WO2019207066A1 - Procédés et compositions destinés au traitement du syndrome de sjögren - Google Patents

Procédés et compositions destinés au traitement du syndrome de sjögren Download PDF

Info

Publication number
WO2019207066A1
WO2019207066A1 PCT/EP2019/060662 EP2019060662W WO2019207066A1 WO 2019207066 A1 WO2019207066 A1 WO 2019207066A1 EP 2019060662 W EP2019060662 W EP 2019060662W WO 2019207066 A1 WO2019207066 A1 WO 2019207066A1
Authority
WO
WIPO (PCT)
Prior art keywords
tlr7
mice
tlr8ko
antagonist
antibodies
Prior art date
Application number
PCT/EP2019/060662
Other languages
English (en)
Inventor
Léna ALEXOPOULOU
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Centre National De La Recherche Scientifique (Cnrs)
Université D'aix Marseille
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by INSERM (Institut National de la Santé et de la Recherche Médicale), Centre National De La Recherche Scientifique (Cnrs), Université D'aix Marseille filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2019207066A1 publication Critical patent/WO2019207066A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders

Definitions

  • the present invention relates to methods and compositions for the treatment of Sjogren’s syndrome.
  • Sjogren’s syndrome is a chronic, systemic autoimmune disease characterized by chronic inflammation of salivary and lacrimal glands, leading to dry mouth and eyes. Clinical presentation can vary considerably from relative mild sicca symptoms, arthralgias, and fatigue to severe systemic symptoms, such as vasculitis, glomerulonephritis, and neurological manifestations (Mavragani, C. P., and H. M. Moutsopoulos. 2014. Sjogren’s syndrome. Annual review of pathology 9: 273-285).
  • SS may occur as an isolated disorder, which is termed primary SS, or it may occur in association with other autoimmune disorders, such as rheumatoid arthritis (SA) or systemic lupus erythematosus (SLE), termed as secondary SS.
  • SA rheumatoid arthritis
  • SLE systemic lupus erythematosus
  • SA rheumatoid arthritis
  • SLE systemic lupus erythematosus
  • SS has a very strong genetic and gender bias, and interaction with environmental factors, microbial infections, particularly those by viruses, have been hypothesized and even experimentally demonstrated to constitute an important trigger for SS (Ohyama, Y., et al. 2006. Severe focal sialadenitis and dacryoadenitis in NZM2328 mice induced by MCMV: a novel model for human Sjogren’s syndrome. J Immunol 177: 7391- 7397).
  • Viral nucleic acids are potent stimulators of Toll- like receptor 3 (TLR3), TLR7, TLR8 and TLR9 that can detect nucleic acids of microbial or endogenous origin. Engagement of these TLRs results in the activation of multiple signalling pathways that culminate in the production of proinflammatory cytokines such as IL-6, TNF-a, and type I interferons (IFNs) (Kopp, E., and R. Medzhitov. 2003. Recognition of microbial infection by Toll-like receptors. Curr Opin Immunol 15: 396-401). A chronic or repeated infection can create an inflammatory environment leading to organ dysfunction and development of autoimmune disease.
  • TLR3 Toll- like receptor 3
  • TLR7, TLR8 and TLR9 can detect nucleic acids of microbial or endogenous origin. Engagement of these TLRs results in the activation of multiple signalling pathways that culminate in the production of proinflammatory cytokines such as IL-6, TNF-a, and type I
  • TLRs and B cells play a pivotal role in the pathogenesis of autoimmune diseases such as SLE, rheumatoid arthritis and SS (Berglova, L, et al. 2011. B cell toll-like receptors with respect to the pathogenesis of Sjogren's syndrome. Acta Medica (Hradec Kralove) 54: 51-57).
  • TLRs and B cells play a pivotal role in the pathogenesis of autoimmune diseases such as SLE, rheumatoid arthritis and SS.
  • Bova, L, et al. 2011 B cell toll-like receptors with respect to the pathogenesis of Sjogren's syndrome. Acta Medica (Hradec Kralove) 54: 51-57).
  • the exact role of individual TLRs in the development and progression of SS is still unknown.
  • TLR7 signalling in the development of SLE in humans has been confirmed by recent studies where increased expression of TLR7 is associated with increased risk for SLE (Deng, Y., et al. 2013. MicroRNA-3l48 modulates allelic expression of toll-like receptor 7 variant associated with systemic lupus erythematosus. PLoS Genet 9: el003336.). Thus, tight control and regulation of TLR7 is pivotal for avoiding SLE development. However, the contribution of TLR7 in SS development and progression is still unknown.
  • the present invention relates to methods and compositions for the treatment of Sjogren’s syndrome.
  • the invention is defined by the claims.
  • the present invention relates to methods and compositions for the treatment of Sjogren’s syndrome (SS). To date effective therapy is not available and treatment has been mainly symptomatic.
  • TLR8ko mice investigated the implication of TLR7 in the development of SS using as animal models TLR8ko that spontaneous develop systemic lupus erythematosus (SLE) due to increased TLR7 signaling.
  • SLE systemic lupus erythematosus
  • TLR8ko mice developed spontaneous SS that is characterized by the presence of inflammatory foci in salivary glands and reduced production of saliva upon pilocarpin injection, increased semm levels of SSA and SSB autoantibodies, and an inflammatory signature in salivary glands similar to that seen in human SS patients.
  • double TLR7/8ko mice did not show any signs of SS.
  • old wild-type mice developed spontaneous inflammatory foci in their salivay glands, but TLR7ko mice were protected.
  • the present invention relates to a method of treating or preventing Sjogren’s syndrome in a subject in need thereof comprising administering the subject with a therapeutically effective amount of an antagonist of Toll-like receptor 7 (TLR7).
  • TLR7 Toll-like receptor 7
  • the TLR7 antagonist is a specific antagonist of TLR7.
  • Sjogren’s syndrome as used herein is an autoimmune disease or disorder in which immune cells attack the glands that produce tears and saliva. The hallmark symptoms of the disorder are dry mouth and dry eyes.
  • Sjogren’s syndrome may cause skin, nose, and vaginal dryness, and may affect other organs of the body including the kidneys, blood vessels, lungs, liver, pancreas, and brain.
  • Sjogren’s syndrome can exist as a primary disorder (“primary Sjogren’s syndrome”) or as a secondary disorder (“secondary Sjogren’s syndrome”) that is associated with and/or secondary to other autoimmune disorders including rheumatic disorders such as rheumatoid arthritis, systemic lupus, polymyositis, scleroderma, and autoimmune hepatitis, lymphomas such as non-Hodgkin's lymphoma, and endocrine disorders such as thyroiditis.
  • the term " Sjogren’s syndrome” as used herein applies to Sjogren’s syndrome no matter what the stage, including both primary and secondary Sjogren’ s syndrome, and no matter what symptoms are evident, provided the diagnosis is made.
  • a subject denotes a mammal, such as a rodent, a feline, a canine, and a primate.
  • a subject according to the invention is a human.
  • the subject is a woman.
  • beneficial or desired clinical results include, but are not limited to, one or more of the following: alleviating one or more symptoms resulting from the disease, diminishing the extent of the disease, stabilizing the disease (e.g., preventing or delaying the worsening of the disease), preventing or delaying the spread of the disease, preventing or delaying the recurrence of the disease, delay or slowing the progression of the disease, ameliorating the disease state, providing a remission (partial or total) of the disease, decreasing the dose of one or more other medications required to treat the disease, delaying the progression of the disease, increasing the quality of life, and/or prolonging survival.
  • treatment encompasses the prophylactic treatment.
  • the term "preventing” refers to the reduction in the risk of acquiring or developing a given condition, or the reduction or inhibition of the recurrence or said condition in a subject who is not ill, but who has been or may be near a subject with the disease.
  • the present invention also refers to a method of treating or preventing salivary glands inflammation in a subject in need thereof comprising administering the subject with a therapeutically effective amount of an antagonist of Toll-like receptor 7 (TLR7).
  • TLR7 Toll-like receptor 7
  • a further object of the present invention relates to a method of treating or preventing inflammatory foci in salivary glands in a subject in need thereof comprising administering the subject with a therapeutically effective amount of an antagonist of Toll-like receptor 7 (TLR7).
  • TLR7 Toll-like receptor 7
  • a further object of the present invention relates to a method of treating or preventing lacrimal glands inflammation in a subject in need thereof comprising administering the subject with a therapeutically effective amount of an antagonist of Toll-like receptor 7 (TLR7).
  • TLR7 Toll-like receptor 7
  • a further object of the present invention relates to a method of preventing immune cell infiltration in a subject in need thereof comprising administering the subject with a therapeutically effective amount of an antagonist of Toll-like receptor 7 (TLR7).
  • TLR7 Toll-like receptor 7
  • a further object of the present invention relates to a method of preventing destruction of the salivary gland structure and/or loss of salivary gland function in a subject in need thereof comprising administering the subject with a therapeutically effective amount of an antagonist of Toll-like receptor 7 (TLR7).
  • TLR7 Toll-like receptor 7
  • a further object of the present invention relates to a method of preventing saliva production decrease in a subject in need thereof comprising administering the subject with a therapeutically effective amount of an antagonist of Toll-like receptor 7 (TLR7).
  • TLR7 Toll-like receptor 7
  • a further object of the present invention relates to a method of preventing circulating antinuclear antibodies production in a subject in need thereof comprising administering the subject with a therapeutically effective amount of an antagonist of Toll-like receptor 7 (TLR7).
  • a further object of the present invention relates to a method of preventing salivary glands cytokines production in a subject in need thereof comprising administering the subject with a therapeutically effective amount of an antagonist of Toll-like receptor 7 (TLR7).
  • TLR7 has its general meaning in the art and refers to the protein (Uniprot reference: Q9NYK1) encoded by the TLR7 gene (NCBI reference gen ID for Homo sapiens: 51284).
  • TLR7 antagonist has its general meaning in the art and refers to any compound, natural or synthetic, that blocks, suppresses, or reduces the biological activity of inhibits the activity of TLR7, or to any compound that inhibit TLR7 gene expression.
  • the TLR7 antagonist is a small organic molecule.
  • small organic molecule refers to a molecule of a size comparable to those organic molecules generally used in pharmaceuticals. The term excludes biological macromolecules (e. g., proteins, nucleic acids, etc.). Preferred small organic molecules range in size up to about 5000 Da, more in particular up to 2000 Da, and most in particular up to about 1000 Da.
  • TLR7 may be present into the cell or/and at the cell surface
  • antibodies and intrabodies may be used as TLR7 antagonists.
  • the TLR7 antagonist is an antibody or a portion thereof.
  • the TLR7 antagonist is an antibody such as chimeric antibodies, humanized antibodies or full human monoclonal antibodies.
  • the TLR7 antagonist is an intrabody.
  • antibody includes both naturally occurring and non-naturally occurring antibodies. Specifically, “antibody” includes polyclonal and monoclonal antibodies, and monovalent and divalent fragments thereof. Furthermore, “antibody” includes chimeric antibodies, wholly synthetic antibodies, single chain antibodies, and fragments thereof. The antibody may be a human or nonhuman antibody. A nonhuman antibody may be humanized by recombinant methods to reduce its immunogenicity in man.
  • the antibody is a monoclonal antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a full human monoclonal antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a polyclonal antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a humanized antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a chimeric antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a light chain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a heavy chain of the antibody.
  • the portion of the antibody comprises a Fab portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a F(ab')2 portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fc portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fv portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a variable domain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises one or more CDR domains of the antibody.
  • Antibodies are prepared according to conventional methodology. Monoclonal antibodies may be generated using the method of Kohler and Milstein (Nature, 256:495, 1975). To prepare monoclonal antibodies useful in the invention, a mouse or other appropriate host animal is immunized at suitable intervals (e.g., twice-weekly, weekly, twice-monthly or monthly) with antigenic forms of TLR7. The animal may be administered a final "boost" of antigen within one week of sacrifice. It is often desirable to use an immunologic adjuvant during immunization.
  • Suitable immunologic adjuvants include Freund's complete adjuvant, Freund's incomplete adjuvant, alum, Ribi adjuvant, Hunter's Titermax, saponin adjuvants such as QS21 or Quil A, or CpG-containing immunostimulatory oligonucleotides.
  • Other suitable adjuvants are well-known in the field.
  • the animals may be immunized by subcutaneous, intraperitoneal, intramuscular, intravenous, intranasal or other routes. A given animal may be immunized with multiple forms of the antigen by multiple routes.
  • recombinant TLR7 may be provided by expression with recombinant cell lines.
  • compositions and methods that include humanized forms of antibodies.
  • humanized describes antibodies wherein some, most or all of the amino acids outside the CDR regions are replaced with corresponding amino acids derived from human immunoglobulin molecules.
  • Methods of humanization include, but are not limited to, those described in U.S. Pat. Nos. 4,816,567, 5,225,539, 5,585,089, 5,693,761, 5,693,762 and 5,859,205, which are hereby incorporated by reference.
  • the above U.S. Pat. Nos. 5,585,089 and 5,693,761, and WO 90/07861 also propose four possible criteria which may used in designing the humanized antibodies.
  • the first proposal was that for an acceptor, use a framework from a particular human immunoglobulin that is unusually homologous to the donor immunoglobulin to be humanized, or use a consensus framework from many human antibodies.
  • the second proposal was that if an amino acid in the framework of the human immunoglobulin is unusual and the donor amino acid at that position is typical for human sequences, then the donor amino acid rather than the acceptor may be selected.
  • the third proposal was that in the positions immediately adjacent to the 3 CDRs in the humanized immunoglobulin chain, the donor amino acid rather than the acceptor amino acid may be selected.
  • the fourth proposal was to use the donor amino acid reside at the framework positions at which the amino acid is predicted to have a side chain atom within 3A of the CDRs in a three dimensional model of the antibody and is predicted to be capable of interacting with the CDRs.
  • the above methods are merely illustrative of some of the methods that one skilled in the art could employ to make humanized antibodies.
  • One of ordinary skill in the art will be familiar with other methods for antibody humanization.
  • the present invention also provides for F(ab')2, Fab, Fv and Fd fragments; chimeric antibodies in which the Fc and/or FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric F(ab')2 fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric Fab fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; and chimeric Fd fragment antibodies in which the FR and/or CDR1 and/or CDR2 regions have been replaced by homologous human or non-human sequences.
  • the present invention also includes so-called single chain antibodies.
  • the various antibody molecules and fragments may derive from any of the commonly known immunoglobulin classes, including but not limited to IgA, secretory IgA, IgE, IgG and IgM.
  • IgG subclasses are also well known to those in the art and include but are not limited to human IgGl, IgG2, IgG3 and IgG4.
  • the antibody according to the invention is a single domain antibody.
  • the term“single domain antibody” (sdAb) or "VHH” refers to the single heavy chain variable domain of antibodies of the type that can be found in Camelid mammals which are naturally devoid of light chains. Such VHH are also called“nanobody®”.
  • the TLR7 antagonist is an intrabody.
  • intrabody refers to an antibody that binds to an intracellular protein, such as TLR7. Intrabodies are defined as antibodies that have been modified for intracellular localization.
  • Intrabodies require special alterations, including the use of single chain antibodies, modification of immunoglobulin VL domains for hyperstability, selection of antibodies resistant to the more reducing intracellular environment, or expression as a fusion protein with maltose binding protein or other stable intracellular proteins. Such optimizations have improved the stability and structure of intrabodies.
  • the TLR7 antagonist is an aptamer.
  • Aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition.
  • Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.
  • Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library.
  • the random sequence library is obtainable by combinatorial chemical synthesis of DNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence.
  • Peptide aptamers consists of a conformationally constrained antibody variable region displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods.
  • the TLR7 antagonist is a polypeptide.
  • the polypeptide is a functional equivalent of TLR7.
  • a“functional equivalent” of TLR7 is a compound which is capable of binding to TLR7 ligand, thereby preventing its interaction with TLR7.
  • the term “functional equivalent” includes fragments, mutants, and muteins of TLR7.
  • the term “functionally equivalent” thus includes any equivalent of TLR7 obtained by altering the amino acid sequence, for example by one or more amino acid deletions, substitutions or additions such that the protein analogue retains the ability to bind to TLR7 ligand. Amino acid substitutions may be made, for example, by point mutation of the DNA encoding the amino acid sequence.
  • the polypeptide of the present invention is fused to a heterologous polypeptide to form a fusion protein.
  • a“fusion protein” comprises all or part (typically biologically active) of a polypeptide of the present invention operably linked to a heterologous polypeptide (i.e., a polypeptide other than the same polypeptide).
  • the term "operably linked” is intended to indicate that the polypeptide of the present invention and the heterologous polypeptide are fused in-frame to each other.
  • the heterologous polypeptide can be fused to the N-terminus or C-terminus of the polypeptide of the present invention. In some embodiment, the heterologous polypeptide is fused to the C- terminal end of the polypeptide of the present invention.
  • polypeptides of the invention may be produced by any suitable means, as will be apparent to those of skill in the art.
  • expression may conveniently be achieved by culturing under appropriate conditions recombinant host cells containing the polypeptide of the invention.
  • the polypeptide is produced by recombinant means, by expression from an encoding nucleic acid molecule.
  • Systems for cloning and expression of a polypeptide in a variety of different host cells are well known. When expressed in recombinant form, the polypeptide is in particular generated by expression from an encoding nucleic acid in a host cell. Any host cell may be used, depending upon the individual requirements of a particular system.
  • Suitable host cells include bacteria mammalian cells, plant cells, yeast and baculovirus systems. Mammalian cell lines available in the art for expression of a heterologous polypeptide include Chinese hamster ovary cells. HeLa cells, baby hamster kidney cells and many others. Bacteria are also preferred hosts for the production of recombinant protein, due to the ease with which bacteria may be manipulated and grown. A common, preferred bacterial host is E coli.
  • polypeptides of the invention, fragments thereof and fusion proteins can exhibit post-translational modifications, including, but not limited to glycosylations, (e.g., N-linked or O-linked glycosylations), myristylations, palmitylations, acetylations and phosphorylations (e.g., serine/threonine or tyrosine).
  • glycosylations e.g., N-linked or O-linked glycosylations
  • myristylations e.g., palmitylations
  • acetylations e.g., serine/threonine or tyrosine
  • polypeptides used in the therapeutic methods of the present invention may be modified in order to improve their therapeutic efficacy.
  • modification of therapeutic compounds may be used to decrease toxicity, increase circulatory time, or modify biodistribution.
  • the toxicity of potentially important therapeutic compounds can be decreased significantly by combination with a variety of drug carrier vehicles that modify biodistribution.
  • adding dipeptides can improve the penetration of a circulating agent in the eye through the blood retinal barrier by using endogenous transporters.
  • the TLR7 antagonist is an inhibitor of expression.
  • said inhibitor of gene expression is a siRNA, an antisense oligonucleotide or a ribozyme.
  • Anti-sense oligonucleotides including anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of target gene mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of TLR7, and thus activity, in a cell.
  • antisense oligonucleotides complementary to unique regions of the mRNA transcript sequence encoding TLR7 can be synthesized, e.g., by conventional phosphodiester techniques.
  • Methods for using antisense techniques for specifically inhibiting gene expression of genes whose sequence is known are well known in the art (e.g. see U.S. Pat. Nos. 6,566,135; 6,566,131; 6,365,354; 6,410,323; 6,107,091; 6,046,321; and 5,981,732).
  • Small inhibitory RNAs can function as inhibitors of TLR7 gene expression for use in the present invention.
  • TLR7 gene expression can be reduced by contacting a subject or cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that TLR7 gene expression is specifically inhibited (i.e. RNA interference or RNAi).
  • dsRNA small double stranded RNA
  • RNAi RNA interference
  • Methods for selecting an appropriate dsRNA or dsRNA-encoding vector are well known in the art for genes whose sequence is known (e.g. see for example Tuschl, T. et al. (1999); Elbashir, S. M. et al. (2001); Hannon, GJ.
  • Ribozymes can also function as inhibitors of TLR7 gene expression for use in the present invention.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of TLR7 mRNA sequences are thereby useful within the scope of the present invention.
  • ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site can be evaluated for predicted structural features, such as secondary structure, that can render the oligonucleotide sequence unsuitable. The suitability of candidate targets can also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using, e.g., ribonuclease protection assays.
  • antisense oligonucleotides and ribozymes useful as inhibitors of TLR7 gene expression can be prepared by known methods. These include techniques for chemical synthesis such as, e.g., by solid phase phosphoramadite chemical synthesis. Alternatively, anti-sense RNA molecules can be generated by in vitro or in vivo transcription of DNA sequences encoding the RNA molecule. Such DNA sequences can be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Various modifications to the oligonucleotides of the invention can be introduced as a means of increasing intracellular stability and half-life.
  • Possible modifications include but are not limited to the addition of flanking sequences of ribonucleotides or deoxyribonucleotides to the 5' and/or 3' ends of the molecule, or the use of phosphorothioate or 2'-0-methyl rather than phosphodiesterase linkages within the oligonucleotide backbone.
  • Antisense oligonucleotides siRNAs and ribozymes of the invention may be delivered in vivo alone or in association with a vector.
  • a "vector" is any vehicle capable of facilitating the transfer of the antisense oligonucleotide siRNA or ribozyme nucleic acid to the cells and preferably cells expressing TLR7.
  • the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector.
  • the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the antisense oligonucleotide siRNA or ribozyme nucleic acid sequences.
  • Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma vims; adenovirus, adeno-associated vims; SV40-type vimses; polyoma vimses; Epstein-Barr vimses; papilloma vimses; herpes vims; vaccinia vims; polio vims; and RNA vims such as a retro vims.
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma vims
  • adenovirus adeno-associated vims
  • SV40-type vimses polyoma vimses
  • Non-cytopathic vimses include retrovimses (e.g., lentivims), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA. Retrovimses have been approved for human gene therapy trials. Most useful are those retrovimses that are replication-deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle). Such genetically altered retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo.
  • viruses for certain applications are the adeno-viruses and adeno-associated viruses, which are double- stranded DNA viruses.
  • the adeno-associated virus can be engineered to be replication deficient and is capable of infecting a wide range of cell types and species.
  • the adeno-associated virus can integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection.
  • wild-type adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event.
  • the adeno-associated virus can also function in an extrachromosomal fashion.
  • Other vectors include plasmid vectors.
  • Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g. Sambrook et a , 1989. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigen-encoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operatively encoded within the plasmid. Some commonly used plasmids include pBR322, pUCl8, pUCl9, pRC/CMV, SV40, and pBlueScript.
  • Plasmids are well known to those of ordinary skill in the art. Additionally, plasmids may be custom designed using restriction enzymes and ligation reactions to remove and add specific fragments of DNA. Plasmids maybe delivered by a variety of parenteral, mucosal and topical routes. For example, the DNA plasmid can be injected by intramuscular, eye, intradermal, subcutaneous, or other routes. It may also be administered by intranasal sprays or drops, rectal and orally. It may also be administered into the epidermis or a mucosal surface using a gene-gun.
  • the plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
  • the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid sequence is under the control of a heterologous regulatory region, e.g., a heterologous promoter.
  • an endonuclease is used for silencing the TLR7 gene.
  • the "CRISPR/Cas9" technology is used for silencing the TLR7 gene.
  • CRISPR has its general meaning in the art and refers to clustered regularly interspaced short palindromic repeats associated which are the segments of prokaryotic DNA containing short repetitions of base sequences.
  • the CRISPR/Cas loci encode RNA-guided adaptive immune systems against mobile genetic elements (viruses, transposable elements and conjugative plasmids).
  • I-III Three types (I-III) of CRISPR systems have been identified.
  • CRISPR clusters contain spacers, the sequences complementary to antecedent mobile elements.
  • CRISPR clusters are transcribed and processed into mature CRTS PR (Clustered Regularly Interspaced Short Palindromic Repeats) RNA (crRNA).
  • the CRISPR- associated endonuclease belongs to the type II CRISPR/Cas system and has strong endonuclease activity to cut target DNA.
  • Cas9 is guided by a mature crRNA that contains about 20 base pairs (bp) of unique target sequence (called spacer) and a trans-activated small RNA (tracrRNA) that serves as a guide for ribonuclease Ill-aided processing of pre-crRNA.
  • spacer unique target sequence
  • tracrRNA trans-activated small RNA
  • the crRNA dracrRNA duplex directs Cas9 to target DNA via complementary base pairing between the spacer on the crRNA and the complementary sequence (called protospacer) on the target DNA.
  • Cas9 recognizes a trinucleotide (NGG) protospacer adjacent motif (PAM) to specify the cut site (the 3rd nucleotide from PAM).
  • the crRNA and tracrRNA can be expressed separately or engineered into an artificial fusion small guide RNA (sgRNA) via a synthetic stem loop to mimic the natural crRNA/tracrRNA duplex.
  • sgRNA like shRNA, can be synthesized or in vitro transcribed for direct RNA transfection or expressed from U6 or Hi-promoted RNA expression vector, although cleavage efficiencies of the artificial sgRNA are lower than those for systems with the crRNA and tracrRNA expressed separately.
  • the CRIS PR-associated endonuclease can be a Cas9 nuclease.
  • the Cas9 nuclease can have a nucleotide sequence identical to the wild type Streptococcus pyrogenes sequence.
  • the CRISPR-associated endonuclease can be a sequence from other species, for example other Streptococcus species, such as thermophilus; Pseudomona aeruginosa, Escherichia coli, or other sequenced bacteria genomes and archaea, or other prokaryotic microogranisms.
  • the wild type Streptococcus pyrogenes Cas9 sequence can be modified.
  • the nucleic acid sequence can be codon optimized for efficient expression in mammalian cells, i.e., "humanized.”
  • a humanized Cas9 nuclease sequence can be for example, the Cas9 nuclease sequence encoded by any of the expression vectors listed in Genbank accession numbers KM099231.1 GL669193757; KM099232.1 GL669193761; or KM099233.l GL669193765.
  • the Cas9 nuclease sequence can be for example, the sequence contained within a commercially available vector such as PX330 or PX260 from Addgene (Cambridge, MA).
  • the Cas9 endonuclease can have an amino acid sequence that is a variant or a fragment of any of the Cas9 endonuclease sequences of Genbank accession numbers KM099231.1 GL669193757; KM099232.1; GL669193761; or
  • the Cas9 nucleotide sequence can be modified to encode biologically active variants of Cas9, and these variants can have or can include, for example, an amino acid sequence that differs from a wild type Cas9 by virtue of containing one or more mutations (e.g., an addition, deletion, or substitution mutation or a combination of such mutations).
  • One or more of the substitution mutations can be a substitution (e.g., a conservative amino acid substitution).
  • a biologically active variant of a Cas9 polypeptide can have an amino acid sequence with at least or about 50% sequence identity (e.g., at least or about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%), 97%), 98%), or 99% sequence identity) to a wild type Cas9 polypeptide.
  • Conservative amino acid substitutions typically include substitutions within the following groups: glycine and alanine; valine, isoleucine, and leucine; aspartic acid and glutamic acid; asparagine, glutamine, serine and threonine; lysine, histidine and arginine; and phenylalanine and tyrosine.
  • the Cas9 nuclease sequence can be a mutated sequence.
  • the Cas9 nuclease can be mutated in the conserved FiNH and RuvC domains, which are involved in strand specific cleavage.
  • an aspartate-to-alanine (D10A) mutation in the RuvC catalytic domain allows the Cas9 nickase mutant (Cas9n) to nick rather than cleave DNA to yield single-stranded breaks, and the subsequent preferential repair through HDR can potentially decrease the frequency of unwanted indel mutations from off-target double- stranded breaks.
  • polypeptides that are biologically active variants of a CRISPR- associated endonuclease can be characterized in terms of the extent to which their sequence is similar to or identical to the corresponding wild-type polypeptide.
  • sequence of a biologically active variant can be at least or about 80% identical to corresponding residues in the wild-type polypeptide.
  • a biologically active variant of a CRISPR-associated endonuclease can have an amino acid sequence with at least or about 80% sequence identity (e.g., at least or about 85%, 90%, 95%, 97%, 98%, or 99% sequence identity) to a CRISPR- associated endonuclease or to a homolog or ortholog thereof.
  • a biologically active variant of a CRIS PR-associated endonuclease polypeptide will retain sufficient biological activity to be useful in the present methods.
  • the biologically active variants will retain sufficient activity to function in targeted DNA cleavage.
  • the biological activity can be assessed in ways known to one of ordinary skill in the art and includes, without limitation, in vitro cleavage assays or functional assays.
  • the endonuclease is CRISPR-Cpfl which is the more recently characterized CRISPR from Provotella and Francisella 1 (Cpfl) in Zetsche et al. (“Cpfl is a Single RNA-guided Endonuclease of a Class 2 CRISPR-Cas System (2015); Cell; 163, 1-13).
  • the TLR7 antagonist is IRS954 developed by Dynavax (see for example Tomai et al., Drug Discovery Today, 2006) or 2’ -O-methyl modified RNAs as described in Robbins et al., Molecular Therapy, 2007.
  • the TLR7 antagonist is AT791 ⁇ 3-[4-(6-(3-
  • the TLR7 antagonist is hydroxychloroquine.
  • the TLR7 antagonist is IMO-8400 (Balak et al., IMO-8400, a toll-like receptor 7, 8, and 9 antagonist, demonstrates clinical activity in a phase 2a, randomized, placebo-controlled trial in patients with moderate-to-severe plaque psoriasis. Clin Immunol. 2017 Jan;l74:63-72).
  • the TLR7 antagonist of the invention is administered to the subject with a therapeutically effective amount.
  • administer refers to the act of injecting or otherwise physically delivering a substance as it exists outside the body (e.g., the TLR7 antagonist of the present invention) into the subject, such as by mucosal, intradermal, intravenous, subcutaneous, intramuscular delivery and/or any other method of physical delivery described herein or known in the art.
  • administration of the substance typically occurs after the onset of the disease or symptoms thereof.
  • administration of the substance typically occurs before the onset of the disease or symptoms thereof.
  • a “therapeutically effective amount” is meant a sufficient amount of TLR7 receptor antagonist to prevent for use in a method for the treatment of SS at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide employed; and like factors well known in the medical arts.
  • the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day.
  • the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject to be treated.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, typically from 1 mg to about 100 mg of the active ingredient.
  • An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
  • compositions according to the invention are formulated for parenteral, transdermal, oral, rectal, intrapulmonary, subcutaneous, sublingual, topical or intranasal administration.
  • Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms.
  • compositions according to the invention are formulated for oral administration.
  • compositions according to the invention are formulated for intravenous administration.
  • compositions according to the invention are formulated for ocular administration.
  • compositions according to the invention are formulated for parental administration.
  • the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected. These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
  • the active ingredient of the present invention i.e. the TLR7 antagonist
  • pharmaceutically acceptable excipients such as biodegradable polymers
  • sustained-release matrices such as biodegradable polymers
  • pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • the carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • the active ingredients of the invention can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports.
  • the TLR7 antagonist of the present invention is administered to the subject in combination with another active ingredient.
  • the TLR7 antagonist of the present invention is administered to the subject in combination with a standard treatment.
  • standard treatment of SS is selected from the group consisting of cyclosporine, lifitegrast, pilocarpine, cevimeline, nonsteroidal anti-inflammatory drugs, hydroxychloroquine or methotrexate.
  • FIGURES are a diagrammatic representation of FIGURES.
  • FIG. 1 Hematoxylin and eosin (H&E) staining of representative salivary gland sections from female WT, TLR8ko and TLR7/8ko mice at the age of 18 months.
  • H&E Hematoxylin and eosin
  • B Histological scoring of H&E salivary gland sections from female WT, TLR8ko or TLR7/8ko mice at the age of 8 and 11-14 months. Each point represents the value of one mouse.
  • FIG. 1 Increased B cells in TLR8ko salivary glands. Salivary glands and spleens of 16 months old TLR8ko and WT mice were analyzed for expression of CD45.2, CD3 and B220. Shown are percentage of T cells (CD3intB220-) and B cells (CD3-B220+) gated on CD45.2. Data are representative of two 3 independent experiments with 3-6 mice per genotype.
  • FIG. 3 Increased levels of anti-SSA/Ro, -SSB/La, -RNA and -DNA autoantibodies in TLR8ko versus WT sera.
  • FIG. 4 Increased expression of TLR7 and cytokines mRNA levels in salivary glands of TLR8ko mice.
  • Total RNA was extracted from the salivary glands of female 8 months WT (white bars) and TLR8ko (black bars) mice, and the expression of TLR7, CXCL13, CXCR5, TNF, LT-a, IL-6 and b-actin were assessed by Q-PCR.
  • Data are representative of 2- 3 independent experiments. *p ⁇ 0.05, **r ⁇ 0.01, ***p ⁇ 0.00l.
  • FIG. 5 Increased TLR7, CXCL13 and CXCR5 mRNA levels in salivary glands of SS patients. Total RNA was extracted from the salivary glands of age matched sicca controls (white circles) and SS patients (black circles).
  • A The expression of TLR7, CXCL13, CXCR5, TNF, LT-ot,and GAPDH were assessed by Q-PCR.
  • FIG. 6 Absence of salivary gland inflammation in male TLR8ko mice is accompanied by reduced salivary gland TLR7 and LT-a mRNA expression compared to female TLR8ko mice.
  • A Histological scoring of H&E salivary gland sections from female and male TLR8ko and WT mice at the age of 8 and 14 months. Each point represents the value of one mouse. **p ⁇ 0.01; ***p ⁇ 0.00l,
  • B Total RNA was extracted from the salivary glands of 14 months old female and male TLR8ko and WT mice and the expression of TLR7, LT-a and b-actin were assessed by Q-PCR.
  • TLR8ko and TLR7ko mice were generated with the automated VelociGene technology. Briefly, the TLR targeting constructs were generated using bacterial homologous recombination to delete 2.3 kb and 2,4 kb of the TLR7 and TLR8 coding sequences, respectively, while inserting a lacZ/neo reporter/selection cassette. For all studies female TLR8ko and TLR7ko mice backcrossed to the C57BL/6 background for more that 10 generations were used, and age-matched wild-type CD57BL/6 mice as controls.
  • MSG biopsies were obtained from 40 SS patients and 11 sicca controls (SC) at the Department of Pathophysiology of the School of Medicine at the University of Athens, Athens, Greece, as a routine part of the diagnostic evaluation for SS, after informed consent was taken. None of the SS patients were complicated by lymphoma. The biopsies were immediately frozen in -80oC. Focus score was determined for each MSG biopsy sample, as previously described (J Clin Pathol. 1968 Sep;2l(5):656-60). 20 out of 21 SS patients and all SC were females with mean age+SD: 59.13+16.60 and 53.25+8.00, respectively. The control group included individuals complaining of sicca symptoms, who did not fulfil the SS criteria ( ⁇ l foci/4 mm2).
  • Spleen and salivary glands isolated from 16 months old TLR8ko and WT mice were minced and digested using collagenase II and DNase I in RPMI, 2% FCS. Spleen and salivary glands cell suspensions were stained with antibodies to the following molecules: CD45.2; B220; IA/IE, CDl lc, SiglecH, CDl lb, Ly6G, F4/80, CD86, NK1.1, CD3, CD4, CD8, CD44, CD26L.
  • germinal centers and plasmablasts populations were assessed using antibodies to CD138, CD38, GL7 and CD19. Type specific cells were quantified and recorded by flow cytometry (FACSCanto IITM and LSRFortessaTM X-20, BD) and analysed with FlowJo 9.9.3 software (Tree Star).
  • mice were anaesthetized by intraperitoneal injection (i.p.) (0.01 mL/g of body weight) with a xylazine - ketamine solution (2.5 mL Rompun 2%, 10 mL Imalgene 500, in sterile saline to a 50 mL final volume).
  • Pilocarpine hydrochloride P6503 SIGMA (244,72 g/mol) was injected intraperitoneally (0.5 pg/g body weight) and saliva was collected for 20 min in microcentrifuge tubes on ice. The tubes were briefly centrifuged and the volume of saliva determined. Saliva was stored at -80°C for further use.
  • TLR7 To investigate the role of TLR7 in SS we performed histological examination of salivary glands in sex and aged matched female TLR8ko and wild-type (WT) mice. Histological scores of inflammatory lesions in the salivary glands of TLR8ko mice at the age of 8 , 11-14 or 18 months were obviously significant higher than those of WT mice (Fig. 1 A & B). The magnitude of immune cell infiltration was progressively increased as the mice aged (Fig. 1B). However, 8 and 11-14 months old female double TLR7/8ko mice did not show any sign of salivary inflammation (Fig. 1 A & B), suggesting that the absence of TLR7 has a protective role in the development of the spontaneous salivary inflammation seen in TLR8ko and old WT mice.
  • TLR7 signalling drives B cells into the dark zone of germinal centers, and may play a role in the development of the antigen activated antibody repertoire
  • anti-Ro/SSA and anti- La/SSB are relatively frequent in the course of autoimmune diseases, particularly in Sjogren’s syndrome and systemic lupus erythematosus (SLE).
  • SLE systemic lupus erythematosus
  • patients with SS can also produce atypical autoantibodies, including anti-DNA and anti-ribonucleoprotein (RNP), some being associated with other systemic autoimmune diseases.
  • RNP anti-DNA and anti-ribonucleoprotein
  • cytokines including TNF, interleukine-6 (IF-6), lymphotoxin-a (FTa), CXCF13, CXR5, in the salivary glands of SS patients, thus we evaluated the mRNA levels of cytokines in the salivary glands of female TFR8ko versus WT mice.
  • Quantitative real-time PCR (Q-PCR) analysis revealed the expression of TFR7 in WT murine salivary glands, where this expression was significantly higher in the absence of TLR8 (Fig. 4).
  • Q-PCR Quantitative real-time PCR
  • TNF, IF-6, FTa, CXCF13, CXR5 A significant increased expression of TNF, IF-6, FTa, CXCF13, CXR5 was detected in 16 months old TFR8ko versus WT mice. However, we did not see any differences regarding the expression of IFN-b and IF-b between WT and TFR8ko mice (data not shown).
  • the chemokine CXCF13 directs B-cell chemotaxis through its cognate receptor CXCR5 and is elevated in several autoimmune diseases.
  • the increased expression of CXCF13 and CXCR5 in TLR8ko salivary glands suggests that the target organ plays an essential role in the inflammatory process by recruiting B.
  • TLR8ko mice had significant increased salivary gland inflammation compared to their female WT controls, (Fig. 6A). Most importantly, 8 months old female TLR8ko mice had significant increased salivary gland inflammation compared to male TLR8ko mice, and this difference become even greater at 14 months (Fig. 6A).
  • TLR7 and LTa mRNA expression in the salivary glands of female and male mice Male TLR8ko mice showed dramatically ( ⁇ 2 times less) reduced TLR7 and LTa mRNA levels compared to female TLR8ko mice (Fig 6B). Thus, male TLR8ko mice do not develop salivary gland inflammation and as consequence SS, and this could be attributed to the decreased salivary gland TLR7 and LTa mRNA expression levels compared to female TLR8ko mice.
  • TLR8ko mice develop spontaneous lupus due to increased TLR7 function in DCs (Desnues B, Macedo AB, Roussel- Queval A, Bonnardel J, Henri S, Demaria O, Alexopoulou L.
  • TLR8 on dendritic cells and TLR9 on B cells restrain TLR7-mediated spontaneous autoimmunity in C57BL/6 mice.
  • TLR8 deficiency leads to autoimmunity in mice. J Clin Invest. 20l0;120 :3651-62). Our current data clearly show that female TLR8ko mice, also develop SS-like autoimmunity that is accompanied by increased TLR7 expression in the salivary glands, while female double TLR7/8ko mice are protected.
  • SS sexual dimorphism
  • female TLR8ko mice developed SS histopathology, while age matched male TLR8 mice were protected (Fig. 6A). While this female bias remains poorly understood, the X chromosome dose effect could be suspected to account for the disease, since the TLR7 gene is located on the X chromosome (both in humans and mice) and may govern gender differences in the development of systemic autoimmune diseases.
  • male TLR8 mice had importantly reduced salivary gland TLR7 mRNA expression compared to female TLR8 mice (Fig 6B).
  • TLR7 overexpression drive the development of SS pathology?
  • TLR8ko mice produced less saliva upon pilocarpine-induced salivation compared to their WT counterparts.
  • stimulation of mice by a TLR3 agonist caused a rapid and significant loss of salivary gland function, without initial lymphocytic foci (Deshmukh, U. et al. 2009. Activation of innate immune responses through Toll-like receptor 3 causes a rapid loss of salivary gland function. Journal of Oral Pathology & Medicine 38: 42- 47).
  • TLR-signalling may affect glandular dysfunction without previous lymphocytic infiltrates.
  • primary SS is an exocrine-tissue specific autoimmune disease and is not associated with other autoimmune disorders such as SLE, whereas secondary SS occurs in addition to other autoimmune disease. Therefore, SS in female TLR8ko mice can be considered as secondary.
  • TLR7/8ko mice are protected from the development of spontaneous inflammatory foci that we observed in older WT salivary glands, and that TLR7/8ko mice do not develop anti-Ro/SSA and anti-La/SSB autoantibodies, it is believed to speculate that TLR7 plays an important role also on the development of primary SS. This hypothesis is also in accordance with the human SS data that we presented, where in the patients with primary SS we also observed an increased salivary gland TLR7 expression that was highly correlated with inflammatory cytokine and chemokine expression.
  • TLR7 plays an important role in the development of SS in female TLR8ko mice, and the inflammatory signature - TLR7 overexpression that was highly correlated with certain cytokines and chemokines - that we observed in their salivary glands was quite similar with the one that we observed in the salivary glands of SS patients.
  • TLR7 seems to be a central player in SS pathology and that could be used as a novel target for the development of new approaches for the prevention and treatment of SS.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne des procédés et des compositions pour le traitement du syndrome de Sjögren (SS). Pour l'instant, aucune thérapie efficace n'est pas disponible et le traitement a été principalement symptomatique. Les inventeurs ont examiné l'implication de TLR7 dans l'apparition du SS à l'aide de modèles animaux TLR8ko qui développent spontanément un lupus érythémateux systémique (LES) en raison d'une signalisation TLR7 augmentée. Les inventeurs ont découvert que les souris TLR8ko développaient spontanément un SS. De manière intéressante, les souris doublement TLR7/8 ko ne présentaient aucun signe de SS. Ces données fournissent une preuve du rôle central de TLR7 dans l'apparition du SS et d'une inflammation des glandes salivaires.
PCT/EP2019/060662 2018-04-26 2019-04-25 Procédés et compositions destinés au traitement du syndrome de sjögren WO2019207066A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP18305523.5 2018-04-26
EP18305523 2018-04-26

Publications (1)

Publication Number Publication Date
WO2019207066A1 true WO2019207066A1 (fr) 2019-10-31

Family

ID=62165506

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2019/060662 WO2019207066A1 (fr) 2018-04-26 2019-04-25 Procédés et compositions destinés au traitement du syndrome de sjögren

Country Status (1)

Country Link
WO (1) WO2019207066A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023135505A1 (fr) * 2022-01-11 2023-07-20 Novartis Ag Antagoniste de tlr7/8 pour le traitement du syndrome de sjögren ou d'une maladie du tissu conjonctif mixte

Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
WO1990007861A1 (fr) 1988-12-28 1990-07-26 Protein Design Labs, Inc. IMMUNOGLOBULINES CHIMERIQUES SPECIFIQUES CONTRE LA PROTEINE TAC p55 DU RECEPTEUR D'IL-2
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
WO1999032619A1 (fr) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Inhibition genetique par de l'arn double brin
US5981732A (en) 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
WO2001036646A1 (fr) 1999-11-19 2001-05-25 Cancer Research Ventures Limited Inhibition d"expression genique a l"aide d"arn bicatenaire
WO2001068836A2 (fr) 2000-03-16 2001-09-20 Genetica, Inc. Procedes et compositions d'interference d'arn
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I expression
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
US6566135B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of caspase 6 expression
US6566131B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of Smad6 expression
US6573099B2 (en) 1998-03-20 2003-06-03 Benitec Australia, Ltd. Genetic constructs for delaying or repressing the expression of a target gene
WO2005007672A2 (fr) 2003-06-20 2005-01-27 Coley Pharmaceutical Gmbh Antagonistes des recepteurs toll (tlr) pour petites molecules
US20060269936A1 (en) 2005-03-24 2006-11-30 Jaromir Vlach Screening assay for TLR7, TLR8 and TLR9 agonists and antagonists
WO2008033432A2 (fr) * 2006-09-12 2008-03-20 Coley Pharmaceutical Group, Inc. Immunomodulation par des ribonucléosides et des oligoribonucléotides chimiquement modifiés

Patent Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
WO1990007861A1 (fr) 1988-12-28 1990-07-26 Protein Design Labs, Inc. IMMUNOGLOBULINES CHIMERIQUES SPECIFIQUES CONTRE LA PROTEINE TAC p55 DU RECEPTEUR D'IL-2
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5693761A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Polynucleotides encoding improved humanized immunoglobulins
US5693762A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
WO1999032619A1 (fr) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Inhibition genetique par de l'arn double brin
US6573099B2 (en) 1998-03-20 2003-06-03 Benitec Australia, Ltd. Genetic constructs for delaying or repressing the expression of a target gene
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
US5981732A (en) 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
WO2001036646A1 (fr) 1999-11-19 2001-05-25 Cancer Research Ventures Limited Inhibition d"expression genique a l"aide d"arn bicatenaire
WO2001068836A2 (fr) 2000-03-16 2001-09-20 Genetica, Inc. Procedes et compositions d'interference d'arn
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I expression
US6566135B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of caspase 6 expression
US6566131B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of Smad6 expression
WO2005007672A2 (fr) 2003-06-20 2005-01-27 Coley Pharmaceutical Gmbh Antagonistes des recepteurs toll (tlr) pour petites molecules
US20060269936A1 (en) 2005-03-24 2006-11-30 Jaromir Vlach Screening assay for TLR7, TLR8 and TLR9 agonists and antagonists
WO2008033432A2 (fr) * 2006-09-12 2008-03-20 Coley Pharmaceutical Group, Inc. Immunomodulation par des ribonucléosides et des oligoribonucléotides chimiquement modifiés

Non-Patent Citations (39)

* Cited by examiner, † Cited by third party
Title
"Genbank", Database accession no. GL669193757
"Genbank", Database accession no. GL669193761
"Genbank", Database accession no. GL669193765
"Genbank", Database accession no. KM099231.1
"Genbank", Database accession no. KM099232.1
"Genbank", Database accession no. KM099233.1
BALAK ET AL.: "IMO-8400, a toll-like receptor 7, 8, and 9 antagonist, demonstrates clinical activity in a phase 2a, randomized, placebo-controlled trial in patients with moderate-to-severe plaque psoriasis", CLIN IMMUNOL., vol. 174, January 2017 (2017-01-01), pages 63 - 72, XP029923566, DOI: doi:10.1016/j.clim.2016.09.015
BERGLOVA, I. ET AL.: "B cell toll-like receptors with respect to the pathogenesis of Sjogren's syndrome", ACTA MEDICA (HRADEC KRALOVE, vol. 54, 2011, pages 51 - 57, XP055487719
DEMARIA O; PAGNI PP; TRAUB S; DE GASSART A; BRANZK N; MURPHY AJ; VALENZUELA DM; YANCOPOULOS GD; FLAVELL RA; ALEXOPOULOU L: "TLR8 deficiency leads to autoimmunity in mice", J CLIN INVEST., vol. 120, 2010, pages 3651 - 62
DENG, Y. ET AL.: "MicroRNA-3148 modulates allelic expression of toll-like receptor 7 variant associated with systemic lupus erythematosus", PLOS GENET, vol. 9, 2013, pages el003336
DESHMUKH, U. ET AL.: "Activation of innate immune responses through Toll-like receptor 3 causes a rapid loss of salivary gland function", JOURNAL OF ORAL PATHOLOGY & MEDICINE, vol. 38, 2009, pages 42 - 47
DESNUES B; MACEDO AB; ROUSSEL-QUEVAL A; BONNARDEL J; HENRI S; DEMARIA O; ALEXOPOULOU L: "TLR8 on dendritic cells and TLR9 on B cells restrain TLR7-mediated spontaneous autoimmunity in C57BL/6 mice", PROC NATL ACAD SCI USA, vol. 111, 2014, pages 1497 - 502, XP055426735, DOI: doi:10.1073/pnas.1314121111
DICARLO ET AL., NUCLEIC ACIDS RES., vol. 41, 2013, pages 4336 - 4343
FABRE ET AL., PLOS NEGL. TROP. DIS., vol. 8, 2014, pages e2671
FOX, R. I. ET AL.: "Update in Sjogren's syndrome", CURRENT OPINION IN RHEUMATOLOGY, vol. 12, 2000, pages 391 - 398, XP009189412
GRATZ ET AL., GENETICS, 2014
GUO ET AL., DEVELOPMENT, vol. 141, 2014, pages 707 - 714
HAI ET AL., CELL RES., 2014
HWANG ET AL., PLOS ONE, vol. 8, 2013, pages e68708
IRENA BERGLOVÁ: "B CELL TOLL-LIKE RECEPTORS WITH RESPECT TO THE PATHOGENESIS OF SJOGREN'S SYNDROME", ACTA MEDICA (HRADEC KRÁLOVÉ), vol. 54, 1 January 2011 (2011-01-01), pages 51 - 57, XP055487719 *
J CLIN PATHOL., vol. 21, no. 5, September 1968 (1968-09-01), pages 656 - 60
KAWAKAMI, A.: "Toll-like receptor in salivary glands from patients with Sjogren's syndrome: functional analysis by human salivary gland cell line", THE JOURNAL OF RHEUMATOLOGY, vol. 34, 2007, pages 1019 - 1026
KOHLER; MILSTEIN, NATURE, vol. 256, 1975, pages 495
KOPP, E.; R. MEDZHITOV: "Recognition of microbial infection by Toll-like receptors", CURR OPIN IMMUNOL, vol. 15, 2003, pages 396 - 401, XP004444344, DOI: doi:10.1016/S0952-7915(03)00080-3
LAMPHIER M ET AL.: "Novel small molecule inhibitors of TLR7 and TLR9: mechanism of action and efficacy in vivo", MOL PHARMACOL., vol. 85, no. 3, March 2014 (2014-03-01), pages 429 - 40, XP009510804, DOI: doi:10.1124/mol.113.089821
MA ET AL., CELL RES., vol. 24, 2014, pages 122 - 125
MALI ET AL., SCIENCE, vol. 339, 2013, pages 823 - 826
MASHIKO ET AL., DEV. GROWTH DIFFER., vol. 56, 2014, pages 122 - 129
MAVRAGANI, C. P.; H. M. MOUTSOPOULOS: "Sjogren's syndrome", ANNUAL REVIEW OF PATHOLOGY, vol. 9, 2014, pages 273 - 285
MAVRAGANI, C. P.; H. M. MOUTSOPOULOS: "The geoepidemiology of Sjogren's syndrome", AUTOIMMUN REV, vol. 9, 2010, pages A305 - 310, XP026940090, DOI: doi:10.1016/j.autrev.2009.11.004
NIU ET AL., CELL, vol. 156, 2014, pages 836 - 843
OHYAMA, Y. ET AL.: "Severe focal sialadenitis and dacryoadenitis in NZM2328 mice induced by MCMV: a novel model for human Sjogren's syndrome", J IMMUNOL, vol. 177, 2006, pages 7391 - 7397
PILLEMER, S. R. ET AL.: "Incidence of physician-diagnosed primary Sjogren's syndrome in residents of Olmsted County, Minnesota", MAYO CLINIC PROCEEDINGS, vol. 76, 2001, pages 593 - 599
QIN B; WANG J; YANG Z ET AL.: "Epidemiology of primary Sjogren's syndrome: a systematic review and meta-analysis", ANN RHEUM DIS., vol. 74, 2015, pages 1983 - 9
ROBBINS ET AL., MOLECULAR THERAPY, 2007
TOMAI ET AL., DRUG DISCOVERY TODAY, 2006
VRECA MISA ET AL: "Expression ofTLR7,TLR9,JAK2, andSTAT3genes in peripheral blood mononuclear cells from patients with systemic sclerosis", JOURNAL OF APPLIED GENETICS: AN INTERNATIONAL JOURNAL OF GENETICS AND BREEDING, SPRINGER, GERMANY, vol. 59, no. 1, 16 November 2017 (2017-11-16), pages 59 - 66, XP036424657, ISSN: 1234-1983, [retrieved on 20171116], DOI: 10.1007/S13353-017-0415-4 *
YANG ET AL., J. MOL. CELL BIOL., vol. 6, 2014, pages 97 - 99
ZETSCHE ET AL.: "Cpfl is a Single RNA-guided Endonuclease of a Class 2 CRISPR-Cas System", CELL, vol. 163, 2015, pages 1 - 13

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023135505A1 (fr) * 2022-01-11 2023-07-20 Novartis Ag Antagoniste de tlr7/8 pour le traitement du syndrome de sjögren ou d'une maladie du tissu conjonctif mixte

Similar Documents

Publication Publication Date Title
JP2009521234A (ja) 補体系を制御する組成および方法
US20120064122A1 (en) Treatment of autoimmune inflammation using mir-155
US9926562B2 (en) Methods for preventing and treating chronic kidney disease (CKD)
US8067577B2 (en) Methods and compositions for the treatment of intestinal conditions
Matsuda et al. Suppression of Murine Experimental Autoimmune Optic Neuritis by Mature Dendritic Cells Transfected with Calcitonin Gene–Related Peptide Gene
US10059946B2 (en) Method for treatment of heart failure
KR20200136939A (ko) 천식 또는 알레르기성 질환을 치료하는 방법
WO2019207066A1 (fr) Procédés et compositions destinés au traitement du syndrome de sjögren
EP3268027B1 (fr) Antagonistes de l'hepcidine destinés à être utilisés dans le traitement d'une inflammation
WO2009033091A1 (fr) Traitement d'une maladie auto-immune médiée par th17 par l'intermédiaire de l'inhibition de stat3
WO2013121034A1 (fr) Procédés et compositions pharmaceutiques pour la réduction d'une inflammation du tissu adipeux
JP5470261B2 (ja) 大動脈瘤非ヒト動物モデルの作出方法
US20120283190A1 (en) Endothelin inhibitors for the treatment of rapidly progressive glomerulonephritis
US20130236480A1 (en) Transglutaminase 2 inhibitors for use in the prevention or treatment of rapidly progressive glomerulonephritis
WO2021048315A1 (fr) Utilisation d'inhibiteurs de duox1 pour le traitement du cancer
US20160304881A1 (en) Ddr1 antagonist or an inhibitor of ddr1 gene expression for use in the prevention or treatment of crescentic glomerulonephritis
ALRASHDI CONTRIBUTION OF THE VOLTAGE-GATED SODIUM CHANNEL NAV1. 6 AND TOLL LIKE RECEPTOR 2 IN THE PATHOPHYSIOLOGY OF EAE
WO2006056479A1 (fr) Inhibiteurs de sly1 en transplantation
WO2011112554A1 (fr) Nouvelles approches thérapeutiques pour le syndrome de birt-hogg-dube (bhd)

Legal Events

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

Ref document number: 19720117

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19720117

Country of ref document: EP

Kind code of ref document: A1