 2003; Komori 2003; Kundu and Liu 2003; Lian et al. 2003; Otto et al. 2003). These act through DNA-binding to a common consensus sequence and through heterodimerization with the common subunit CBFα (also called PEBP2α): RUNXl (also called AML1, CBFα 2 or PEBP2α B), RUNX2 (also called AML3, CBFα 1 or PEBP2α A) and RUNX3 (also called AML2, CBFα 3 or PEBP2α C) (Adya et al. 2000; Bushweller 2000; Backstrom et al. 2001; Backstrom et al. 2002; Cameron et al. 2003). RUNX proteins can bind the same target sequence TG(T/C)GGT and perform activation or inhibition of transcription depending on the
±. presence of other interacting proteins (Adya et al. 2000; Bushweller 2000; Backstrom et al. 2001; Backstrom et al. 2002; Cameron et al. 2003). Whether one or the other of the RUNX proteins bind to the sequence is a matter of relative amounts of each in a given cell at a given time (Choi et al. 2001; Harrington et al. 2002). In general, inactivation, or insufficiency of the RUNX proteins leads to increased proliferation of undifferentiated cells. RUNXl can be inactivated by translocations or point mutations, leading to acute myeloid and other types of leukemia and familial thrombocytopenia, functional insufficiency of RUNXl is also associated with the inability of myeloid cells to enter apoptosis (Lutterbach et al. 2000; Okuda et al. 2001; Michaud et al. 2002; Walker et al. 2002); mutations in RUNX2 (AML3) lead to arrest of osteoblast maturation and bone formation and have been found associated with cleidocranial dysplasia (CCD) (Leboy et al. 2001; Kundu et al. 2002; Otto et al. 2002; Komori 2003; Lian and Stein 2003; McCarthy et al. 2003); inactivation (loss-of-function) of the RUNX3 (AML2) gene has been associated with hyperproliferation of gastric epithelial cells and their failure to differentiate, leading to gastric cancer (Moss 2003). It appears that the role of different RUNX proteins in disease is similar - depending on time and level of their expression and interacting partners. These define whether the cells proliferate, undergo terminal differentiation or die. The immune system recognizes foreign molecules and initiates an immune response against these. At times, as in autoimmune diseases, the response is against self- components, such as membrane phospholipids, nuclear proteins, components of the splicing or ribosomal complexes or even against double stranded DNA. In autoimmune diseases, individuals have in their serum large quantities of antibodies (i.e. autoantibodies) reacting against such self-components. Examples of autoimmune diseases are systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, psoriasis, Sjδgren's syndrome, insulin-dependent diabetes (also known as type 1 diabates mellitus or IDDM), myasthenia gravis, and primary biliary cirrhosis. Systemic lupus erythematosus, a systemic disease is diagnosed using the criteria from the American College of Rheumatology Criteria established in 1982 (Tan et al. 1982). The classification of SLE patients is based on the presence, at any time and in a cumulative fashion of at least 4 out of 11 criteria. Recently, manifestations related with the antiphospholipid syndrome were also added to the criteria to classify lupus, in 1997 (Hochberg 1997). Systemic lupus erythematosus is mainly characterized by the presence of skin rashes (butterfly rash), or discoid lesions, arthralgias and arthritis, typically non-destructive but at times similar to that of rheumatoid arthritis, oral ulcers, alopecia (hair fall), inflammation of small blood vessels, inflammation of the kidney glomeruli (glomerulonephritis), lymphopenia and leucopenia, hemolytic anemia and thrombocytopenia. Patients present with high levels of immunoglobulin and antibodies in their sera and with abnormal behaviour of their peripheral blood mononuclear cells. Lymphocytes of the B class are hyperactive and produce the high levels of antibodies, while Tcells are hyperreactive and produce various cytokines and interleukins.
Cytokines are soluble factors that can be produced by various differenTcell types depending on their function that stimulate or inhibit other cells activity at a distance or within localized compartments in secondary lymphoid organs. In addition, cells of myeloid origin, such as monocytes, macrophages or dendritic cells, also produce various substances that normally stimulate lymphoid cells. These soluble factors are produced abnormally in systemic lupus erythematosus as well as in other autoimmune diseases (Sebbag et al. 1997; Feldmann et al. 1998; Magnusson et al. 2001; Prud'homme et al. 2001). The normal immune reaction includes first the appearance of the non-adaptive, also called innate immune system where monocytes and dendritic cells first encounter antigen. In a posterior phase, the adaptive immune system is engaged, as the monocytes and dendritic cells initiate the interaction between the Tcell receptor in CD4+ or CD8+ cells with antigen presenting cells, showing small peptides within the peptide groove of the major histocompatibility complex (HLA) class I or class II molecules depending on the nature of the antigen. During such an interaction, other molecules are accessory and help to amplify or inhibit the activating effect, proliferation or cell cycle entry of the cells as well as the production of the various cytokines (or lymphokines) is initiated. There are several accessory molecules such as CD28 or CTLA-4. For ex. CD28 is an amplifier of activation, while CTLA-4 is an inhibitor of activation. However at various levels of the immune response several other
fiiΪRSlΪTUTE SHEET (RULE 26) molecules may be involved. Such is the case of PD-1, coded by a gene in chromosome 2q37.3 known as PDCDl. The complete gene sequence (including previously unknown introns) of PDCDl is part of a previous patent application (US Application Serial No. 10/219,446). PD-1 (50-55kDa type I transmembrane receptor) is an iinmuno-receptor tyrosine inhibitory motif-containing molecule expressed on early lymphocytes in thymus and bone marrow and in activated T and B cells. PD-1 appears to inhibit lymphocyte activation by inhibiting proliferation at the Gl phase of the cell cycle and cytokine production without causing cell death, though an activating role has also been described (Shinohara et al. 1994; Agata et al. 1996; Finger et al. 1997; Nishimura and Honjo 2001; Carreno and Collins 2002; Greenwald et al. 2002). PD-1 belongs to the immunoglobulin super-family, containing a single Ig-like variable domain in the extra-cellular region, while the cytoplasmic region contains an inhibitory motif (ITIM) known to recruiTcellular phosphatases and transmit in this fashion negative signals. PD-1 has been shown to recruit the src homology 2-domain- containing tyrosine phosphatase 2 (SHP-2 or PTPN11) during inhibition of B cell receptor signalling (Okazaki et al. 2001). As all other proteins, the expression of PD-1 is dependent on the presence of transcription factors not only within the promoter region, but also through enhancers and silencers.
In a previous patent, such an enhancer for PDCDl was identified and used for the detection of the nucleotide sequence serving as binding site for a RUNXl transcription factor (US Application Serial No. 10/219,446) (Prokunina et al. 2002). A polymorphism in the enhancer changed the binding site for the transcription factor. The variant disrupting the binding site of the transcription factor was associated with systemic lupus erythematosus and serves as an indication for susceptibility and risk to develop the disease. The polymorphism is described in a patent application (US Application Serial No. 10/219,446). As the binding site for the transcription factors RUNXl, RUNX2 and RUNX3 is shared between the three, any of these can be involved in the regulation of the PDCDl gene, it is required to know which of the RUNX genes regulates PDCDl and also their expression in autoimmune diseases. Not all RUNX proteins can be found in the same cell at the same time and if they do, there is strong competition for the binding site dependent on affinity, dose and quantity of each in the cellular milieu. Also, it is important to know if there are abnormalities in the tissue expression of these genes including also PD-1 or not in patients with SLE and in whaTcells this abnormal expression takes place.
It has now turned out that PD-1, Runxl, Runx2 and Runx3 are differently expressed in Tcells from SLE patients than in healthy individuals. Thus, PD1 is more expressed in CD4+ cells from SLE patients, Runxl more expressed in Tcells and Runx2 and Runx3 more expressed in Tcells and CD 4+ cells than in the corresponding cells from healthy persons (Example 3, tables 1 and 2). It has also turned out that Runxl and Runx2 are expressed in fϊbroblast synovial cells from patients with rheumatoid arthritis (Example 4). Thus, the inventors have detected the cellular subpopulation where the gene expression is to be analyzed and the patterns of expression in various tissues for comparison.
Summary of the Invention
The present invention relates to a method for screening and/or diagnosis of autoimmune diseases, such as rheumatic disorders and SLE based on the detection of levels of expression of PDCDl, RUNXl, RUNX2 and RUNX3. Further, the method detects the cellular subpopulation where the gene expression is to be analyzed and the patterns of expression in various tissues for comparison. The invention also includes a method for the preparation of nucleic acids such as cellular RNA and DNA from specific cellular populations. Further, the invention includes a kit to detect cDNA in cells of patients with SLE or any other autoimmune disease. Further, the invention includes the detection of the cDNA expression of the gene alone or in combinations for the diagnosis of SLE or other related autoimmune conditions. Moreover, the invention includes a method for the follow-up of patients with SLE with various treatments, where levels of PDCDl, RUNXl, RUNX2 and/or RUNX3 can be detected and followed longitudinally. Further, the invention describes a method where the levels of PDCDl, RUNXl, RUNX2 and RUNX3 can be used for screening of therapeutic agents to treat or cure SLE that modify the expression of PDCDl, RUNXl, RUNX2 or RUNX3 or all of these combined or in variable combinations.
The invention also relates to a pharmaceutical composition, comprising at least one compound that modulate PDCDl, RUNXl, RUNX2 or RUNX3, the use of such a compound for the preparation of a pharmaceutical composition for the treatment of autoimmune diseases and a method for the treatment of a patient suffering from an autoimmune disease.
The invention is further elucidated by way of the annexed figures of which:
Figure 1. Expression of PD-1 (PDCDl) and RUNX genes in tissues and blood fractions of normal controls. Prepared cDNAs from the cell subpopulations and tissues were obtained from CLONTECH.
Figure 2 Expression of the PD-1 gene in blood fractions of controls and SLE patients.
Figure 3. Expression of RUNXl genes in blood fractions of controls and SLE patients
Figure 4. Expression of RUNX2 gene in blood fractions of controls and SLE patients
Figure 5. Expression of RUNX3 gene in blood fractions of controls and SLE patients Thus, all three RUNX genes were expressed in human Tcells. RUNX2 and RUNX3 genes showed increased expression in activated Tcells of SLE patients (Table 1 and 2), but no difference was observed in non-Tcells.
Figure 6. Fibroblast synovial cells obtained from patients with rheumatoid arthritis were cultured in RPMI and 10% fetal calf serum with or without TGFbeta under normoxic or hypoxic conditions (normal=cells alone, hypo= with less that 1% 02, TGF=only TGFbeta, hypo/tgf=hypoxia and TGFbeta). RNA was prepared from the cells after harvesting at the points in time shown in the figure. As shown, TGFbeta induces strong expression of RUNXl and RUNX2, while hypoxia tends to reduce the levels, in particular after 4, 6 and 24 hours.
Figure 7. a. Endogenous expression of the RUNX proteins in Jurkat Tcells. Interestingly RUNXl expression decreases when JurkaTcells are stimulated, b. PD-1 expression is conversely increased during Tcell stimulation. Over- expression of RUNXl reduces endogenous PD-1 expression in stimulated cells.
Detailed description of the Invention
Diagnostic Assays
The invention relates to a method for screening and/or diagnosis of autoimmune disorders comprising the step of: a) obtaining biological sample from a test subject b) contacting the biological sample with at least one agent capable of detecting at least one of PDCDl, RUNXl, RUNX2 and/or RUNX3 nucleic acid or the corresponding expression product coded for by one or more of these genes or a sub fragment thereof, c) comparing the expression rate of at least one expression product from PDCD 1 , RUNXl , RUNX2 and/or RUNX3 nucleic acid or a subfragment thereof from a healthy test subject with the expression rate thereof from another test subject.
The presence or absence, increase or decrease of RUNXl, RUNX2 and/or RUNX3 and/or PDCDl or the expression products thereof may be analysed in the biological sample. One or a combination of several of the above mentioned genes or their expression products may be analysed.
The biological sample may be chosen from peripheral blood mononuclear cells such as lymphocytes, especially T-lymphocytes or CD4+ cells. Preferably the lymphocytes are activated. This may be done e.g. with anti-CD3 antibodies, PMA (phorbol-12- myristate- 13 -acetate) and Ionomycin (I), PWM (pokeweed mitogen) or a combination thereof.
The nucleic acid which is detected may be RNA, such as cellular RNA, mRNA, or
DNA such as cDNA or genomic DNA. The at least one agent may be a nucleic acid probe capable of hybridizing to any of
PDCD, RUNXl, RUNX2 or RUNX3 mRNA, cDNA or genomic DNA.
Suitable probes for use in the diagnostic assays of the invention are described herein.
In one embodiment, the biological sample contains mRNA molecules from the test subject or genomic DNA. In another embodiment, the methods further involve obtaining control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting mRNA, cDNA or genomic
DNA in the biological sample, and comparing the presence or absence or increased or decreased levels of mRNA in the test sample.
Usable probes may be a nucleic acid that comprises at least 20, at least 30, preferably at least 40 and most preferred at lest 50 nucleotides capable of hybridizing to any sequence chosen from sequences that differ between RUNXl, RUNX2 or RUNX3, or chosen from the PDCDl sequence.
RUNXl differentiates from the sequence of RUNX2 and RUNX3 at sequence SEQ
No.l covering 378 bp of contiguous nucleotide sequence, covering nucleotides 1141- 1469 bp from GENBANK sequence NM_001754.
In a preferred embodiment, the sequence of 135 bp is used covering nucleotides 1186-
1320 (SEQ No. 2), corresponding to amino acids - 248-292, in the junction of exons 6 and 7b. This sequence can be detected using SEQ No. 3 primer RUNX1F and SEQ
No.4 primer RUNXlR. In a still preferred embodiment, detection is improved using a probe described in SEQ.
No. 5.
The minimal sequence that can be detected, but which is not preferred comprises the minimal sequence of 1231-1281 bp, a nucleotide sequence of 50bp (SEQ No. 6) or also the nucleotide sequence comprising as minimal sequence 1391-1441 bp, as a contiguous sequence of 48 bp (SEQ No. 7). The invention also describes a method for detection of variation in expression of the cDNA of the RUNX2 gene. RUNX2 differentiates from the sequence of RUNXl and RUNX3 at SEQ No. 8, covering base pairs 721-1140 bp (based on Genebank sequence NM_004348). In a preferred embodiment, the sequence of bp 757-858, corresponding to amino acids 253-286, a total of 102 contiguous nucleotides is used (SEQ No. 9). This sequence is recognized and hybridizes the nucleotides from the kit known as primer RUNX2F (SEQ No. 10) and SEQ No. 11, known as Primer RUNX2R. In a preferred embodiment, detection is improved using a probe described in SEQ. No. 12. In a further embodiment, the minimal sequence that can be detected, but which is not preferred comprises the minimal sequence of 791-841 bp (SEQ 13) comprised of 50 contiguous nucleotides or the sequence bp 951-1061 (junction over two exons) comprised by a contiguous sequence of 110 nucleotides (SEQ No. 14). The invention also describes a method for detection of variation in expression of the cDNA of the RUNX3 gene specifically at a region covering a 340 bp sequence, SEQ no. 15 unique for RUNX3 gene from bp 581-921, a contiguous nucleotide sequence of 340bp (based on Genebank sequence: NM_004350).
In a preferred embodiment, the method detects and produces an amplicon of 125 contiguous nucleotides at the borders of exons 4 and 5, corresponding to bp 220-260, corresponding to amino acids 220-260 of the RUNX 3 gene (SEQ No.16).
This sequence is recognized and hybridizes the nucleotides from the kit known as SEQ
No. 17 primer RUNX3F and also with sequence No. 18: primer RUNX3 R.
In a preferred embodiment, detection is improved using a probe described in SEQ. No.
19. In a further embodiment, the minimal sequence that can be detected, but which is not preferred comprises the minimal sequence of 681-741 bp or a contiguous nucleotide sequence of 60 bp (SEQ No. 20).
The invention also describes a method for detection of variation in expression of the cDNA of the PDCDl gene specifically at a region that hybridizes any of the consecutive nucleotide sequences, SEQ no. 21, of 50 bp, SEQ NO. 22, of 50 bp, SEQ NO: 23, of 50 bp, SEQ NO. 24 of 50 bpbased and SEQ NO. 25 of 65 consecutive nucleotides (numbering is based on the Genbank sequence NM_005018) being these also the minimal sequences that can be detected. This sequence is recognized and hybridizes the nucleotides from the kit known as SEQ NO. 26 PDlforw and SEQ NO: 27 PDlrev. In a preferred embodiment, detection is improved using a probe described in SEQ. No. 28.
In a first embodiment, detection can be done using the polymerase chain reaction and detection using gel electrophoresis and staining with ethidium bromide. In a preferred embodiment, detection can be performed using real-time quantitative PCR. Detection can be performed by labelling of the probes with SYBRgreen and the use of a laser based detector or with radioactive isotopes and the use of autoradigraphic films or a phosphoimager (Hernandez et al. 2003; Maeda et al. 2003; Von Samson- Himmelstjerna et al. 2003; Luthra et al. 2004). In a further embodiment, the oligonucleotides are used for hybridization in microarrays. In a first embodiment, the probe can be detected through hybridization and autoradiography. In a preferred embodiment, the detection can be done using fluorescent labels such as FAM and TAMRA, or other amidites such as ROX, or other labels such as VIC, JOE, etc or other labels that can use fluorescent detection. Preferred nucleic acid probes are, for detection of RUNXl those set forth in SEQ ID NO. 3, 4, 5; for detection of RUNX2 10, 11, 12; for detection of RUNX3, 17, 18, 19; and for PDCDl, SEQ NO. 26, 27 and 28 or a portion thereof. Preferably the probe is labelled.
The autoimmune disorder that may be screened and /or diagnosed may be chosen from: a) multiple sclerosis, b) myasthenia gravis, c) Type 1 diabetes, d) rheumatoid arthritis, e) Sjδgrens syndrome f) atopy, g) allergy, or
10 h) systemic lupus erythematosus, where systemic lupus erythematosus (SLE) is characterised by conditions selected from the group including any one or more; fatigue, fever, loss of appetite, nausea, weight loss, hives, loss of scalp hair, red "butterfly rash" and raised rash, sensitivity to sun, ulcers in mouth, nose, or vagina, arthritis, joint pain, loss of blood supply to bone, pain, infections within joints, decrease in kidney function including, blood, aberrant amounts of protein or white blood cells in urine, intracerebral haemorrhage, headaches, loss of coordination, memory loss, seizures, strokes, anaemia, low white blood cell or low platelet count, pericardial effusion, heart attack, inflammation in the heart, infection in the heart, inflammation of the lining of the heart, infection of the lining of the heart, heart valve problems, shortness of breath, cough, inflammation of the lungs, inflammation of the lining of the lungs, abdominal distress, diarrhoea, enlargement of the liver, loss of appetite, nausea and vomiting, blindness, visual impairment, dryness of the eyes and dryness of the mouth.
The invention especially relates to the screening or diagnosing of SLE and rheumatoid arthritis.
Thus, SLE may be diagnosed when the rate of expression of PDCDl in CD4+ cells is higher in the tested subject that in a healthy test subject; when the rate of expression of RUNXl in T - cells is higher in the tested subject that in a healthy test subject ; when the rate of expression of RUNXl and/or RUNX3 in T - cells and/or CD4+cells is higher in the tested subject that in a healthy test subject.
Rheumatoid arthritis may be diagnosed when the rate of expression of RUNXl and 2 in synovial fibroblasTcells is modulated in the tested subject compared to a healthy test subject when subjected to hypoxia or to TGFbeta.
According to the invention SLE may be diagnosed when PD-1 is at least 2 fold higher, especially at least 3 fold higher, preferably at least 4 fold higher and most preferred at least 4,5 fold higher in lymphocytes of SLE patients compared to controls. Preferably lymphocytes, such as Tcells, preferably CD4+cells are used. The cells are preferably activated. According to another embodiment SLE may be diagnosed when RUNX2 is at least 2 fold higher, especially at least 3 fold higher, preferably at least 4 fold higher in lymphocytes of SLE patients compared to controls. Preferably lymphocytes, such as Tcells, preferably CD4+cells are used. The cells are preferably activated.
According to still another embodiment SLE may be diagnosed when RUNX3 is at least 2 fold higher, especially at least 3 fold higher in lymphocytes of SLE patients compared to controls. Preferably lymphocytes, such as Tcells, preferably CD4+cells are used. The cells are preferably activated.
According to the invention rheumatoid arthritis may be diagnosed when RUNXl and/or RUNX2 are at least 2 fold higher, especially at least 3 fold higher, preferably at least 4 fold higher in fibroblast synovial cells from patients with rheumatoid arthritis compared to controls.
Kits
The invention also encompasses kits for detecting the presence of PDCDl, RUNXl, RUNX2 and/or RUNX3 in a biological sample. For example, the kit can comprise a labelled or unlabeled compound or agent capable of detecting PDCDl, RUNXl,
RUNX2 and/or RUNX3 mRNA cDNA or genomic DNA in a biological sample; means for determining the amount in the sample, means of comparing the amounts in the sample with a standard and instructions for the use of the kit. Examples of how this may be done are to be found in the examples below. The compound or agent can be packaged in a suitable container. A kit comprising at least one, nucleic acid probe capable of hybridizing to mRNA, cDNA or genomic DNA from RUNXl, RUNX2,
RUNX3, PDCDl.
The kit may comprise at least 20, preferably at least 30, such as at least 40, most preferred at least 50 nucleotides capable of hybridizing to any of SEQ ID NO 1, 2, 6,
7, 8, 9, 13, 14, 15, 16, 20, 21, 22, 23, 24, 25.
12 In a preferred embodiment the kit comprises at least one nucleic nucleic acid probe chosen from SEQ ID NO. 3, 4, 5; 10, 11, 12; 17, 18, 19; 26, 27 and 28 or a portion thereof.
Preferably the at least one probe is labelled, preferably as described above. The kit can further comprise instructions for using the kit to detect PDCD 1 , RUNXl , RUNX2 and/or RUNX3 nucleic acid, preferably mRNA and instruction for the statistical comparison with a set standard as well as preparation of the standard.
Screening Assays The invention provides a method (also referred herein as screening assay) for identifying modulators, i.e. candidate or test compounds or agents that modify PDCDl, RUNXl, RUNX2 and/or RUNX3 expression, have an induction or reduction effect on their expression. Such a screening assay comprises the step of: a) obtaining biological sample from a test subject b) contacting the biological sample with at least one agent capable of detecting the PDCDl, RUNXl, RUNX2 and/or RUNX3 nucleic acid or the corresponding expression product coded for by these gene or a subfragment thereof, c) optionally adding a compound to be tested for modulation d) determining the rate of expression of at least one of the PDCDl, RUNXl, RUNX2 and/or RUNX3 expression products e) determining if the compound has induction or reduction effect on their expression. In one embodiment, the invention provides assays for screening candidate or test compounds which alter or modulate the expression of the RUNX genes. In a preferred embodiment, the compounds normalize the expression of the genes, making them candidates for treatment of systemic lupus erythematosus. The test compounds of the present invention can be obtained using any of numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries, synthetic library methods using affinity chromatography selection, and other methods to peptide libraries, non-peptide oligomer or small molecule libraries of compounds (DeWitt et al. 1993; Lam et al. 1997).
Monitoring the ability of compounds to modulate the RUNX genes can be accomplished by monitoring in a cell-based assay the effect of the compound on the expression of the genes, by adding the compound in the cellular culture. In a further embodiment, the compound can be tested in transfection assays, where any compound is added and expression of the RUNX genes is tested. In a further embodiment, compounds that modulate Tcell activation can be identified by determining the ability of the compound to modulate expression of PDCDl, RUNXl, RUNX2 and RUNX3. Furthermore, compounds that modulate myeloid activation can also be tested for their ability to modulate expression of PDCDl, RUNXl, RUNX2 and RUNX3.
Prognostic Assays The diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk of developing systemic lupus erythematosus or any disorder associated with aberrant or unwanted PDCDl, RUNXl, RUNX2, or RUNX3 activity, alone or in combination, known to be an autoimmune disease. As used herein "aberrant" includes a PDCDl, RUNXl, RUNX2 and/or RUNX3 expression or activity which deviates from the wild-type PDCDl, RUNXl, RUNX2 and/or RUNX3 expression. Aberrant expression includes increased, decreased expression or activity alone for each of the genes or in combination.
Furthermore, the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g. agonist, antagonist, peptidomimetic, peptide, nucleic acid, small molecule or other drug or candidate) to treat an autoimmune disease or disorder with aberrant expression of PDCDl, RUNXl, RUNX2 and/or RUNX3. For example, such methods can be used to determine whether a subject can be effectively treated for an autoimmune disorder, including systemic lupus erythematosus, rheumatoid arthritis, or other disorder known in the art to be of autoimmune nature.
14 The methods of the invention can also be used to detect genetic alterations in the RUNXl, RUNX2 and/or RUNX3 gene, thereby determining if a subject with the altered gene is at risk for a disorder characterized by missregulation of RUNXl, RUNX2 and or RUNX3 in particular if this is an autoimmune disorder. In preferred embodiments, the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic alteration characterized by mis-expression of PDCDl, RUNXl, RUNX2 and/or RUNX3, in particular in autoimmune disorders.
Monitoring of Effects During Clinical Trials Monitoring the influence of agents (e.g. drugs) on the expression or activity of PDCDl, RUNXl, RUNX2 and/or RUNX3 can be applied not only to basic drug screening, but also in clinical trials. For example the effectiveness of an agent determined by a screening assay as described herein to increase or decrease PDCDl, RUNXl, RUNX2 and/or RUNX3 expression, can be monitored in clinical trials for subjects with systemic lupus erythematosus or other autoimmune disease exhibiting increased, decreased or aberrant levels or pattern of expression of the RUNXl, RUNX2 and/or RUNX3 genes alone or in combination with or without the expression of PDCDl. For example, and not by way of limitation, genes, including PDCDl, RUNXl, RUNX2 and RUNX3, that are modulated in cells by treatment with an agent (e.g. compound, drug or small molecule) which modulate PDCDl, RUNXl, RUNX2 and/or RUNX3 activity can be identified. Thus, to study the effect of agent on PDCDl, RUNXl, RUNX2 and/or RUNX3 -associated disorders, for example in a clinical trial, cells can be isolated and RNA prepared and analyzed for the levels of expression of PDCD 1 , RUNXl , RUNX2 and/or RUNX3 alone or in combination. The levels of gene expression (e.g. a gene expression pattern) can be quantified by Northern blot analysis or RT-PCR as described herein, by one of the methods described herein. In this way, the gene expression can serve as a marker, indicative of the physiological response of the cells to the agent. Accordingly, this response state may be determined before, and at various points during treatment of the individual with the agent.
15 In a preferred embodiment, the present invention provides a method for monitoring the effectiveness of treatment of a subject with an agent including a pre-administration sample from a subject prior to administration of the agent; detecting the expression of PDCDl, RUNXl, RUNX2 and/or RUNX3 in the preadministration sample; obtaining one or more postadministration samples and; measuring expression of the genes in the postadministration samples; comparing the levels of expression in the preadministration sample or postadministration sample; and altering the administration of the agent to the subject accordingly. According to such an embodiment, PDCDl, RUNXl, RUNX2 and/or RUNX3 expression alone or in combination may be used as an indicator of the effectiveness of the agent, even in the absence of an observable phenotypic response.
Pharmaceutical compositions and treatment.
The invention also relates to a pharmaceutical composition comprising a compound that modulates PDCD 1 , RUNXl , RUNX2 or RUNX3 expression identified by the step of: a) obtaining biological sample from a test subject b) contacting the biological sample with at least one agent capable of detecting the PDCDl, RUNXl, RUNX2 and/or RUNX3 nucleic acid or the corresponding expression product coded for by these gene or a subfragment thereof, c) optionally adding a compound to be tested for modulation d) determining the rate of expression of at least one of the PDCD 1 , RUNXl, RUNX2 and/or RUNX3 expression products e) determining if the compound has induction or reduction effect on their expression.
According to one embodiment of the invention the compound is an agonist, antagonist to the expression products of PDCDl, RUNXl, RUNX2 or RUNX3, such as peptidomimetics, peptides, nucleic acids such as anti sense DNA, antisense RNA, such
SUBSTITUTE feT (RULE 2 as siRNA or ribozyme, small molecules or drugs, antibodies, including mouse or humanized antibodies.
The invention includes the construction of a pharmaceutical compound used to over express the cDNA and the protein for PDCDl, RUNXl, RUNX2 or RUNX3 in cases where the expression of these genes is deficient. They may be pair of nucleotide primers that over express the cDNA of the genes. The compounds may be used as gene therapy. Especially the invention relates to compounds, such as nucleotides that over express RUNXl and RUNX3 such as the primers SEQ ID NO 29, 30 and 31, 32.
The oligonucleotides from SEQ. 29 and 30 are used to amplify the full-length cDNA that produces the peptide known as the AML-lc isoform of RUNXl shown in SEQ. NO. 31 of 1464bp including a 4bp sequence CACCT at the beginning used to clone in the vector.
The oligonucleotides from SEQ. 32 and 33 are used to amplify the full-length cDNA that produces the peptide for RUNX3 shown in SEQ. NO. 34 of 1258bp including a 4bp sequence CACCT at the beginning used to clone in the vector.
The nucleotides may be used with a vector that is introduced onto cells. As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of preferred vector is an episome, i.e., a nucleic acid capable of extra-chromosomal replication. Preferred vectors are those capable of autonomous replication and/or expression of nucleic acids to which they are linked. Vectors capable of directing the expression of genes to which they are operatively linked are referred to herein as "expression vectors". In general, expression vectors of utility in recombinant DNA techniques are often in the form of "plasmids" which refer generally to circular double stranded DNA loops which, in their vector form are not bound to the chromosome. In the present specification, "plasmid" and "vector" are used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors which serve equivalent functions and which become known in the art subsequently hereto.
The invention is intended to include all forms of expression vectors including replication defective retroviruses, adenoviruses and adeno-associated viruses and lentiviruses. One type of vector used is a plasmid, which refers to a circle double stranded DNA loop into which additional DNA segments can be ligated. Certain vectors are capable of directing the expression of genes.
The invention further includes pharmaceutical compositions, which comprise an expression vector comprising at least one DNA molecule of PDCDl, RUNXl, RUNX2 or RUNX3 cloned into the expression vector in an antisense orientation. The DNA molecule may be operatively linked to a regulatory sequence in a manner, which allows for expression of an RNA molecule, which is antisense to PDCDl, RUNXl, RUNX2 or RUNX3 mRNA. The system includes the use of RNA interference technology where persistent expression of a silencer RNA molecule known as siRNA, is allowed in order to silence the gene desired. This siRNA is an antisense of PDCDl, RUNXl, RUNX2 or RUNX3 mRNA or a subfragment of PDCDl, RUNXl, RUNX2 or RUNX.
The invention especially relates to the nucelotides SEQ ID 35, 36 and 37 that target the positions of the RUNXl gene at 505-525 (SEQ. NO. 35), 959-978 (SEQ. NO. 36) and 1242-1263 (SEQ. NO. 37).
A lentiviral vector (LW) may be used. The ideal vector for gene therapy should be first of all safe, non-immunogenic, non-toxic, non-allergic, enabling ease, efficient and stable gene transfer to targeTcells, which are often non-replicating and terminally differentiated. Such vector should have a satisfactory cloning capacity. Among different viral and non- viral vectors LW (LentiViral-based Vector) of the third generation with increased bio-safety and capacity have some essential advantages (Dull et all 998, Zufferey et al.1998). Lentiviruses are a class of retroviruses that can
18 infect both dividing and non-dividing cells. This property makes them especially valuable gene delivery vehicles in terms of gene therapy. Although LW share some concerns with oncoretroviral vectors (e.g. possibility of insertional mutagenesis) it also offers many benefits. Self-inactivating lentiviruses with all deleted viral genes can accommodate about 8.5kb of DNA fragment. This is highly desirable, as some therapeutic genes together with regulatory elements may be long. Viral particles are packed in a special cell line with help of three independent non-overlapping split- genome packaging constructs, two of them expressing HIV proteins necessary for vector propagation and a third expressing heterologous envelope (vesicular stomatitis virus glycoprotein, VSVG) protein. The major feature of self-inactivating viral vectors is a deletion in the U3 region of the 3'- LTR, making them incapable to produce packageble viral genomes after integration into the host genomic DNA increasing biosafety.
Since lupus is characterized by reiterated periods of flares and remissions, it is important to have systems allowing regulation of expression of a therapeutic gene at the proper time. Up-regulation of a gene caused by a single drug administration will ameliorate disease and improve the health of the patient. First of all such regulated system should not influence the transcription of endogenous genes and provide low- level (ideally undetectable) basal activity and rapid induction of a drug dose-response. The system must consist of artificial promoters and regulator proteins to prevent interference with cellular processes.
The important part of the regulated system is the inducer that should not be toxic, do not cause adverse effects (i.e. be fully physiologically inert compound) and be preferably orally bio-viable to ease treatment. Unfortunately none of the existing regulated systems can fully fit to all of these demands. They all have advantages as well as disadvantages and probably might be used only for animal model study, at present. Rapamycin in the dimerization-based regulation system interferes with cellular signaling through inhibition of evolutionary conserved signal transduction pathways and cause immunosuppressive effects (Fruman et el. 1994). Ecdysone is quite complicated and overexpression of one of the regulator protein RXRa may exert pleiotropic effects because of its human origin (No et el. 1996, Bugge et al. 1992). The most developed Tet-system has serious drawbacks such as high basal level activity of the therapeutic gene (tet-on system, Freundlieb et al. 1999, Urlinger et al. 2000, Lamartina et al. 2002). The tet-off system is not suitable for long-term applications since un-induced state is maintained by continuous treatment with Tet or Dox. The expression of the gene is activated only after withdrawal of drug. Best suited for testing different gene therapy approaches is the last of the modifications of the RU486-dependent regulated system, called GeneSwitch (Abruzzese et al. 2000). The system includes a chimeric regulator protein and an autoinducible promoter consisting of four copies of Gal4 binding sites linked to a minimal thymidine kinase promoter. The promoter/enhancer for the therapeutic gene is composed of six copies of Gal4 sites followed by the TATA sequence from the Adenoviral Elb promoter. Thus the regulatory GeneSwitch protein contains the following functional domains: a) Mutated human progesterone receptor ligand binding domain (LPR-LBDDdelta) capable of changing its conformation upon binding to the progesterone antagonist, mifepristone (RU486); b) Yeast Gal4 DNA binding domain (Gal4-DBD); c) Transcription activation domain from the human NF-kB transcription factor p65 (p65AD). Expression of the regulator as well as the transgene is ligand-dependent and inducible. In normal, un-induced states the system provides weak expression of the inactive regulator. After a single oral or i.p. administration of mifepristone (RU486) the regulator is activated and provides an increase of transcription of its own gene and of the therapeutic gene. The concentration of mifepristone needed for maximal induction of gene expression (250-350 mg/kg) is far below the levels at which mifepristone has antiprogesterone and antiglucocorticoid activity (Abruzzese et al. 200). The advantage is that the recombinant regulatory protein is synthesized at high levels only when mifepristone is administered. The maximum level of expression varies in different models and for different transgenes ranging from 25% to 80% of the CMV-driven expression but it is high enough to achieve therapeutic effect. The GeneSwitch system is preferred.
20 Further, the pharmaceutical compounds described herein can be used to screen cells from various individuals or cell lines and study how the overexpression or silencing of the RUNX genes modulate the expression of other molecules, genes or peptides. This is also done using stable or temporary transfection for which an example is provided.
The invention also relates to the use of such a compound for the preparation of a pharmaceutical composition for the treatment of the above mentioned diseases or disorders.
The compound used in the composition is present in an effective amount. The phrase "an effective amount", as used herein, means a therapeutically effective amount of a compound or composition large enough to modify the symptoms and/or condition to be treated, but small enough to avoid serious side effects (at a reasonable benefit/risk ratio), within the scope of sound medical judgment. The effective amount of active ingredient for use in the pharmaceutical compositions and the methods of the invention herein will vary depending upon the severity of the diseases, the duration of the treatment, the nature of concurrent therapy, the particular active ingredient being employed, the particular pharmaceutically-acceptable excipients utilized, and like factors within the knowledge and expertise of the attending physician.
For pharmaceutical compounds, the dosage may range from about 0.0001 to lg/kg, such as 0,01 mg to 500 mg/kg and more usually 0.1 to 5 lOOmg/kg, e.g. 0,5-50 mg of the host body weight.
Doses for nucleic acids may range from about 10 ng to lg, 100 ng to 100 mg, 1. μg to
10 mg, or 30-300 μg DNA per patient.
The term "pharmaceutically-acceptable excipients", as used herein, includes any physiologically inert, pharmacologically inactive material known to one skilled in the art, which is compatible with the physical and chemical characteristics of the particular azapeptide compound active ingredient selected for use. Pharmaceutically-acceptable
21 excipients include, but are not limited to, polymers, resins, plasticizers, fillers, binders, lubricants, glidants, disintegrates, solvents, co-solvents, buffer systems, surfactants, preservatives, sweetening agents, flavouring agents, pharmaceutical grade dyes or pigments, and viscosity agents.
Methods of treatment.
Further, the invention relates to a method for the treatment of a patient suffering from an autoimmune disease wherein a compound that is an agonist, antagonist to the expression products of PDCDl, RUNXl, RUNX2 or RUNX3, such as peptidomimetic, peptide, nucleic acid such as anti sense DNA or RNA is administrated.
While the invention has been described in relation to certain disclosed embodiments, the skilled person may foresee other embodiments, variations, or combinations which are not specifically mentioned but are nonetheless within the scope of the appended claims.
All references cited herein are hereby incorporated by reference in their entirety.
The expression "comprising" as used herein should be understood to include, but not be limited to, the stated items.
The invention will now be described by way of the following non-limiting examples.
EXAMPLES
The present invention is used to detect variable levels of PDCD 1 , RUNXl , RUNX2 and/or RUNX3 in normal individuals and in patients with SLE, rheumatoid arthritis or other diseases of the immune system including autoimmune diseases. The kit described here allows for the detection of levels of each gene individually or in combination forming a pattern characteristic of patients with SLE as compared to normal individuals.
22 Peripheral blood mononuclear cells (PBMC) are purified from heparin-treated blood samples by Fycoll-hypaque gradient centrifugation (Bouyant density gradients). For further separation of cell types, T and non-Tcells are separated using sheep red blood cells and 95% purity of separated Tcells is obtained, as determined with anti-CD3 antibodies, a specific peripheral Tcell marker.
In a preferred embodiment, total PBMC are fractionated using cell sorting (FACS) where PBMC are incubated with antibodies to CD14, CD19, CD4 and CD8, conjugated with fluorescent dyes (antiCD14-PE, antiCD19-FITC, antiCD4-APC and antiCD8-PC5) for 20 min on ice, washed and subjected to FACS separation. The purity of each population is confirmed by FACS analysis and is not less than 95%. In a first embodiment, several CD4+ and CD8+ samples are activated using anti-CD3 antibodies for 3 days. In a preferred embodiment, cells are activated by a combination of PMA (phorbol-12-myristate- 13 -acetate, 20ng/ml) and Ionomycin (I) (0.5mM) (Sigma) and incubated for 2, 4 and 6 hours. In a further embodiment, non-Tcells or CD 19+ cells are activated by PMA/I for 4 hours or PWM (pokeweed mitogen) for 3 days.
Total RNA is extracted using Trizol reagent (Life Technologies) and cDNAs is prepared with TaqMan Reverse Transriptase Kit (PE Biosystem). The amount of cDNA generated is measured using the TaqMan assays with a 7700 ABI PRISM sequence detector system (PE Biosystem). TaqMan primers and probes are designed with Primer Express Software (PE Biosystem) so that they covered exon junctions to assure for the sole amplification of cDNA and none of genomic DNA. All samples are run at least twice and mean values are used for calculations. Standard curves are constructed for all assays from serial dilutions of cDNA from the JurkaTcell line. Standard curves are used to calculate the level of expression of all the genes according to the manufacturers instructions. Absolute values are calculated as the proportion of the amount of transcript of the query genes normalized to the amount of transcript of endogenous control. Two endogenous controls: glyceraldehydephosphate dehydrogenase (GAPDH) and B2-microglobulin (B2M) are run for each sample in duplicates and Ct values are calculated. To choose endogenous control, cDNAs were prepared from 2 ug of total
23 RNA extracted from non-treated non-Tcells and activated Tcells (30 control samples in total) and cDNAs were diluted so that 100 ng of total RNA is used for each RT- PCR reaction. The mean Ct value for GAPDH is 23.7 +/- 2.17 with coefficient of variation of 9.2%. For B2M the mean Ct value is 21.0+/- 0.82 and coefficient of variation of 3.9%. Based on these calculations B2M is a more stable endogenous control for the study and used to calculate the results from the various experiments. In a further embodiment, the invention provides a method to detect mRNA from PDCDl, RUNXl, RUNX2 and RUNX3 alone or in combination in cells cultured in vitro and stimulated with various substances.
Example 1 PD-1 expression
To evaluate the optimal conditions for PD-1 gene expression, PBMC of 5 controls were treated by PMA/I for 2, 4 and 6 hours and compared with non-treated samples. Expression of PD-1 gene is increased by 2.7 folds at 2 hours (P-0.04), by 3.7 folds at 4 hours (P=0.002) and is not different from untreated samples at 6 hours. Therefore, activation by PMA/I for 4 hours is the standard.
PD-1 expression is not detected in resting B-cells (cell line Daudi, peripheral blood B cells, bone marrow, tonsils) or myeloid cells (cell lines THP-1, U937, K562, KM3 and peripheral CD 14+ cells). Activation of B cells by PMA/I for 4 hours or by PWM for 4 days and activation of monocytes by PMA/I for 4 hours does not induce PD-1 expression. PD-1 expression is observed in the Molt 4 cell line (representing CD4+CD8+ thymic cells) and in cDNA from the thymus, spleen and lymph nodes. The low expression is increased by 3-4 fold by PMA/I activation in peripheral blood CD4+ cells and in the Jurkat CD4+ cell line. Expression is not detected in either resting or activated peripheral blood CD8+ cells and the HSB2 cell line (CD8+). These results indicate that PD-1 is expressed in differentiating and mature CD4+ Tcells (Figure l,a).
Example 2. Gene expression of RUNXL RUNX2 and RUNX3 Highest expression of RUNXl is detected in myeloid cells including peripheral CD 14+ cells and the cell lines THP, KM3 and K562. Expression of RUNXl is also
24 observed in both CD4+ and CD8+ Tcells: Molt 4, Jurkat and HSB2 cells and in peripheral CD4+ and CD8+ cells. Low expression is observed in B cells (Figurel,b) The RUNX2 gene was mostly expressed in myeloid cells and in peripheral CD4+ and CD8+ Tcells and Tcell lines (Molt4, Jurkat and HSB2), but was not observed in B cells (Figure l,c)
Finally, highest expression of the RUNX3 gene was observed in CD8+ cells, both in the HSB2 cell line and in peripheral CD8 + cells. RUNX3 was also detected in resting and activated CD4+ cells and B cells, but was not present in myeloid cells (Figure l,d).
Example 3. Expression of the PD-1 and the RUNX genes in SLE PD-1 was expressed higher in activated PBMC of patients compared to controls (P=0.0006) (Figure 2). We decided to clarify further which blood fractions contribute to this difference. There was no difference of the PD-1 expression in non-Tcells (resting and activated B cells and monocytes) of patients compared to controls, while expression was 4.5 fold higher in activated Tcells of patients (P=0.00004) as compared with activated Tcells from controls. Interestingly, the difference in expression was due to increased PD-1 expression in activated CD4+ cells of patients, while expression in resting and activated CD8+ cells of both patients and controls was not detectable. Thus patients with SLE have an increased expression of PD-1 in activated CD4+ cells (Figure 2).
Expression of all RUNX genes in non-Tcells (resting and activated B cells and monocytes) did not differ between patients and controls. In Tcells activated with PMA/I for 4 hours, expression of RUNXl did not differ in patients compared to controls while it was almost 4 fold higher (p=0.003) for RUNX2 and 3 fold higher (P=0.0001) for RUNX3 genes (Table 1 and Figures 3-5). Table 1. Pattern of gene expression in blood fractions of controls
Cells PD-1 RUNXl RUNX2 RUNX3
Total PBMC 0.0004 0.0002 0.009 0.019
Total PBMC activ 0.0014 0.0003 0.027 0.026
Total T activ 0.0001 0.0012 0.005 0.009
CD4+ 0.0003 0.0005 0.074 0.018
CD4+ activ 0.0008 0.0003 0.02 0.006
CD8+ 0.0001 0.0007 0.07 0.017
CD8+ activ 0.0009 0.0003 0.05 0.016
CD 19+ 0.00001 0.0002 0.0004 0.019
CD 19+ activ 0.00001 0.0001 0.005 0.003
CD 14+ 0.00001 0.001 0.012 0.003
Table 2. Gene expression, in systemic lupus erythematosus (SLE) compared to normal pattern. Significant differences from controls are shown in bold; a tendency is shown in italic (Results are expressed as ratio of each gene/GAPDH or β2M)
Cells PD-1 RUNXl RUNX2 RUNX3
Total PBMC 0.0008
Total PBMC activ 0.007
Total T activ 0.0034 0.002 0.019 0.025
CD4+ 0.001 0.0006 0.036 0.009
CD4+ activ 0.003 0.0006 0.07 0.012
CD8+ 0.0004 0.0006 0.03 0.014
CD8+ activ 0.002 0.0006 0.05 0.014
Non-Tcells 0.0001 0.0008 0.016 0.004
Figure imgf000028_0001
In a further embodiment, the levels of PDCDl, RUNXl, RUNX2 and/or RUNX3 can be compared to those of normal controls using statistical methods such as non-paired Student' s T test and determination of a p value.
Example 4. Expression of RUNX genes in fibroblast synovial cells from patients with rheumatoid arthritis
In a second example, synovial fibroblast-like cells were obtained through arthroscopy from the knee joint of patients with rheumatoid arthritis. Using methods known in the art, the cells were grown in culture in standard conditions or were subjected to hypoxa or to tumor growth factor beta (TGFbeta). After 2, 4, 6 or 24 hours the cells were harvested and RNA was prepared using described methodology. Using the kit described herein for RUNXl, RUNX2 and RUNX3, the expression of these genes was determined. RUNX3 and PDCDl were not detected, while changes were observed with RUNXl and RUNX2. Overall the levels of RUNX2 were higher than for RUNXl. This is shown in figure 6 where hypoxia reduced the overall transcription of RUNXl and RUNX2 while TGFbeta enhanced their production substantially. As hypoxia is suggested to be possibly involved in the pathogenesis of rheumatoid arthritis in the joint, this effect could be mediated by RUNXl and RUNX2. The results are shown in figure 6. (Jackson, Minton et al. 1997; Berse, Hunt et al. 1999; Demasi, Cleland et al. 2004)
Example 5. Overexpression of RUNXl and RUN3 genes
The full-length cDNA sequence of RUNXl is amplified using the oligonucleotides described in SEQ. 29 and 30 (that recognize nucleotides 425-447 and 1868-1887 in the sequence NM_001754 and amplify the portion known as AML-lc described in SEQ. 33). That for RUNX3 is obtained by PCR using the oligonucleotides described in SEQ. No. 31 and 32 (and recognize positions 1-24 and 1237-1258 in the sequence NM_004350 and amplify the portion described in SEQ. 34). Both and cloned into ρcDNA3.1D/V5-His-TOPO vector (Invitrogen, K 4900-01). JurkaTcells were transfected with Lipofectamine™ 2000 (Invitrogen) in 24-well plates according to the manufacturer's protocol. Briefly, 0.35xl06 cells were seeded in each well in antibiotic-free RPMI medium supplemented with 10% FBS. 0.4 μg of the pcDNA vector carrying either of the Runx genes or empty vector for negative control, were incubated in 50 μl of Opti-MEM medium (GIBCO) for 5 min. Then diluted DNA was combined with diluted in 50 μl of Opti-MEM Lipofectamine 2000, incubated at room temperature for 20 min and added to the cells. Cells were incubated at 37°C in a C02 incubator for 48 hours. Stimulation was done for 10 hours with 20 ng/ml of PMA and 0.5μM of ionomycin. Transfection of THP-1 and Daudi cells was carried out by DEAE/Dextran method. 5xl06 cells/sample were washed once in PBS and once in RPMI medium and resuspended in 750 μL of RPMI medium. 2.5 μg of plasmid DNA were mixed with 50 μg DEAE/Dextran in 250 μl RPMI medium and incubated at RT for 10 min before it was added to the cells. Cells are incubated at 37°C in a C02 incubator for 30 min and then washed carefully with excessive amount of RPMI medium and resuspended in 3 mL of RPMI supplemented with 10% FBS. Cells were incubated at 37°C in a C02 incubator for 48 h. Daudi cells are stimulated with PMA and ionomycin, as described for Jurkat, and THP cells are stimulated for 10 hours with 100 ng/ml of LPS and 10 ng/ml of interferon γ.
Example 6. Silencing of RUNXl gene
For silencing of the Runxl gene several constructs targeting to the positions 505-525 959-978 and 1242-1263 in the sequence NM_001754 were prepared as follows. Single cassettes encoding sense, 9-base loop (TTCAAGAGA), antisense and terminator (TTTTTT) were prepared according to the protocol from OligoEngine and cloned into pSUPERIOR.puro vector (OligoEngine). The constructs were transfected as described above.
The sequences used are those described in SEQ. No. 35, 36 and 37. Human monocytic leukemia cells (THP-1), human Tcell leukemia cells (Jurkat) and Burkitt's lymphoma cells (Daudi) were grown in 37°C in 5% C02 with RPMI 1640 media (GIBCO), supplemented with 10% fetal bovine serum (FBS) (GIBCO), penicillin (100 U/mL) and streptomycin (100 μg/mL) (GIBCO). The medium was replaced every third day. THP-1 cells, JurkaTcells and Daudi can be obtained from The American Type Culture Collection (ATCC).
The effects of the overexpression of the RUNXl, RUNX2 and RUNX3 genes on the expression of PDCDl is shown in Figure 7, as an example.
29 References
Abruzzese RV, Godin D, Mehta V, Perrard JL, French M, Nelson W, Howell G, Coleman M, O'Malley BW, Nordstrom JL. Mol Ther 2:276-87, 2000.
Adya N, Castilla LH, Liu PP (2000) Function of CBFbeta/Bro proteins. Semin Cell Dev Biol 11 :361-368
Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubata T, Yagita H, Honjo T (1996) Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol 8:765-772
Backstrom S, Huang SH, Wolf-Watz M, Xie XQ, Hard T, Grundstrom T, Sauer UH (2001) Crystallization and preliminary studies of the DNA-binding runt domain of AML1. Acta Crystallogr D Biol Crystallogr 57:269-271
Backstrom S, Wolf-Watz M, Grundstrom C, Hard T, Grundstrom T, Sauer UH (2002) The RUNXl Runt domain at 1.25 A resolution: a structural switch and specifically bound chloride ions modulate DNA binding. J Mol Biol 322:259- 272
Barnes GL, Javed A, Waller SM, Kamal MH, Hebert KE, Hassan MQ, Bellahcene A, Van Wijnen AJ, Young MF, Lian JB, Stein GS, Gerstenfeld LC (2003) Osteoblast-related transcription factors Runx2 (Cbfal/AML3) and MSX2 mediate the expression of bone sialoprotein in human metastatic breast cancer cells. Cancer Res 63:2631-2637
Berse, B., J. A. Hunt, et al. (1999). "Hypoxia augments cytokine (transforming growth factor-beta (TGF-beta) and IL-l)-induced vascular endothelial growth factor secretion by human synovial fibroblasts." Clin Exp Immunol 115(1): 176-82
Bugge TH, Pohl J, Lonnoy O, Stunnenberg HG. EMBO J 11:1409-18, 1992 Bristow CA, Shore P (2003) Transcriptional regulation of the human MIP-1 alpha promoter by RUNXl and MOZ. Nucleic Acids Res 31 :2735-2744
Burns CE, DeBlasio T, Zhou Y, Zhang J, Zon L, Nimer SD (2002) Isolation and characterization of runxa and runxb, zebrafish members of the runt family of transcriptional regulators. Exp Hematol 30:1381-1389 Bushweller JH (2000) CBF~a biophysical perspective. Semin Cell Dev Biol 11 :377- 382 Cameron ER, Blyth K, Hanlon L, Kilbey A, Mackay N, Stewart M, Terry A, Vaillant F, Wotton S, Neil JC (2003) The Runx genes as dominant oncogenes. Blood Cells Mol Dis 30:194-200 Choi JY, Pratap J, Javed A, Zaidi SK, Xing L, Balint E, Dalamangas S, Boyce B, van Wijnen AJ, Lian JB, Stein JL, Jones SN, Stein GS (2001) Subnuclear targeting of Runx Cbfa/AML factors is essential for tissue-specific differentiation during embryonic development. Proc Natl Acad Sci U S A 98:8650-8655 Coffinan JA (2003) Runx transcription factors and the developmental balance between cell proliferation and differentiation. Cell Biol Int 27:315-324 Cohen MM, Jr. (2001) RUNX genes, neoplasia, and cleidocranial dysplasia. Am J Med Genet 104:185-188 Demasi, M., L. G. Cleland, et al. (2004). "Effects of hypoxia on the expression and activity of cyclooxygenase 2 in fibroblast-like synoviocytes: interactions with monocyte-derived soluble mediators." Arthritis Rheum 50(8): 2441-9 DeWitt SH, Kiely JS, Stankovic CJ, Schroeder MC, Cody DM, Pavia MR (1993) "Diversomers": an approach to nonpeptide, nonoligomeric chemical diversity. Proc Natl Acad Sci U S A 90:6909-6913 Eggers JH, Stock M, Fliegauf M, Vonderstrass B, Otto F (2002) Genomic characterization of the RUNX2 gene of Fugu rubripes. Gene 291:159-167 Feldmann M, Brennan FM, Maini R (1998) Cytokines in autoimmune disorders. Int Rev Immunol 17:217-228 Finger LR, Pu J, Wasserman R, Vibhakar R, Louie E, Hardy RR, Burrows PD, Billips LG (1997) The human PD-1 gene: complete cDNA, genomic organization, and developmentally regulated expression in B cell progenitors. Gene 197:177-187 Freundlieb S, Schirra-Muller C, Bujard H. J Gene Med 1 :4- 12, 1999 Fruman DA, Burakoff SJ, Bierer BE. FASEB J 8:391-400, 1994 Greenwald RJ, Latchman YE, Sharpe AH (2002) Negative co-receptors on lymphocytes. Curr Opin Immunol 14:391-396
Harrington KS, Javed A, Drissi H, McNeil S, Lian JB, Stein JL, Van Wijnen AJ, Wang YL, Stein GS (2002) Transcription factors RUNX1/AML1 and
31 RUNX2/Cbfal dynamically associate with stationary subnuclear domains. J Cell Sci 115:4167-4176 Hernandez M, Rodriguez-Lazaro D, Esteve T, Prat S, Pla M (2003) Development of melting temperature-based SYBR Green I polymerase chain reaction methods for multiplex genetically modified organism detection. Anal Biochem 323 : 164- 170 Hochberg MC (1997) Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 40:1725 Jackson, J. R., J. A. Minton, et al. (1997). "Expression of vascular endothelial growth factor in synovial fibroblasts is induced by hypoxia and interleukin lbeta." J Rheumatol 24(7): 1253-9 Javed A, Barnes GL, Jasanya BO, Stein JL, Gerstenfeld L, Lian JB, Stein GS (2001) runt homology domain transcription factors (Runx, Cbfa, and AML) mediate repression of the bone sialoprotein promoter: evidence for promoter context- dependent activity of Cbfa proteins. Mol Cell Biol 21:2891-2905 Komori T (2003) Requisite roles of Runx2 and Cbfb in skeletal development. J Bone Miner Metab 21 :193-197 Kundu M, Javed A, Jeon JP, Horner A, Shum L, Eckhaus M, Muenke M, Lian JB, Yang Y, Nuckolls GH, Stein GS, Liu PP (2002) Cbfbeta interacts with Runx2 and has a critical role in bone development. Nat Genet 32:639-644 Kundu M, Liu PP (2003) Cbfbeta is involved in maturation of all lineages of hematopoietic cells during embryogenesis except erythroid. Blood Cells Mol Dis 30:164-169 Lam KS, Lebl M, Krchnak V ( 1997) The "One-Bead-One-Compound" Combinatorial Library Method. Chem Rev 97:411-448 Lamartina S, Roscilli G, Rinaudo CD, Sporeno E, Silvi L, Hillen W, Bujard H, Cortese R, Ciliberto G, Toniatti C. Hum Gene Ther 13:199-210, 2002 Leboy P, Grasso-Knight G, DAngelo M, Volk SW, Lian JV, Drissi H, Stein GS, Adams SL (2001) Smad-Runx interactions during chondrocyte maturation. J Bone Joint Surg Am 83-A Suppl 1:S 15-22 Lian JB, Balint E, Javed A, Drissi H, Vitti R, Quinlan EJ, Zhang L, Van Wijnen AJ, Stein JL, Speck N, Stein GS (2003) Runxl/AMLl hematopoietic transcription factor contributes to skeletal development in vivo. J Cell Physiol 196:301-311
Lian JB, Stein GS (2003) Runx2/Cbfal: a multifunctional regulator of bone formation. Curr Pharm Des 9:2677-2685
Lund AH, van Lohuizen M (2002) RUNX: a trilogy of cancer genes. Cancer Cell 1:213-215
Luthra R, Sanchez- Vega B, Jeffrey Medeiros L (2004) TaqMan RT-PCR assay coupled with capillary electrophoresis for quantification and identification of bcr-abl transcript type. Mod Pathol 17:96-103
Lutterbach B, Westendorf JJ, Linggi B, Isaac S, Seto E, Hiebert SW (2000) A mechanism of repression by acute myeloid leukemia- 1, the target of multiple chromosomal translocations in acute leukemia. J Biol Chem 275:651-656
Maeda H, Fujimoto C, Haruki Y, Maeda T, Kokeguchi S, Petelin M, Arai H, Tanimoto I, Nishimura F, Takashiba S (2003) Quantitative real-time PCR using TaqMan and SYBR Green for Actinobacillus actinomycetemcomitans, Porphyromonas gingivalis, Prevotella intermedia, tetQ gene and total bacteria. FEMS Immunol Med Microbiol.39:81-86
Magnusson V, Nakken B, Bolstad Al, Alarcon-Riquelme ME (2001) Cytokine polymorphisms in systemic lupus erythematosus and Sjogren's syndrome. Scand J Immunol 54:55-61
McCarthy TL, Chang WZ, Liu Y, Centrella M (2003) Runx2 integrates estrogen activity in osteoblasts. J Biol Chem
Michaud J, Wu F, Osato M, Cottles GM, Yanagida M, Asou N, Shigesada K, Ito Y, Benson KF, Raskind WH, Rossier C, Antonarakis SE, Israels S, McNicol A, Weiss H, Horwitz M, Scott HS (2002) In vitro analyses of known and novel RUNXl/AMLl mutations in dominant familial platelet disorder with predisposition to acute myelogenous leukemia: implications for mechanisms of pathogenesis. Blood 99:1364-1372 Moss SF (2003) RUNX 3, apoptosis 0: a new gastric tumour suppressor. Gut 52:12-13
Figure imgf000035_0001
Nishimura H, Honjo T (2001) PD-1: an inhibitory immunoreceptor involved in peripheral tolerance. Trends Immunol 22:265-268 No D, Yao TP, Evans RM. Proc Natl Acad Sci U S A 93:3346-51, 1996 Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T (2001) PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad Sci U S A 98:13866-13871 Okuda T, Nishimura M, Nakao M, Fujita Y (2001) RUNX1/AML1: a central player in hematopoiesis. Int J Hematol 74:252-257 Otto F, Kanegane H, Mundlos S (2002) Mutations in the RUNX2 gene in patients with cleidocranial dysplasia. Hum Mutat 19:209-216 Otto F, Lubbert M, Stock M (2003) Upstream and downstream targets of RUNX proteins. J Cell Biochem 89:9-18 Perry C, Eldor A, Soreq H (2002) Runxl/AMLl in leukemia: disrupted association with diverse protein partners. Leuk Res 26:221-228
Prokunina L, Castillejo-Lopez C, Oberg F, Gunnarsson I, Berg L, Magnusson V, Brookes AJ, Tentler D, Kristjansdottir H, Grondal G, Bolstad Al, Svenungsson E, Lundberg I, Sturfelt G, Jonssen A, Truedsson L, Lima G, Alcocer-Varela J, Jonsson R, Gyllensten UB, Harley JB, Alarcon-Segovia D, Steinsson K, Alarcon-Riquelme ME (2002) A regulatory polymorphism in PDCD 1 is associated with susceptibility to systemic lupus erythematosus in humans. Nat Genet 32:666-669 Prud'homme GJ, Lawson BR, Theofilopoulos AN (2001) Anticytokine gene therapy of autoimmune diseases. Expert Opin Biol Ther 1:359-373 Sebbag M, Parry SL, Brennan FM, Feldmann M (1997) Cytokine stimulation of T lymphocytes regulates their capacity to induce monocyte production of tumor necrosis factor-alpha, but not interleukin-10: possible relevance to pathophysiology of rheumatoid arthritis. Eur J Immunol 27:624-632 Shinohara T, Taniwaki M, Ishida Y, Kawaichi M, Honjo T (1994) Structure and chromosomal localization of the human PD-1 gene (PDCDl). Genomics 23:704-706 Strieker S, Fundele R, Vortkamp A, Mundlos S (2002) Role of Runx genes in chondrocyte differentiation. Dev Biol 245:95-108 Tan EM, Cohen AS, Fries JF, Masi AT, McShane DJ, Rothfield NF, Schaller JG, Talal N, Winchester RJ (1982) The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 25:1271-1277
Urlinger S, Baron U, Thellmann M, Hasan MT, Bujard H, Hillen W. Proc Natl Acad Sci U S A 97:7963-8, 2000 Von Samson-Himmelstjerna G, Buschbaum S, Wirtherle N, Pape M, Schnieder T (2003) TaqMan minor groove binder real-time PCR analysis of beta-tubulin codon 200 polymorphism in small strongyles (Cyathostomin) indicates that the TAG allele is only moderately selected in benzimidazole-resistant populations. Parasitology 127:489-496 Walker LC, Stevens J, Campbell H, Corbett R, Spearing R, Heaton D, Macdonald DH, Morris CM, Ganly P (2002) A novel inherited mutation of the transcription factor RUNXl causes thrombocytopenia and may predispose to acute myeloid leukaemia. Br J Haematol 117:878-881 Zufferey R, Dull T, Mandel RJ, Bukovsky A, Quiroz D, Naldini L, Trono D. J Virol 72:9873-80, 1998
35

Claims

1. A method for screening and/or diagnosis of autoimmune disorders comprising the step of : a) obtaining biological sample from a test subject b) contacting the biological sample with at least one agent capable of detecting at least one of PDCDl, RUNXl, RUNX2 and/or RUNX3 nucleic acid or the corresponding expression product coded for by one or more of these genes or a subfragment thereof. c) comparing the expression rate of at least one expression product from PDCD 1 , RUNXl, RUNX2 and/or RUNX3 nucleic acid or a subfragment thereof from a healthy test subject with the expression rate thereof from another test subject.
2. The method according to claim 1, wherein the biological sample is chosen from pheripheral blood mononuclear cells such as lymphocytes, especially T- lymphocytes or CD4+ cells.
3. The method according to claim 2, wherein the lymphocytes are activated.
4. The method according to any of claims 1-3, wherein the nucleic acid which is detected is RNA such as mRNA, or DNA such as cDNA or genomic DNA.
5. The method according to any of claims 1-4, wherein the autoimmune disorder is chosen from : a) multiple sclerosis, b) myasthenia gravis, c) Type 1 diabetes, d) rheumatoid arthritis, e) Sjδgrens syndrome f) atopy, g) allergy, or
45 h) systemic lupus erythematosus, where systemic lupus erythematosus (SLE) is characterised by conditions selected from the group including any one or more; atigue, fever, loss of appetite, nausea, weight loss, hives, loss of scalp hair, red "butterfly rash" and raised rash, sensitivity to sun, ulcers in mouth, nose, or vagina, arthritis, joint pain, loss of blood supply to bone, pain, infections within joints, decrease in kidney function including, blood, aberrant amounts of protein or white blood cells in urine, intracerebral haemorrhage, headaches, loss of coordination, memory loss, seizures, strokes, anaemia, low white blood cell or low platelet count, pericardial effusion, heart attack, inflammation in the heart, infection in the heart, inflammation of the lining of the heart, infection of the lining of the heart, heart valve problems, shortness of breath, cough, inflammation of the lungs, inflammation of the lining of the lungs, abdominal distress, diarrhoea, enlargement of the liver, loss of appetite, nausea and vomiting, blindness, visual impairment, dryness of the eyes and dryness of the mouth.
6. The method according to any of claims 1-5, wherein the at least one agent is a nucleic acid probe capable of hybridizing to any of PDCD, RUNXl, RUNX2 or RUNX3 mRNA, cDNA or genomic DNA.
The method according to any of claims 1-6, wherein the probe is labelled.
8. The method according to any of claims 1-7, wherein the nucleic acid probe for detection of RUNXl is chosen from SEQ ID NO. 3, 4, 5; for detection of RUNX2 is chosen from SEQ ID NO 10, 11, 12; for detection of RUNX3, is chosen from SEQ ID NO 17, 18, 19; and for detection of PDCDl is chosen from SEQ ID NO SEQ NO. 26, 27 and 28 or a portion thereof.
9. The method according to any of claims 1-8, wherein systemic lupus erythematosus (SLE) is diagnosed when the rate of expression of PDCDl in CD4+ cells is higher in the tested subject that in a healthy test subject.
46
10. The method according to any of claims 1-8, wherein systemic lupus erythematosus (SLE) is diagnosed when the rate of expression of RUNXl in T - cells is higher in the tested subject that in a healthy test subject.
11. The method according to any of claims 1-8, wherein systemic lupus erythematosus (SLE) is diagnosed when the rate of expression of RUNXl and/or RUNX3 in T - cells and/or CD4+cells is higher in the tested subject that in a healthy test subject.
12. The method according to any of claims 1-8, wherein rheumatoid arthritis is diagnosed when the rate of expression of RUNXl and 2 in synovial fibroblasTcells is modulated in the tested subject compared to a healthy test subject when subjected to hypoxia or to TGFbeta.
13. A kit comprising at least one, nucleic acid probe capable of hybridizing to mRNA, cDNA or genomic DNA from RUNXl, RUNX2, RUNX3, PDCDl.
14. The kit according to claim 13, comprising at least 20, preferably at least 30, such as at least 40, most preferred at least 50 nucleotides capable of hybridizing to any of SEQ ID NO 1, 2, 6, 7, 8, 9, 13, 14, 15, 16, 20, 21, 22, 23, 24, 25.
15. The kit according to claim 13, comprising at least one nucleic nucleic acid probe chosen from SEQ ID NO. 3, 4, 5; 10, 11, 12; 17, 18, 19; 26, 27 and 28 or a portion thereof.
16. The kit according to any of claims 13-15, wherein the at least one probe is labelled.
47
17. A method for identifying compounds, that modulate PDCD 1 , RUNXl , RUNX2 or RUNX3 expression comprising step of : a) obtaining biological sample from a test subject 5 b) contacting the biological sample with at least one agent capable of detecting the PDCDl, RUNXl, RUNX2 and/or RUNX3 nucleic acid or the corresponding expression product coded for by these gene or a subfragment thereof, c) optionally adding a compound to be tested for modulation I o d) determining the rate of expression of at least one of the PDCD 1 , RUNX 1 , RUNX2 and/or RUNX3 expression products e) determining if the compound has induction or reduction effect on their expression.
15 18. A Pharmaceutical composition comprising a compound that modulates PDCDl, RUNXl, RUNX2 or RUNX3 expression identified by the step of : a) obtaining biological sample from a test subject b) contacting the biological sample with at least one agent capable of detecting the PDCDl, RUNXl, RUNX2 and/or RUNX3 nucleic acid or
20 the corresponding expression product coded for by these gene or a subfragment thereof, c) optionally adding a compound to be tested for modulation d) determining the rate of expression of at least one of the PDCD 1 , RUNX 1 , RUNX2 and/or RUNX3 expression products
25 e) determining if the compound has induction or reduction effect on their expression.
19. The composition according to claim 16, characterised in that the compound is an agonist, antagonist to the expression products of PDCDl, RUNXl, RUNX2 30 or RUNX3, peptidomimetic, peptide, nucleic acid such as anti sense DNA or RNA.
48
20. The composition according to claim 19, characterised in that the composition comprises a vector comprising SEQ ID NO 29 and 30 or SEQ ID NO 31 and 32 or anyone of SEQ ID NO 35-37.
21. Use of a composition according to any of claims 18 or 19 for the preparation of a pharmaceutical composition for the treatment of autoimmune disorders such as: a) multiple sclerosis, b) myasthenia gravis, c) Type 1 diabetes, d) rheumatoid arthritis, e) Sjδgrens syndrome f) atopy, g) allergy, or h) systemic lupus erythematosus, where systemic lupus erythematosus (SLE) is characterised by conditions selected from the group including any one or more; fatigue, fever, loss of appetite, nausea, weight loss, hives, loss of scalp hair, red "butterfly rash" and raised rash, sensitivity to sun, ulcers in mouth, nose, or vagina, arthritis, joint pain, loss of blood supply to bone, pain, infections within joints, decrease in kidney function including, blood, aberrant amounts of protein or white blood cells in urine, intracerebral haemorrhage, headaches, loss of coordination, memory loss, seizures, strokes, anaemia, low white blood cell or low platelet count, pericardial effusion, heart attack, inflammation in the heart, infection in the heart, inflammation of the lining of the heart, infection of the lining of the heart, heart valve problems, shortness of breath, cough, inflammation of the lungs, inflammation of the lining of the lungs, abdominal distress, diarrhea, enlargement of the liver, loss of appetite, nausea and vomiting, blindness, visual impairment, dryness of the eyes and dryness of the mouth.
49 A method for the treatment of a patient suffering from an autoimmune disease wherein a compound that is an agonist, antagonist to the expression products of PDCDl, RUNXl, RUNX2 or RUNX3, peptidomimetic, peptide, nucleic acid such as anti sense DNA or RNA. Is administrated.
50
PCT/SE2005/000414 2004-03-22 2005-03-22 Method and kit for screening, pharmaceutical compositions and method for treatment of autoimmune disorders WO2005089080A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US55517904P 2004-03-22 2004-03-22
US60/555,179 2004-03-22

Publications (2)

Publication Number Publication Date
WO2005089080A2 true WO2005089080A2 (en) 2005-09-29
WO2005089080A3 WO2005089080A3 (en) 2005-11-17

Family

ID=34994135

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SE2005/000414 WO2005089080A2 (en) 2004-03-22 2005-03-22 Method and kit for screening, pharmaceutical compositions and method for treatment of autoimmune disorders

Country Status (1)

Country Link
WO (1) WO2005089080A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018084706A1 (en) * 2016-11-04 2018-05-11 Erasmus University Medical Center Rotterdam Markers for identifying patient classes and use thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003022875A2 (en) * 2001-09-07 2003-03-20 Everygene Ab Polymorphisms of pd-1
US20030232323A1 (en) * 2001-11-13 2003-12-18 Wyeth Agents that modulate immune cell activation and methods of use thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003022875A2 (en) * 2001-09-07 2003-03-20 Everygene Ab Polymorphisms of pd-1
US20030232323A1 (en) * 2001-11-13 2003-12-18 Wyeth Agents that modulate immune cell activation and methods of use thereof

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
ALARCON-RIQUELME M.E. ET AL: 'A runx trio with a taste for autoimmunity' NATURE GENETICS vol. 35, no. 4, December 2003, pages 299 - 300, XP002991618 *
ALARCON-RIQUELME M.E. ET AL: 'Finding genes for SLE: complex interactions and complex populations' JOURNAL OF AUTOIMMUNITY vol. 21, 2003, pages 17 - 20, XP002991619 *
DATABASE GENBANK [Online] 02 July 2003 XP002991615 Retrieved from NCBI Database accession no. (AX770853) *
HATACHI S. ET AL: 'DC4+ PD-1+ T Cells Accumulate as Unique Anergic Cells in Rheumatoid Arthritis Synovial Fluid' J RHEUMATOL vol. 30, 2003, pages 1410 - 1419, XP008053453 *
NIELSEN C. ET AL: 'Association of a putative regulatory polymorphism in the PD-1 gene with susceptibility to type 1 diabetes' TISSUE ANTIGENS vol. 62, no. 6, December 2003, pages 492 - 497, XP002991620 *
NISHIMURA H. ET AL: 'Development of Lupus-like Autoimmune Diseases by Disruption of the PD-1 Gene Encoding an ITIM Motif-Carrying Immunoreceptor' IMMUNITY vol. 11, August 1999, pages 141 - 151, XP000971788 *
PROKUNINA L. ET AL: 'A regulatory polyorphism in PDCD1 is associated with susceptibility to systemic lupus erythematosus in humans' NATURE GENETICS vol. 32, December 2002, pages 666 - 669, XP002243477 *
PROKUNINA L. ET AL: 'The genetic basis of systemic lupus erythematosus-knowledge of today and thoughts for tomorrow' HUMAN MOLECULAR GENETICS vol. 13, no. 1, 2004, pages R143 - R148, XP002991616 *
TOKUHIRO S. ET AL: 'An intronic SNP in a RUNX1 binding site of SLC22A4, encoding an organic cation transorter, is associated with rheumatoid arthritis' NATURE GENETICS vol. 35, no. 4, December 2003, pages 341 - 348, XP002991617 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018084706A1 (en) * 2016-11-04 2018-05-11 Erasmus University Medical Center Rotterdam Markers for identifying patient classes and use thereof

Also Published As

Publication number Publication date
WO2005089080A3 (en) 2005-11-17

Similar Documents

Publication Publication Date Title
Lu et al. Interferon (IFN) beta acts downstream of IFN-gamma-induced class II transactivator messenger RNA accumulation to block major histocompatibility complex class II gene expression and requires the 48-kD DNA-binding protein, ISGF3-gamma.
Kim et al. Both integrated and differential regulation of components of the IL-2/IL-2 receptor system
Grumont et al. Rel induces interferon regulatory factor 4 (IRF-4) expression in lymphocytes: modulation of interferon-regulated gene expression by rel/nuclear factor κB
Rosselli et al. Abnormal lymphokine production: a novel feature of the genetic disease Fanconi anemia. II. In vitro and in vivo spontaneous overproduction of tumor necrosis factor alpha
Gong et al. Cytokine-dependent Blimp-1 expression in activated T cells inhibits IL-2 production
Neurath et al. The transcription factor T-bet regulates mucosal T cell activation in experimental colitis and Crohn's disease
Niu et al. BCL6 controls the expression of the B7-1/CD80 costimulatory receptor in germinal center B cells
Kusam et al. Inhibition of Th2 differentiation and GATA-3 expression by BCL-6
Li et al. Kaposi’s sarcoma-associated herpesvirus viral interferon regulatory factor
Nourbakhsh et al. Constitutive silencing of IFN-β promoter is mediated by NRF (NF-κB-repressing factor), a nuclear inhibitor of NF-κB
Yang et al. Identification of a distant T-bet enhancer responsive to IL-12/Stat4 and IFNγ/Stat1 signals
Kim et al. CREB/ATF-dependent T cell receptor–induced FoxP3 gene expression: a role for DNA methylation
Friedman Leukemogenesis by CBF oncoproteins
Zhang et al. Interleukin 2 receptor signaling regulates the perforin gene through signal transducer and activator of transcription (Stat) 5 activation of two enhancers
Ott et al. Superinduction of IL-8 in T cells by HIV-1 Tat protein is mediated through NF-κB factors
Park et al. Comparative analysis of genes regulated by PML/RARα and PLZF/RARα in response to retinoic acid using oligonucleotide arrays
Villiger et al. IL-6 production by human T lymphocytes. Expression in HTLV-1-infected but not in normal T cells.
Bruns et al. Expression of a constitutively active Stat6 in vivo alters lymphocyte homeostasis with distinct effects in T and B cells
Lee et al. Differential regulation of constitutive major histocompatibility complex class I expression in T and B lymphocytes
Kirchhoff et al. Viral IFN-regulatory factors inhibit activation-induced cell death via two positive regulatory IFN-regulatory factor 1-dependent domains in the CD95 ligand promoter
CA2143261A1 (en) Method of diagnosing cancer, precancerous state, or susceptibility to other forms of diseases by analysis of irf-1-specific rna in biopsy samples
Li‐Weber et al. A regulatory element in the CD95 (APO‐1/Fas) ligand promoter is essential for responsiveness to TCR‐mediated activation
Wu et al. Genomic organization and characterization of mouse SAP, the gene that is altered in X-linked lymphoproliferative disease
Li et al. MxA overexpression reveals a common genetic link in four Fanconi anemia complementation groups.
Zhao et al. Stat2-dependent regulation of MHC class II expression

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

NENP Non-entry into the national phase in:

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

122 Ep: pct application non-entry in european phase