WO2015054210A2 - T. suis excretory/secretory products and methods of their use - Google Patents

T. suis excretory/secretory products and methods of their use Download PDF

Info

Publication number
WO2015054210A2
WO2015054210A2 PCT/US2014/059440 US2014059440W WO2015054210A2 WO 2015054210 A2 WO2015054210 A2 WO 2015054210A2 US 2014059440 W US2014059440 W US 2014059440W WO 2015054210 A2 WO2015054210 A2 WO 2015054210A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
subject
composition
acid sequence
polypeptides
Prior art date
Application number
PCT/US2014/059440
Other languages
French (fr)
Other versions
WO2015054210A3 (en
Inventor
Matthew Ross HEPWORTH
Susanne Hartmann
Friederike EBNER
Marion MULLER
Katharina JANEK
Original Assignee
Coronado Biosciences, Inc.
Freie Universitat Berlin
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 Coronado Biosciences, Inc., Freie Universitat Berlin filed Critical Coronado Biosciences, Inc.
Publication of WO2015054210A2 publication Critical patent/WO2015054210A2/en
Publication of WO2015054210A3 publication Critical patent/WO2015054210A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0003Invertebrate antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43536Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from worms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/577Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 tolerising response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons

Definitions

  • the present invention relates generally to helminthic excretory/secretory products. More particularly, the present invention relates to biologically active polypeptides secreted by Trichuris suis and to the use of such isolated polypeptides for modulating an immune response of a subject or in the treatment of certain medical disorders, including those that are Thl- and Th2-mediated medical disorders.
  • Thl- and Th2-mediated disorders such as allergy, inflammatory and autoimmune diseases in the western world has been correlated with the loss of helminth infections as a consequence of increased hygiene and reduced exposure (Maizels et al. J.Exp. Med. 2009; Maizels et al. Immun.Rev. 2004).
  • Treatment of patients in helminth- endemic countries with antiheminthic drugs has been shown to lead to enhanced allergic reactivity (Flohr et al. Clin. Exp. Allergy 2009; van Biggelaar et al. J. Inf. Dis. 2004) whereas, infection of allergic patients with helminths has been linked to improved health and reduced disease symptoms (Feary et al. Clin. Exp. Allergy 2010).
  • gastrointestinal helminths can be considered as a beneficial commensal macroflora, which act to induce immunoregulatory pathways necessary for immune homeostasis and the health of the host.
  • T. suis The helminth Trichuris suis (T. suis), for example, naturally infects the caecum and colon of pigs.
  • T. suis ova TSO
  • TSO administration to patients suffering from Crohn's disease and Ulcerative colitis resulted in a significant reduction in clinical symptoms of disease and the onset of remission in many patients (Summers et al. Gut 2005; Summers et al. Gastroenterology 2005), which suggests that TSO administration has therapeutic potential for the treatment of inflammatory human diseases in the clinic.
  • TSO therapy may also have immunomodulatory and therapeutic potential in patients suffering from multiple sclerosis (Fleming et al. Mult. Scler. 201 1; Benzel et al. J. Helminth. 2011), although larger clinical trials are desired to confirm the efficacy of this therapy.
  • the present invention relates to novel excretory/secretory polypeptides isolated and purified from a larval stage (LI) of T. suis, as well as recombinant versions of the same.
  • the polypeptides themselves are shown to demonstrate therapeutic and immunomodulatory properties in vivo and may be administered as a pharmaceutical composition, preferably in the absence of the helminth, its ova, or its larvae.
  • one aspect of the present invention provides one or more isolated helminthic excretory/secretory polypeptides, said one or more polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-4 and 37-45, or a biologically active or immunogenic fragment thereof.
  • the amino sequences of SEQ ID NOs: 1-3 and 37-45 include a secretion signal peptide sequence.
  • the mature form of the proteins (i.e., protein minus signal peptide) of SEQ ID NOs: 1-3 and 37-45 consist of the amino acid sequences shown in SEQ ID NOs: 5-7 and 46-54, respectively.
  • Another aspect of the present invention provides one or more isolated helminthic excretory/secretory polypeptides, said one or more polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 5-7 and 46-54 or a biologically active or immunogenic fragment thereof.
  • the one or more polypeptides are isolated from Trichuris suis.
  • Another aspect of the present invention provides one or more recombinant polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54, or a biologically active or immunogenic fragment thereof.
  • such recombinant polypeptides may be obtained by a process comprising the steps of culturing a cell transformed with a polynucleotide comprising a sequence that encodes an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54, under conditions suitable for polypeptide expression, and recovering the polypeptide so expressed.
  • the polynucleotide with which the cell is transformed comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 8-28 and 30-36.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of one or more recombinant polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54, or a biologically active or immunogenic fragment thereof; and a pharmaceutically acceptable carrier.
  • a composition comprising one or more isolated Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, said one or more Ts E/S polypeptides having a molecular weight ranging from about 20 kD and about 80 kD.
  • the one or more polypeptides are recombinant.
  • the one or more polypeptides may have an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54, or a biologically active or immunogenic fragment thereof.
  • the one or more polypeptides may have a molecular weight of about 58 kD, about 50 kD, about 45 kD, about 35 kD, or about 20 kD.
  • the polypeptides may be obtained from in vitro hatched T. suis first- stage (LI) larvae.
  • a method for modulating a Thl- or Th2-mediated immune response in a subject comprising administering to a subject whose Thl- or Th2-mediated immune response is in need of modulation an effective amount of one or more isolated polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54, a recombinant version thereof, or a biologically active or immunogenic fragment of the foregoing.
  • a method for modulating a Thl- or Th2-mediated immune response in a subject comprising administering to a subject whose Thl- or Th2 -mediated immune response is in need of modulation a composition comprising an effective amount of one or more polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54, a recombinant version thereof, or a biologically active or immunogenic fragment of the foregoing, wherein the composition is substantially or totally free of Ts eggs and larvae.
  • the present invention provides a method of modulating a Thl- or Th2-mediated immune response in a subject comprising administering to a subject whose Thl- or Th2-mediated immune response is in need of modulation a composition comprising Ts E/S supernatants but substantially or totally free of Ts eggs and larvae.
  • the present invention provides a method of modulating a Thl- or Th2-mediated immune response in a subject comprising contacting one or more T regulatory cells of a subject with a composition comprising Ts E/S supernatants but substantially or totally free of Ts eggs and larvae.
  • the present invention provides a method of regulating expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN- ⁇ in a subject comprising administering to a subject whose expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN- ⁇ are in need of regulation an effective amount of a composition comprising one or more isolated Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, each having a molecular weight ranging from about 20 kD and about 80 kD, and a carrier.
  • Ts Trichuris suis
  • E/S excretory/secretory
  • the present invention provides a method of regulating expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN- ⁇ in a subject comprising administering to a subject whose expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN- ⁇ are in need of regulation an effective amount of a composition comprising one or more Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, each having a molecular weight ranging from about 20 kD and about 80 kD, and a carrier, wherein the composition is substantially or totally free of Ts eggs and larvae.
  • Ts Trichuris suis
  • E/S excretory/secretory
  • the present invention provides a method of regulating expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN- ⁇ in a subject comprising administering to a subject whose expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN- ⁇ are in need of regulation a composition comprising Ts E/S supernatants but substantially or totally free of Ts eggs and larvae.
  • Still another aspect of the present invention provides a method of treating allergic airway hyperreactivity or inflammation in a subject, said method comprising administering to a subject in need thereof an effective amount of a composition comprising one or more isolated Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, each having a molecular weight ranging from about 20 kD and about 80 kD and a carrier.
  • Ts Trichuris suis
  • E/S excretory/secretory
  • Still another aspect of the present invention provides a method of treating allergic airway hyperreactivity or inflammation in a subject, said method comprising administering to a subject in need thereof an effective amount of a composition comprising one or more Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, each having a molecular weight ranging from about 20 kD and about 80 kD and a carrier, wherein the composition is substantially or totally free of Ts eggs and larvae.
  • Ts Trichuris suis
  • E/S excretory/secretory
  • Still another aspect of the present invention provides a method of treating allergic airway hyperreactivity or inflammation in a subject, said method comprising administering to a subject in need thereof a composition comprising Ts E/S supernatants but substantially or totally free of Ts eggs and larvae
  • the present invention provides a composition
  • a composition comprising a mixture of Trichuris suis (Ts) excretory/secretory (E/S) polypeptides obtained by culturing Ts first stage larvae in an aqueous medium, harvesting a portion of the medium at predetermined intervals while replenishing the medium over a period of about one to two weeks.
  • Ts Trichuris suis
  • E/S secretory
  • 50,000 larvae/well may be cultured on a 24-well plate using 0.5 mL of serum- free medium per well.
  • 0.4 mL of supernatant may be discarded after a period of 24 hours with the medium being replenished.
  • 0.4 mL of supernatant is subsequently harvested every 48 hours with the medium being replenished over a period of ten days. In yet further embodiments, 0.4 mL of supernatant may be subsequently harvested every 48 hours with the medium being replenished over a period of fourteen days.
  • Another aspect of the present invention provides a protein pellet obtained by acidifying the composition described above.
  • the protein pellet may, in some embodiments, be obtained by adding 100% trichloroacetic acid (TCA) at 1 :5 ratio of TCA volume to sample volume.
  • TCA trichloroacetic acid
  • a method for modulating an immune response of a subject comprising administering to a subject whose immune response is in need of modulation an effective amount of a composition comprising a mixture of isolated Ts E/S polypeptides.
  • splenocytes of a treated subject may exhibit suppressed IFN- ⁇ and IL-4 production compared to splenocytes of a control subject.
  • dendritic cells of a treated subject may exhibit no upregulation in their expression of CD86 and MHC class II molecules compared to dendritic cells of a control subject.
  • the dendritic cells of a treated subject may, in some embodiments, exhibit suppressed IL-6 production compared to dendritic cells of a control subject.
  • a method for modulating an immune response of a subject comprising administering to a subject whose immune response is in need of modulation an effective amount of a composition comprising a mixture of Ts E/S polypeptides, wherein the composition is substantially or totally free of Ts eggs and larvae.
  • Yet another aspect of the present invention provides a method of alleviating allergic airway hyperreactivity or inflammation comprising administering to a subject in need thereof an effective amount of a composition comprising a mixture of isolated Ts E/S polypeptides.
  • treated subjects exhibit reduced lymphocyte and eosinophil infiltration compared to a control subject.
  • treated subjects may exhibit suppressed IL-4, IL-5 and IL-13 production compared to a control subject.
  • Yet another aspect of the present invention provides a method of alleviating allergic airway hyperreactivity or inflammation comprising administering to a subject in need thereof an effective amount of a composition comprising a mixture of Ts E/S polypeptides, wherein the composition is substantially or totally free of Ts eggs and larvae.
  • polypeptides that are the subject of the present invention may comprise, or alternatively consist essentially of, or yet further consist of, a "biologically equivalent” or “biologically active” polypeptide encoded by equivalent polynucleotides as described herein.
  • polypeptides may possess at least 60%, or alternatively, at least 65%, or alternatively, at least 70%, or alternatively, at least 75%, or alternatively, at least 80%, or alternatively at least 85%, or alternatively at least 90%, or alternatively at least 95% or alternatively at least 98%, identical primary amino acid sequence to the reference polypeptide (i.e., a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54 or a fragment thereof) when compared using sequence identity methods run under default conditions.
  • reference polypeptide i.e., a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54 or a fragment thereof
  • sequence identity methods run under default conditions.
  • biologically active is used to refer to a protein or polypeptide that induces a desired beneficial effect when orally administered to a subject and includes agents that are therapeutically and/or prophylactically effective as pharmaceuticals ("pharmacologically active agents").
  • pharmacologically active agents agents that are therapeutically and/or prophylactically effective as pharmaceuticals.
  • an “effective" amount of an active agent is meant a nontoxic but sufficient amount of an active agent to provide the desired beneficial effect.
  • isolated peptide fragment is meant to include peptide fragments which are not naturally occurring as fragments and would not be found in the natural state.
  • isolated is also used herein to refer to polypeptides and proteins that are isolated from other cellular proteins and is meant to encompass both purified and recombinant polypeptides.
  • isolated means separated from constituents, cellular and otherwise, in which the cell, tissue, polynucleotide, peptide, polypeptide, protein, antibody or fragment(s) thereof, which are normally associated in nature.
  • an isolated cell is a cell that is separated form tissue or cells of dissimilar phenotype or genotype.
  • a non-naturally occurring polynucleotide, peptide, polypeptide, protein, antibody or fragment(s) thereof does not require “isolation” to distinguish it from its naturally occurring counterpart.
  • polypeptide is used interchangeably with the term “protein” and in its broadest sense refers to a compound of two or more subunit amino acids, amino acid analogs or peptidomimetics.
  • the subunits may be linked by peptide bonds. In another embodiment, the subunit may be linked by other bonds, e.g., ester, ether, etc.
  • amino acid refers to natural and/or unnatural or synthetic amino acids, including glycine and both the D and L optical isomers, amino acid analogs and peptidomimetics.
  • a peptide of three or more amino acids is commonly called an oligopeptide if the peptide chain is short.
  • peptide chain is commonly called a polypeptide or a protein.
  • peptide fragment also refers to a peptide chain.
  • Homology or “identity” or “similarity” are synonymously and refers to sequence similarity between two peptides or between two nucleic acid molecules. Homology can be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same base or amino acid, then the molecules are homologous at that position. A degree of homology between sequences is a function of the number of matching or homologous positions shared by the sequences. An "unrelated” or “non-homologous” sequence shares less than 40% identity, or alternatively less than 25% identity, with one of the sequences of the present invention.
  • culture refers to the in vitro propagation of cells or organisms on or in media of various kinds. It is understood that the descendants of a cell grown in culture may not be completely identical (i.e., morphologically, genetically, or phenotypically) to the parent cell.
  • composition is intended to mean a combination of active polypeptides and another compound or composition, inert (e.g. a detectable label) or active (e.g. a delivery vehicle) alone or in combination with a carrier which can in one embodiment be a simple carrier like saline or pharmaceutically acceptable or a solid support as defined below.
  • inert e.g. a detectable label
  • active e.g. a delivery vehicle
  • carrier which can in one embodiment be a simple carrier like saline or pharmaceutically acceptable or a solid support as defined below.
  • a "pharmaceutical composition” is intended to include the combination of an active polypeptide, polynucleotide or antibody with a carrier, inert or active such as a solid support, making the composition suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo.
  • the term "pharmaceutically acceptable carrier” encompasses any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents.
  • the compositions also can include stabilizers and preservatives.
  • stabilizers and adjuvants see Martin (1975) Remington's Pharm. Sci., 15th Ed. (Mack Publ. Co., Easton).
  • a "subject,” “individual,” or “patient” is used interchangeably herein, and refers to a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murines, rats, rabbits, simians, bovines, ovines, porcines, canines, felines, farm animals, sport animals, pets, equines, and primates, particularly humans.
  • the terms "disease” and “disorder” are used inclusively and refer to any condition associated with improper function of the immune system of a subject.
  • egg refers to either embryonated or nonembryonated, viable T. suis ova.
  • Applicants have provided herein the polypeptide and/or polynucleotide sequences for use in gene and protein transfer and expression techniques described below. It should be understood, although not always explicitly stated that the sequences provided herein can be used to provide the expression product as well as substantially identical sequences that produce a protein that has the same biological properties. These "biologically equivalent” or “biologically active” polypeptides are encoded by equivalent polynucleotides.
  • They may possess at least 60%, or alternatively, at least 65%, or alternatively, at least 70%, or alternatively, at least 75%, or alternatively, at least 80%, or alternatively at least 85%, or alternatively at least 90%, or alternatively at least 95% or alternatively at least 98%, identical primary amino acid sequence to the reference polypeptide when compared using sequence identity methods run under default conditions.
  • Specific polypeptide sequences are provided as examples of particular embodiments. Modifications to the sequences to amino acids with alternate amino acids that have similar charge.
  • FIG. 1A depicts T. suis eggs (TSO), first stage larvae (LI) and the timeline of experimentation in which the eggs were hatched, washed thoroughly and cultured in vitro for 10-14 days during which time supernatants were harvested every two days (arrows).
  • Figure IB is a photograph showing electrophoresis results characterizing T. suis first stage larval E/S proteins.
  • Figure 2 is a series of graphs showing the suppression of antigen-specific CD4+ T cell cytokine production by Ts E/S, as well as modulation of dendritic cell activation in vitro.
  • I A and B show the proliferation of ovalbumin-specific CD4+ T cells treated with decreasing volumes of Ts E/S supematants for 72 hours and (A) incubated with 3H thymidine for a further 20 hours to assess proliferation, (B) and supematants analyzed via ELISA for production of IFN- ⁇ or IL-4. C+D.
  • Bone marrow-derived dendritic cells were pre- treated with Ts E/S supernatant prior to pulsing with (C) the TLR-9 agonist CpG or (D) the TLR-4 agonist LPS and DC activation was assessed via quantification of cells expressing the co-stimulatory markers CD86 and MHCII via flow cytometry and by production of the proinflammatory cytokines IL-6 and IL-12/23p40 by ELISA.
  • Figure 3(A)-(F) is a series of graphs and photographs showing the suppression of the development of allergic airway inflammation in mice that were sensitized by intraperitoneal injection of PBS or OVA.
  • A is a line graph showing airway hyperreactivity in response to increasing concentrations of metacholine;
  • B is a series of cytology results showing cellular infiltrate into bronchiolar lavage fluid;
  • C is a series of bar graphs showing total cytokine concentrations in BAL;
  • (D) is a series of bar graphs showing ELISA results of total cytokine concentrations in splenocytes following OVA restimulation;
  • E is a series of photographs and of histological staining (RELM-a-specific antibody staining in the lung parenchyma;
  • F is a series of graphs showing concentration of ova-specific cytokine isotopes in serum of PBS, OVA, and Ts E/S mice.
  • Figure 4 is two sets of graphs showing that Ts E/S treatment induces a T. suis- specific Th2 response and antibody production.
  • Figure 2(A) is a series of six bar graphs showing Ts E/S-specific cytokine production assessment via ELISA in culture supematants of splenocytes following restimulation with Ts E/S ex vivo for 24 hours.
  • Figure 2(B) is a series of three plots showing Ts E/S-specific antibody titres in the serum.
  • FIG. 5 shows that Ts E/S amelioration of allergic airway disease is not associated with increased Treg frequencies but is partially dependent on IL-10 production.
  • A is a quantification of FoxP3 expression in splenocytes in vitro stimulated with ConA +/- Ts E/S or Hp E/S or heat inactivated E/S proteins.
  • B is a quantification of CD25+ FoxP3+ regulatory T cells as a frequency of the CD4+ T cell compartment in the spleen and peribroncheolar lymph nodes (PBLN) by flow cytometry
  • PBLN peribroncheolar lymph nodes
  • C is a quantification of FoxP3+ cells in the lung parenchyma by immunohistochemistry staining.
  • (D) is an experiment schematic for antibody neutralization of IL-10R in OVA-induced allergic airway hyperreactive mice.
  • E is a quantification and differentiation of cell infiltrate in the BAL.
  • F is a quantification of IL- 5 and IL-13 in BAL fluid via ELISA.
  • G is a quantification of OVA-specific antibody titers in the serum of OVA-induced allergic airway hyperreactive mice +/- alL-lOR treatment.
  • FIG. 6 shows PCR amplification of SEQ ID NO: 1.
  • Figure 7 shows secretion of recombinant polypeptide SEQ ID NO: 1.
  • Figure 8 shows purification of recombinant polypeptide SEQ ID NO: 1.
  • Coomassie stained SDS-PAGE of His-tagged polypeptide SEQ ID NO: 1 HPLC purification showing representative aliquots of wash (Lane 2, Ctrl) and elution (Lane 3, HisTrap Elution) fractions.
  • FIG. 9 shows that SEQ ID NO: 1 suppresses antigen-specific proliferation of T cells co-cultured with Ova pulsed DCs.
  • Purified CD4+ T cells from Ova-tg D01 1.10 mice were labeled with CFSE cell proliferation dye and co-cultured with Ova-peptide pulsed DCs in the absence (+ Ctrl.) or presence of recombinant SEQ ID NO: 1 or heat-inactivated SEQ ID NO: 1.
  • Figure 10 shows that recombinant SEQ ID NO: 1 increases IL-10 secretion in CD4+ T cell - DC co-cultures.
  • SEQ ID NO: 1 is the full length amino acid sequence of a T. suis Locus_7131_l/l protein.
  • SEQ ID NO: 2 is the full length amino acid sequence of a T. suis Locus_1063_l/1 T. suis protein.
  • SEQ ID NO: 3 is the full length amino acid sequence of a T. suis Locus_1815_l/l protein.
  • SEQ ID NO: 4 is the amino acid sequence of a T. suis protein homolog of T. muris accession number TMC00067 1.
  • SEQ ID NO: 5 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 1 (i.e., SEQ ID NO: 1 minus the signal peptide sequence).
  • SEQ ID NO: 6 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 2 (i.e., SEQ ID NO: 2 minus the signal peptide sequence).
  • SEQ ID NO: 7 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 3 (i.e., SEQ ID NO: 3 minus the signal peptide sequence).
  • SEQ ID Nos: 8-10 and 30 are exemplary cDNA sequences encoding the full length protein of SEQ ID NO: 1.
  • SEQ ID Nos: 11-13 and 31 are exemplary cDNA sequences encoding the full length protein of SEQ ID NO: 2.
  • SEQ ID Nos: 14-16 and 32 are exemplary cDNA sequences encoding the full length protein of SEQ ID NO: 3.
  • SEQ ID Nos: 17-19 and 33 are exemplary cDNA sequences encoding the protein of SEQ ID NO: 4.
  • SEQ ID Nos: 20-22 and 34 are exemplary cDNA sequences encoding the mature protein of SEQ ID NO: 5.
  • SEQ ID Nos: 23-25 and 35 are exemplary cDNA sequences encoding the mature protein of SEQ ID NO: 6.
  • SEQ ID Nos: 26-28 and 36 are exemplary cDNA sequences encoding the mature protein of SEQ ID NO: 7.
  • SEQ ID NO: 29 is the amino acid sequence of a Trichuris muris protein.
  • SEQ ID NO: 37 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide.
  • SEQ ID NO: 46 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 37.
  • SEQ ID NO: 38 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide.
  • SEQ ID NO: 47 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 38.
  • SEQ ID NO: 39 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide.
  • SEQ ID NO: 48 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 39.
  • SEQ ID NO: 40 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide.
  • SEQ ID NO: 49 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 40.
  • SEQ ID NO: 41 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide.
  • SEQ ID NO: 50 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 41.
  • SEQ ID NO: 42 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide.
  • SEQ ID NO: 51 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 42.
  • SEQ ID NO: 43 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide.
  • SEQ ID NO: 52 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 43.
  • SEQ ID NO: 44 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide.
  • SEQ ID NO: 53 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 44.
  • SEQ ID NO: 45 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide.
  • SEQ ID NO: 54 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 45.
  • Ts E/S E/S products derived from the larval stage of T. suis
  • Ts E/S T. suis
  • Administration of soluble products derived from helminths is advantageous as it negates the need to expose patients to live parasites.
  • Ts E/S excretory/secretory supernatants
  • T. suis proteins Proteomic analysis of Ts E/S identified 4 T. suis proteins by LC-MS/MS spectrometry and subsequent database searches. Three of the identified proteins are specific for T. suis and could only be found in the T. suis transcriptome data base (www.helmdb.org) in contrast to the available Trichuris EST database "NEMBASE4" (www.nematodes,org) comprising of data from Trichuris muris, Trichineiia spiralis and Trichuris vulpis or the NCBI database. HelmDB comprises 1288 protein sequences translated from transcriptomic data of T. suis.
  • Polypeptides comprising, or alternatively consisting essentially of, or yet further consisting of, the amino acid sequences of the invention (SEQ ID NOs: 1-7 and 37-54 or a fragment or variant thereof) can be prepared by expressing polynucleotides encoding the polypeptide sequences of this invention in an appropriate host cell. This can be accomplished by methods of recombinant DNA technology known to those skilled in the art, employing, for example, a cDNA having a sequence selected from the group consisting of SEQ ID NOs: 8- 28 and 30-36.
  • this invention also provides methods for recombinantly producing the polypeptides of this invention in a eukaryotic or prokaryotic host cell, which in one aspect is further isolated from the host cell.
  • the amino acid sequences of SEQ ID NOs: 1-3 and 37-45 are full length proteins, i.e., they include a secretion signal peptide. Accordingly, the nucleotide sequences encoding SEQ ID NOs: 1-3 and 37-45 include a portion that encodes the secretion signal peptide.
  • the protein of any of SEQ ID NOs: 1-3 and 37-45 minus the secretion signal peptide is referred to as the mature protein.
  • the mature proteins of the invention comprise or consist of an amino acid sequence selected from the group consisting of SEQ ID NO: 5 (the mature protein of SEQ ID NO: 1), SEQ ID NO: 6 (the mature protein of SEQ ID NO: 2), SEQ ID NO: 7 (the mature protein of SEQ ID NO: 3), SEQ ID NO: 46 (the mature protein of SEQ ID NO: 37), SEQ ID NO: 47 (the mature protein of SEQ ID NO: 38), SEQ ID NO: 48 (the mature protein of SEQ ID NO: 39), SEQ ID NO: 49 (the mature protein of SEQ ID NO: 40), SEQ ID NO: 50 (the mature protein of SEQ ID NO: 41), SEQ ID NO: 51 (the mature protein of SEQ ID NO: 42), SEQ ID NO: 52 (the mature protein of SEQ ID NO: 43), SEQ ID NO: 53 (the mature protein of SEQ ID NO: 44), and SEQ ID NO: 54 (the mature protein of SEQ ID NO: 45).
  • SEQ ID NO: 5 the mature protein of SEQ ID NO
  • a polynucleotide sequence encoding a mature protein having a sequence selected from SEQ ID NOs: 5-7 and 46-54 may be linked to a polynucleotide sequence encoding a particular secretion signal peptide to produce a full length nucleotide sequence for expression/secretion in a particular cell type.
  • the proteins and peptide fragments of this invention also can be obtained by chemical synthesis using a commercially available automated peptide synthesizer such as those manufactured by Perkin Elmer/Applied Biosystems, Inc., Model 430A or 431A, Foster City, CA, USA.
  • the synthesized protein or polypeptide can be precipitated and further purified, for example by high performance liquid chromatography (HPLC).
  • HPLC high performance liquid chromatography
  • this invention also provides a process for chemically synthesizing the proteins of this invention by providing the sequence of the protein and reagents, such as amino acids and enzymes and linking together the amino acids in the proper orientation and linear sequence.
  • compositions for Therapy are Compositions for Therapy
  • compositions can be further combined with a carrier, a pharmaceutically acceptable carrier or medical device which is suitable for use of the compositions in diagnostic or therapeutic methods.
  • the compositions comprise, or alternatively consist essentially of, or yet further consists of, one or more of the above compositions described above in combination with a carrier, a pharmaceutically acceptable carrier or medical device.
  • the carrier can be a liquid phase carrier or a solid phase carrier, e.g., bead, gel, microarray, or carrier molecule such as a liposome.
  • the composition can optionally further comprise at least one further compound, protein or composition.
  • carriers includes therapeutically active agents such as another peptide or protein (e.g., an Fab' fragment).
  • an antibody of this invention, derivative or fragment thereof can be functionally linked (e.g., by chemical coupling, genetic fusion, noncovalent association, or otherwise) to one or more other molecular entities, such as another antibody (e.g., to produce a bispecific or a multispecific antibody), a cytotoxin, a cellular ligand, or an antigen.
  • this invention encompasses a large variety of antibody conjugates, bi- and multispecific molecules, and fusion proteins, whether or not they target the same epitope as the antibodies of this invention.
  • organic molecules also termed modifying agents or activating agents, that can be covalently attached, directly or indirectly, to an antibody of this invention. Attachment of the molecule can improve pharmacokinetic properties (e.g., increased in vivo serum half-life).
  • organic molecules include, but are not limited to a hydrophilic polymeric group, a fatty acid group or a fatty acid ester group.
  • fatty acid encompasses mono-carboxylic acids and di- carboxylic acids.
  • the polypeptides of this invention modulate a Thl- or Th2-mediated immune response in a subject and regulate expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN- ⁇ .
  • the Thl- or Th2-mediated response is allergic airway hyperreactivity or inflammation.
  • methods to achieve such in vitro or in vivo responses are provided by contacting or administering an effective amount of the polypeptide of this invention to a subject in need of such treatment. Administration can be by any suitable method and effective amounts can be empirically determined by a treating physician.
  • An "effective amount" is an amount sufficient to effect beneficial or desired results. An effective amount can be administered in one or more administrations, applications or dosages.
  • an "effective amount” corresponds to an amount of polypeptide administered to prevent or reduce an inflammatory response. In some embodiments, an "effective amount” corresponds to the amount of polypeptide given in a pre-challenge.
  • the "therapeutically effective amount” will vary depending on the polypeptide, the Thl- or Th2-mediated disorder and its severity and the age, weight, etc., of the patient to be treated all of which is within the skill of the attending clinician. It is contemplated that a therapeutically effective amount of a polypeptide described herein will decrease levels of inflammation in the patient as compared to the levels of inflammation in the absence of treatment. A therapeutically effective amount is distinguishable from an amount having a biological effect (a "biologically effective amount"). A biological effect, however, may not result in any clinically measurable therapeutically effect as described above as determined by methods within the skill of the attending clinician.
  • Administration in vivo can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell, solid tumor or cancer being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents can be found below.
  • the pharmaceutical compositions can be administered orally, intranasally, parenterally, injection, orally and may take the form of tablets, lozenges, granules, capsules, pills, ampoules, suppositories or aerosol form.
  • compositions may also take the form of suspensions, solutions and emulsions of the active ingredient in aqueous or nonaqueous diluents, syrups, granulates or powders.
  • the pharmaceutical compositions can also contain other pharmaceutically active compounds or a plurality of compounds of the invention.
  • an agent of the present invention also referred to herein as the active ingredient, may be administered for therapy by any suitable route including oral, rectal, nasal, topical (including transdermal, aerosol, buccal and sublingual), vaginal, intraperitoneal, parenteral (including subcutaneous, intramuscular, intravenous and intradermal) and pulmonary. It will also be appreciated that the preferred route will vary with the condition and age of the recipient, and the disease being treated.
  • the agent should be administered to achieve peak concentrations of the active compound at sites of disease. This may be achieved, for example, by the intravenous injection of the agent, optionally in saline, or orally administered, for example, as a tablet, capsule or syrup containing the active ingredient. Desirable blood levels of the agent may be maintained by a continuous infusion to provide a therapeutic amount of the active ingredient within disease tissue.
  • operative combinations is contemplated to provide therapeutic combinations requiring a lower total dosage of each component agent than may be required when each individual therapeutic compound or drug is used alone, thereby reducing adverse effects.
  • the agent While it is possible for the agent to be administered alone, it is preferable to present it as a pharmaceutical formulation comprising at least one active ingredient, as defined above, together with one or more pharmaceutically acceptable carriers therefor and optionally other therapeutic agents.
  • Each carrier must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets, each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder (e.g., povidone, gelatin, hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (e.g., sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose) surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
  • Formulations suitable for parenteral administration include aqueous and nonaqueous isotonic sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents, and liposomes or other microparticulate systems which are designed to target the compound to blood components or one or more organs.
  • the formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • sterile liquid carrier for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example, those suitable for oral administration may include such further agents as sweeteners, thickeners and flavoring agents. It also is intended that the agents, compositions and methods of this invention be combined with other suitable compositions and therapies.
  • T. suis secretome In order to demonstrate the efficacy of helminthic secretory proteins in combating Thl- and Th2-mediated disorders, an exemplary T. suis secretome is first obtained. Ova of Trichuris suis, were incubated, hatched into larvae, maintained as larvae, and induced to secrete products, which were then collected, as described below.
  • Ts E/S Trichuris suis Excretory/Secretory supernatants
  • T. suis ova 250,000 purified T. suis ova (Ovamed GmbH) were resuspended in 8 ml PBS mixed with 12 ml 6% Hypochloride solution and incubated for 45 minutes at 37°C, 5% CC ⁇ with periodical agitation. T. suis ova were washed via multiple rounds of centrifugation at 2000 RPM (425 x g) for 10 minutes and resuspended in serum free RPMI-1640 media containing 20 mM L-glutamine, 100 U/ml penicillin and 100 ⁇ g/ml streptomycin (Invitrogen). Hatched T.
  • protein pellet was used for LC-MS/MS spectrometry to identify the protein content.
  • supernatants were concentrated 20x with centrifugal concentrators with a molecular weight cut off of 5000 PES (Vivaspin 6; Viva products GmbH) and used as indicated below. Final Endotoxin levels were detected by a Limulus amoebocyte lysate test (Cambrex) and consistently found to be ⁇ 0.2 pg/ ⁇ . E/S containing supernatants were then concentrated/precipitated and the concentrated E/S was analysed via silver staining (Fig IB). LI T. suis E/S (Ts E/S) contained at least 7 distinct protein bands.
  • TCA-precipitated dried protein pellets of Ts E/S were suspended in 25 ⁇ of 0.1M ammonium bicarbonate buffer.
  • 4 ⁇ dithiotreitol 45 mM in 0.1 M ammonium bicarbonate
  • the mixture was incubated at 56°C for 45 min.
  • 4 ⁇ iodoacetamide solution 100 mM in 0.1 M ammonium biocarbonate
  • the sample was kept in the dark for 30 min. Then the solution was diluted with 95 ⁇ of 0.1 M ammonium bicarbonate.
  • Trypsin was dissolved in 50 mM acedic acid und diluted to a final concentration of 0.1 ⁇ g/ ⁇ l and kept at -80°C until use. In-solution digests were performed with 0.5 or ⁇ ⁇ of trypsin (0.1 ⁇ g/ ⁇ L) for 16-18 h at 37°C. The reaction was stopped with 2.5 ⁇ of 20% (v/v) trifluoroacetic acid (TFA) in water and samples were dried in a vacuum centrifuge.
  • TFA trifluoroacetic acid
  • LC-MS/MS analyses were performed on a 4700 proteomics Analyzer (ABSCIEX, Framingham, MS) off-line coupled with a Thermo Scientific Dionex UltiMate 3000 RSLC system and Probot fractionation device (Thermo Scientific, Idstein, Germany). Dried peptides were reconstituted in 12 ⁇ of 0.1% (v/v) TFA in water. Ten ⁇ of the sample were concentrated on a trap column (Acclaim-PepMap CI 8, 3 ⁇ , 2 cm x 75 ⁇ i.d. Thermo Scientific).
  • LC separations were performed on an analytical column (Acclaim-PepMap CI 8, 2 ⁇ , 150 mm x 75 ⁇ , Thermo Scientific) at a flow rate of 300 nl/min.
  • Mobile phase (A) was 0.1% (v/v) TFA in water and (B) was 80:20 (v/v) acetonitrile/water containing 0.08% (v/v) TFA.
  • the gradient was 3-15% B in 2 min and 15-60% B in 60 min.
  • MS mass spectra
  • T. suis LI E/S compared the observed MS spectra to databases containing known nematode proteins ( CBI, NemBase4, and HelmDB).
  • CBI known nematode proteins
  • HelmDB known nematode proteins
  • the second column is the respective database specific "accession ID,” followed by internal “description and species.”
  • the software calculates an overall protein score (Mascot score).
  • the column “Mascot score” reflects the combined scores of all observed mass spectra that can be matched to amino acid sequences within that protein. The higher the Mascot score, the more confident is the match.
  • the column “Peptide overage” indicates the percent of protein sequence information that is covered by the sum of all matched peptides.
  • the next two columns list peptide mass specific information, by showing the observed peptide mass obtained from the spectra (Peptide mass exp) and the calculated mass (Peptide mass calc) resulting from theoretical tryptic digests of the database sequence information.
  • the "peptide score” is a measure of how well the observed MS/MS spectra match to the stated peptide.
  • the following "Identity score” is a probability score that comprises the peptide length, the trypsin target sites and the possibility of incomplete proteolytic cleavage. Notably, the peptide score should be higher than the identity score to significantly identify a protein.
  • the "p- value” indicates the probability that the observed match between spectra and peptide sequence has been found by chance. In the last column the matching peptide "sequences" are listed. [0071] The tryptic digested LI E/S was analyzed by LC-MS/MS spectrometry as described above.
  • Locus_7131_l/l sequence identified no homology to any known sequences and no gene ontologies.
  • Three individual peptides matched the protein sequence of Locus_1063_l/1 (T. suis transcriptome, HelmDB, 26.5% sequence coverage) with a Mascot Score of 128. This locus describes a protein with an estimated mass of 15737 Da.
  • Locus_1815_l/l (40146 Da) was identified with a Mascot score of 83 by two different peptides covering 6% sequence information (T. suis transcriptome, HelmDB).
  • Further database search predicted the gene ontology terms: serine-type endopeptidase activity; proteolysis.
  • C carb carbadidomethylated cysteine
  • pyr Q pyroglutamat
  • MS and MS/MS peaklists were generated by the "Peak to Mascot" tool of the 4000er Series Expolerer v 3.6.
  • MASCOT server version 2.2, Matrixscience, London, UK
  • Data base searches were performed using NCBI (31.601.460 protein sequences), Helm database (1288 protein sequences translated from transcriptome data of T. suis, (www.helmdb.org), and Trichuris EST database (in-house database composed of 12.895 cDNA sequences from Trichuris muris, Trichinella spiralis und Trichuris vulpis, made from www.nematodes.org/nembase4/overview.shtml).
  • Protein identification revealed a number of protein matches based on NemBase4, HelmDB and NCBI.
  • Table 2 lists all proteins centered information according to an internal ranking (first column).
  • Table 2 lists the expected protein weight in Da, which is calculated based on the available sequence information.
  • the last column illustrates the complete protein sequence obtained from the indicated database. The respective peptides matching to each protein are underlined.
  • T. suis LI (lmg) will be ground in liquid nitrogen before being homogenized with the QIAshredder column (Quiagen). RNA will be isolated from the lysate using RNeasy Mini Kit (Quiagen) according to manufacturer's instruction. Integrity of the total RNA will be proofed by gel electrophoresis in 1% agarose, which have to yield two bands corresponding to 28 S and 18 S ribosomal RNA showing no smear confirming the integrity of the total RNA.
  • PCR amplified cDNA of the respective proteins will be cloned into pGEM-T Easy vector (Promega). Subsequently, for expression in E. coli the cDNA will be subcloned into pET 28 (Merck Millipore) with a 6-His tag at the N- and C-terminal end of the sequence. Alternatively, pQE 30 (Quiagen) will be used for only N-terminal 6-His tag expression, depending on keeping or eliminating the stop codon within the cloning strategy. Recombinant proteins will be purified by affinity chromatography using a Ni-NTA column and dialysed against PBS. Removal of endotoxin contaminations will be done by EndoTrap columns (Profos) and final endotoxin concentrations will be detected by Limulus amoebocyte lysate test (Cambrex).
  • Profiles EndoTrap columns
  • the first larval stages (LI) of Trichuris suis is likely to be the most clinical relevant stage of the life cycle in the context of therapeutic application in humans as it has been shown that the development to sexually mature adult stages is absent in TSO treated humans and efficacious treatment requires repeat applications of high doses of embryonated TSO suggesting that helminth establishment is transient in the human host.
  • the proteins of excretory/secretory (E/S) products derived from the LI T. suis in culture supernatants were determined. T. suis eggs were hatched, washed thoroughly and cultured in vitro for 10-14 days and supernatants were harvested every two days (Figure 1A).
  • IFN- ⁇ , IL-4, IL-5, IL-6, IL-10, IL-12/IL-23p40, IL-13 and IL-17A were determined using Cytometric Bead Assay (CBA, BD Biosciences) as per manufacturer's instructions or commercially available ELISA antibody pairs (BD Biosciences, eBioscience) following manufacturer's instructions.
  • CBA Cytometric Bead Assay
  • ELISA plates were coated with either 50 ⁇ g/ml OVA or 50 ⁇ Ts E/S in bicarbonate buffer at 4°C overnight along with serial dilutions of recombinant IgM, IgG, or IgE (SantCruz Biotech).
  • Plates were blocked with 3% BSA for one hour, washed in PBS 0.05% Tween and incubated for 2 hours with murine serum at multiple dilutions. Plate-bound antibody was then detected by probing with HRP-conjugated anti-mouse IgM, IgGl, or IgE secondary antibody and developed with TMB substrate (R&D Systems).
  • Example 3 71 suis Excretory/Secretory Proteins Have Immunomodulatory Properties in vivo
  • Ts E/S suppresses the onset of OVA-speciflc allergic airway inflammation
  • mice and DO 11.10 transgenic mice were purchased from Charles River GmbH and Female BALB/c were purchased from Harlan Olac Ltd. Allergic airway hyperreactivity was induced as described in Schn511er et al. J. Immunol. 2008). Briefly, 8 week old female BALB/c mice were sensitized three times (day 0, 14 and 21) via intraperitoneal injection of 20 ⁇ g OVA (grade VI, Sigma- Aldrich) or an equal volume of PBS (control) emulsified in 2mg of Alum (Pierce).
  • mice received ⁇ concentrated Ts E/S supernatant i.p. on day 0, 7, 14 and 21 days post-sensitization.
  • mice received 0.5mg anti-IL-lOR (clone 1B1, a kind gift from the DRFZ, Berlin) i.p. or rat IgG (control) on day 21, 23 and 26 post-sensitization. All mice were subsequently challenged via intranasal administration of 50 ⁇ g OVA in sterile PBS and sacrificed two days later.
  • Airway hyperreactivity was assessed on day 30 via whole-body plethysomography in unrestrained mice in response to increasing doses of metacholine (MCh, Sigma-Aldrich) as described previously (Schn511er et al. 2008, Hamelmann et al. 1997). All mice were maintained in the animal house of the University in accordance with the national animal protection guidelines and approved by the German animal ethics committee.
  • MCh metacholine
  • Lymphocytes from the lung-draining peribroncheolar lymph node (PBLN) and spleen were isolated via manual disruption through a 70 ⁇ nylon mesh and cultured in RPMI 1640 media containing 10% fetal calf serum (FCS), 20mM L-glutamine, 100 U/ml penicillin and 100 ⁇ g/ml streptomycin.
  • FCS fetal calf serum
  • Cells were plated at lxlO 6 cells/ml in a 96 well plate and restimulated with 50 ⁇ g/ml Ovalbumin, 20 ⁇ 1 TsE/S or ⁇ g aCD3 and aCD28 mAb for 48 hours at 37 °C/5% CO 2 and supernatants assessed for cytokine production via ELISA.
  • Intracellular cytokine staining was performed by stimulating 5xl0 6 cells/well with ⁇ g/ml PMA and ⁇ g/ml Ionomycin for 30 minutes followed by the addition of Brefeldin A (5 ⁇ g/ml) (Sigma Aldrich) and a further 3 hour incubation. Cellular proliferation was analyzed via a further 20-hour culture in fresh media containing 1 ⁇ methyl- [ 3 H] -thymidine (Amersham Pharmacia) and subsequent detection using a ⁇ -counter (Perkin-Elmer). De novo regulatory T cell induction assays were performed as previously described by Grainger et al. 2010.
  • Lung tissue was degassed and fixed with 3.7% paraformaldehyde and embedded in paraffin. 2 ⁇ sections were cut, deparaffinised and stained with hematoxylin and eosin (H&E) or Periodic Acid Schiff s reagent to visualize goblet cells. In some experiments sections of lung were stained with an antibody against RELM-a (R&D systems) to visualize alternatively activated macrophages. Goblet cells and RELM-a+ cells were quantified by counting number of cells in 10 HPF (400x magnification). Broncheolar lavage fluid (BAL) cells were differentiated via centrifugation onto frosted slides and differentiated via H&E staining. A minimum of 200 lymphocytes were counted per slide and the percentage of each cell type used to calculate total BAL cell numbers.
  • H&E hematoxylin and eosin
  • R&D systems Periodic Acid Schiff s reagent to visualize goblet cells.
  • Goblet cells and RELM-a+ cells were quant
  • mice that were sensitized and challenged intranasally with ovalbumin (OVA) developed pronounced airway hyperreactivity (AHR) were compared to mice that were sensitised with PBS vehicle only prior to OVA challenge (Fig 3A).
  • Ts E/S treated mice In line with a reduced airway hyperreactivity the local inflammatory infiltrate in the bronchoalveolar lavage fluid (BAL) was found to be ameliorated in Ts E/S treated mice (Fig 3B). In comparison to disease control mice, Ts E/S treated mice had significantly reduced total cell numbers in the BAL, as well as a complete reduction in lymphocyte and eosinophil infiltration (Fig 3B). Furthermore, whereas the Th2 cytokines IL-4, IL-5, IL-10 and IL-13 were found to be elevated in the BAL of disease control mice, IL-4, IL-5 and IL-13 (but not IL-10) were suppressed in the Ts E/S treated group (Fig 3C).
  • Ts E/S treatment resulted in significant reductions in OVA-specific IgM and IgE levels (Fig 3F), and a trend towards reduced OVA-specific IgGl (Fig 3F).
  • Ts E/S treatment elicits a Ts E/S-speciflc Th2 response and antibody production
  • Ts E/S treatment during the priming phase of OVA-specific airway inflammation resulted in a significant reduction in disease-associated inflammation, local cytokine production and OVA-specific antibody production.
  • nematode-derived proteins also elicited potent immune responses. Restimulation of splenocytes with Ts E/S led to the antigen-specific production of IL-4, IL-5, and IL-13 and a significant induction of IL-10 in Ts E/S treated mice (Fig 4A), whereas little cytokine production was detected following restimulation of PBS and OVA control groups that had not received Ts E/S treatment (Fig 4A).
  • Ts E/S treatment In line with a Th2 skewed immune response to Ts E/S we were unable to detect IFN- ⁇ or IL-17A in Ts E/S restimulated cultures (Fig 4A). Furthermore, Ts E/S treatment elicited significant titres of Ts E/S-specific IgM, IgGl and IgE, which could not be detected in control groups (Fig 4B). These results suggest that in addition to modulating responses to bystander antigens (i.e. OVA), Ts E/S treatment also elicits a host Th2 response and production of antibodies directed against these helminth-derived products.
  • OVA bystander antigens
  • Immunomodulatory helminth E/S products have previously been shown to ameliorate inflammation via the induction of regulatory immune cell types, particularly CD4+CD25+FoxP3+ regulatory T cells (Treg).
  • E/S products derived from the murine helminth Heligmosomoides polygyrus bakeri (Hp) have been reported to contain TGF-P-like activity and to directly induce de novo Treg differentiation in vitro and in vivo (Grainger et al. 2010).
  • Ts E/S has a similar potential to induce Treg differentiation whole splenocytes with Ts E/S or Hp E/S were cultured in the presence or absence of the potent T cell stimulating mitogen, ConA (Fig 5A).
  • Ts E/S was unable to induce FoxP3 expression in stimulated and sorted CD4+Foxp3- T cells in vitro (Fig 5A).
  • suppression of allergic airway inflammation was not associated with increased frequencies of Tregs in the spleen, lung draining lymph node (PBLN) or in the lung parenchyma (Fig 5B).
  • Nematodes commonly elicit a so-called "modified Th2 response" in their host, which is associated with the production of high levels of IL-10 leading to the persistence of the nematode infection, but also to suppression of bystander immune responses to unrelated antigens.
  • modified Th2 response is associated with the production of high levels of IL-10 leading to the persistence of the nematode infection, but also to suppression of bystander immune responses to unrelated antigens.
  • Ts E/S induction of IL-10 is at least partially responsible for the suppression of local airway inflammation, although IL-10 independent mechanisms may also contribute to suppression, particularly in regard to antibody production.
  • PCR amplified cDNA of SEQ ID NO: 1 was cloned into pGEM-T Easy vector (Promega) and transformed into competent E. coli cells (JM109).
  • SEQ ID NO: 1 cDNA was subcloned into a eukaryotic expression system of of Leishmania tarentolae (LEXSY, Jena Bioscience), a unicellular-flagellated protozoa. Genome integrated cloning, a Leishmanian secretion system and a C-terminal His-tag enabled recombinant polypeptide purification of SEQ ID NO: 1 from liquid culture media (Fig. 7).
  • Recombinant polypeptide SEQ ID NO: 1 was purified by affinity chromatography using HisTrap excel columns (GE Healthcare) prepacked with Ni Sepharose excel affinity media for capture and purification of histidine-tagged proteins secreted into eukaryotic cell culture supernatants (Fig. 8).
  • Recombinant polypeptide SEQ ID NO: 1 was dialysed against PBS and endotoxin concentrations, detected by Limulus amoebocyte lysate test (Cambrex), were consistently lower than 0.6pg LPS ⁇ g protein.
  • T. suis counterpart of the T. muris protein TMC00067_1

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Toxicology (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Helminthic excretory/secretory products are disclosed herein, which include biologically active polypeptides secreted by Trichuris suis. Such products are useful for modulating an immune response of a subject or in the treatment of certain medical disorders, including those that are Th1- and/or Th2-mediated medical disorders.

Description

T. SUIS EXCRETORY/SECRETORY PRODUCTS AND METHODS OF THEIR USE
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Provisional Application No. 61/887,900 entitled "T. SUIS EXCRETORY/SECRETORY PRODUCTS AND METHODS OF THEIR USE" filed October 7, 2013; and Provisional Application No. 61/940,166 entitled "T. SUIS EXCRETORY/SECRETORY PRODUCTS AND METHODS OF THEIR USE" filed February 14, 2014, and assigned to the assignee hereof and hereby expressly incorporated by reference herein.
FIELD OF THE INVENTION
[0002] The present invention relates generally to helminthic excretory/secretory products. More particularly, the present invention relates to biologically active polypeptides secreted by Trichuris suis and to the use of such isolated polypeptides for modulating an immune response of a subject or in the treatment of certain medical disorders, including those that are Thl- and Th2-mediated medical disorders.
BACKGROUND OF THE INVENTION
[0003] The prevalence of Thl- and Th2-mediated disorders such as allergy, inflammatory and autoimmune diseases in the western world has been correlated with the loss of helminth infections as a consequence of increased hygiene and reduced exposure (Maizels et al. J.Exp. Med. 2009; Maizels et al. Immun.Rev. 2004). Treatment of patients in helminth- endemic countries with antiheminthic drugs has been shown to lead to enhanced allergic reactivity (Flohr et al. Clin. Exp. Allergy 2009; van Biggelaar et al. J. Inf. Dis. 2004) whereas, infection of allergic patients with helminths has been linked to improved health and reduced disease symptoms (Feary et al. Clin. Exp. Allergy 2010). As such, under certain circumstances gastrointestinal helminths can be considered as a beneficial commensal macroflora, which act to induce immunoregulatory pathways necessary for immune homeostasis and the health of the host.
[0004] The helminth Trichuris suis (T. suis), for example, naturally infects the caecum and colon of pigs. In recent years, administration of T. suis ova (TSO) has been proposed as a novel therapeutic in chronic human inflammatory diseases, due to the immunosuppressive potential of therapeutically administered helminth infections. In particular, TSO administration to patients suffering from Crohn's disease and Ulcerative colitis resulted in a significant reduction in clinical symptoms of disease and the onset of remission in many patients (Summers et al. Gut 2005; Summers et al. Gastroenterology 2005), which suggests that TSO administration has therapeutic potential for the treatment of inflammatory human diseases in the clinic. Recently preliminary phase I studies have suggested TSO therapy may also have immunomodulatory and therapeutic potential in patients suffering from multiple sclerosis (Fleming et al. Mult. Scler. 201 1; Benzel et al. J. Helminth. 2011), although larger clinical trials are desired to confirm the efficacy of this therapy.
[0005] The potential success of therapeutic helminth application in patients suffering from autoimmune, allergic or inflammatory diseases may also be hindered by problems with patient compliance to live helminth administration.
[0006] There remains, therefore, a need for improved helminthic therapies that do not require administration of the entire helminth or whole or partial ova or larvae thereof.
SUMMARY OF THE INVENTION
[0007] The present invention relates to novel excretory/secretory polypeptides isolated and purified from a larval stage (LI) of T. suis, as well as recombinant versions of the same. The polypeptides themselves are shown to demonstrate therapeutic and immunomodulatory properties in vivo and may be administered as a pharmaceutical composition, preferably in the absence of the helminth, its ova, or its larvae.
[0008] In particular, one aspect of the present invention provides one or more isolated helminthic excretory/secretory polypeptides, said one or more polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-4 and 37-45, or a biologically active or immunogenic fragment thereof. The amino sequences of SEQ ID NOs: 1-3 and 37-45 include a secretion signal peptide sequence. The mature form of the proteins (i.e., protein minus signal peptide) of SEQ ID NOs: 1-3 and 37-45 consist of the amino acid sequences shown in SEQ ID NOs: 5-7 and 46-54, respectively. Another aspect of the present invention provides one or more isolated helminthic excretory/secretory polypeptides, said one or more polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 5-7 and 46-54 or a biologically active or immunogenic fragment thereof. In some embodiments, the one or more polypeptides are isolated from Trichuris suis. Another aspect of the present invention provides one or more recombinant polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54, or a biologically active or immunogenic fragment thereof. In some embodiments such recombinant polypeptides may be obtained by a process comprising the steps of culturing a cell transformed with a polynucleotide comprising a sequence that encodes an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54, under conditions suitable for polypeptide expression, and recovering the polypeptide so expressed. In some embodiments, the polynucleotide with which the cell is transformed comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 8-28 and 30-36.
[0009] In another aspect, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of one or more recombinant polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54, or a biologically active or immunogenic fragment thereof; and a pharmaceutically acceptable carrier. In yet another aspect, a composition is provided comprising one or more isolated Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, said one or more Ts E/S polypeptides having a molecular weight ranging from about 20 kD and about 80 kD. In some embodiments, the one or more polypeptides are recombinant. In further embodiments, the one or more polypeptides may have an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54, or a biologically active or immunogenic fragment thereof. In still further embodiments, the one or more polypeptides may have a molecular weight of about 58 kD, about 50 kD, about 45 kD, about 35 kD, or about 20 kD. In yet further embodiments, the polypeptides may be obtained from in vitro hatched T. suis first- stage (LI) larvae.
[0010] In yet another aspect of the present invention, a method is provided for modulating a Thl- or Th2-mediated immune response in a subject comprising administering to a subject whose Thl- or Th2-mediated immune response is in need of modulation an effective amount of one or more isolated polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54, a recombinant version thereof, or a biologically active or immunogenic fragment of the foregoing. In yet another aspect of the present invention, a method is provided for modulating a Thl- or Th2-mediated immune response in a subject comprising administering to a subject whose Thl- or Th2 -mediated immune response is in need of modulation a composition comprising an effective amount of one or more polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54, a recombinant version thereof, or a biologically active or immunogenic fragment of the foregoing, wherein the composition is substantially or totally free of Ts eggs and larvae. In yet another aspect, the present invention provides a method of modulating a Thl- or Th2-mediated immune response in a subject comprising administering to a subject whose Thl- or Th2-mediated immune response is in need of modulation a composition comprising Ts E/S supernatants but substantially or totally free of Ts eggs and larvae. In yet another aspect, the present invention provides a method of modulating a Thl- or Th2-mediated immune response in a subject comprising contacting one or more T regulatory cells of a subject with a composition comprising Ts E/S supernatants but substantially or totally free of Ts eggs and larvae.
[0011] In another aspect, the present invention provides a method of regulating expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN-γ in a subject comprising administering to a subject whose expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN-γ are in need of regulation an effective amount of a composition comprising one or more isolated Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, each having a molecular weight ranging from about 20 kD and about 80 kD, and a carrier. In another aspect, the present invention provides a method of regulating expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN-γ in a subject comprising administering to a subject whose expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN-γ are in need of regulation an effective amount of a composition comprising one or more Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, each having a molecular weight ranging from about 20 kD and about 80 kD, and a carrier, wherein the composition is substantially or totally free of Ts eggs and larvae. In another aspect, the present invention provides a method of regulating expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN-γ in a subject comprising administering to a subject whose expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN-γ are in need of regulation a composition comprising Ts E/S supernatants but substantially or totally free of Ts eggs and larvae. [0012] Still another aspect of the present invention provides a method of treating allergic airway hyperreactivity or inflammation in a subject, said method comprising administering to a subject in need thereof an effective amount of a composition comprising one or more isolated Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, each having a molecular weight ranging from about 20 kD and about 80 kD and a carrier. Still another aspect of the present invention provides a method of treating allergic airway hyperreactivity or inflammation in a subject, said method comprising administering to a subject in need thereof an effective amount of a composition comprising one or more Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, each having a molecular weight ranging from about 20 kD and about 80 kD and a carrier, wherein the composition is substantially or totally free of Ts eggs and larvae. Still another aspect of the present invention provides a method of treating allergic airway hyperreactivity or inflammation in a subject, said method comprising administering to a subject in need thereof a composition comprising Ts E/S supernatants but substantially or totally free of Ts eggs and larvae
[0013] In yet another aspect, the present invention provides a composition comprising a mixture of Trichuris suis (Ts) excretory/secretory (E/S) polypeptides obtained by culturing Ts first stage larvae in an aqueous medium, harvesting a portion of the medium at predetermined intervals while replenishing the medium over a period of about one to two weeks. In some embodiments, 50,000 larvae/well may be cultured on a 24-well plate using 0.5 mL of serum- free medium per well. 0.4 mL of supernatant may be discarded after a period of 24 hours with the medium being replenished. In further embodiments, 0.4 mL of supernatant is subsequently harvested every 48 hours with the medium being replenished over a period of ten days. In yet further embodiments, 0.4 mL of supernatant may be subsequently harvested every 48 hours with the medium being replenished over a period of fourteen days.
[0014] Another aspect of the present invention provides a protein pellet obtained by acidifying the composition described above. The protein pellet may, in some embodiments, be obtained by adding 100% trichloroacetic acid (TCA) at 1 :5 ratio of TCA volume to sample volume.
[0015] In still another aspect of the present invention, a method is provided for modulating an immune response of a subject comprising administering to a subject whose immune response is in need of modulation an effective amount of a composition comprising a mixture of isolated Ts E/S polypeptides. In some embodiments, splenocytes of a treated subject may exhibit suppressed IFN-γ and IL-4 production compared to splenocytes of a control subject. In further embodiments, dendritic cells of a treated subject may exhibit no upregulation in their expression of CD86 and MHC class II molecules compared to dendritic cells of a control subject. The dendritic cells of a treated subject may, in some embodiments, exhibit suppressed IL-6 production compared to dendritic cells of a control subject. In still another aspect of the present invention, a method is provided for modulating an immune response of a subject comprising administering to a subject whose immune response is in need of modulation an effective amount of a composition comprising a mixture of Ts E/S polypeptides, wherein the composition is substantially or totally free of Ts eggs and larvae.
[0016] Yet another aspect of the present invention provides a method of alleviating allergic airway hyperreactivity or inflammation comprising administering to a subject in need thereof an effective amount of a composition comprising a mixture of isolated Ts E/S polypeptides. In some embodiments, treated subjects exhibit reduced lymphocyte and eosinophil infiltration compared to a control subject. In further embodiments, treated subjects may exhibit suppressed IL-4, IL-5 and IL-13 production compared to a control subject. Yet another aspect of the present invention provides a method of alleviating allergic airway hyperreactivity or inflammation comprising administering to a subject in need thereof an effective amount of a composition comprising a mixture of Ts E/S polypeptides, wherein the composition is substantially or totally free of Ts eggs and larvae.
[0017] In yet another aspect, the polypeptides that are the subject of the present invention may comprise, or alternatively consist essentially of, or yet further consist of, a "biologically equivalent" or "biologically active" polypeptide encoded by equivalent polynucleotides as described herein. They may possess at least 60%, or alternatively, at least 65%, or alternatively, at least 70%, or alternatively, at least 75%, or alternatively, at least 80%, or alternatively at least 85%, or alternatively at least 90%, or alternatively at least 95% or alternatively at least 98%, identical primary amino acid sequence to the reference polypeptide (i.e., a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 and 37-54 or a fragment thereof) when compared using sequence identity methods run under default conditions. [0018] As used herein, "about" will be understood by persons of ordinary skill in the art and will vary to some extent depending upon the context in which it is used. If there are uses of the term which are not clear to persons of ordinary skill in the art given the context in which it is used, "about" will mean up to plus or minus 10% of the particular term.
[0019] As used herein, the term "biologically active" is used to refer to a protein or polypeptide that induces a desired beneficial effect when orally administered to a subject and includes agents that are therapeutically and/or prophylactically effective as pharmaceuticals ("pharmacologically active agents"). By an "effective" amount of an active agent is meant a nontoxic but sufficient amount of an active agent to provide the desired beneficial effect.
[0020] The term "isolated" peptide fragment is meant to include peptide fragments which are not naturally occurring as fragments and would not be found in the natural state. The term "isolated" is also used herein to refer to polypeptides and proteins that are isolated from other cellular proteins and is meant to encompass both purified and recombinant polypeptides. In other embodiments, the term "isolated" means separated from constituents, cellular and otherwise, in which the cell, tissue, polynucleotide, peptide, polypeptide, protein, antibody or fragment(s) thereof, which are normally associated in nature. For example, an isolated cell is a cell that is separated form tissue or cells of dissimilar phenotype or genotype. As is apparent to those of skill in the art, a non-naturally occurring polynucleotide, peptide, polypeptide, protein, antibody or fragment(s) thereof, does not require "isolation" to distinguish it from its naturally occurring counterpart.
[0021] The term "polypeptide" is used interchangeably with the term "protein" and in its broadest sense refers to a compound of two or more subunit amino acids, amino acid analogs or peptidomimetics. The subunits may be linked by peptide bonds. In another embodiment, the subunit may be linked by other bonds, e.g., ester, ether, etc. As used herein the term "amino acid" refers to natural and/or unnatural or synthetic amino acids, including glycine and both the D and L optical isomers, amino acid analogs and peptidomimetics. A peptide of three or more amino acids is commonly called an oligopeptide if the peptide chain is short. If the peptide chain is long, the peptide is commonly called a polypeptide or a protein. The term "peptide fragment," as used herein, also refers to a peptide chain. [0022] "Homology" or "identity" or "similarity" are synonymously and refers to sequence similarity between two peptides or between two nucleic acid molecules. Homology can be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same base or amino acid, then the molecules are homologous at that position. A degree of homology between sequences is a function of the number of matching or homologous positions shared by the sequences. An "unrelated" or "non-homologous" sequence shares less than 40% identity, or alternatively less than 25% identity, with one of the sequences of the present invention.
[0023] The terms "culture" or "culturing" refer to the in vitro propagation of cells or organisms on or in media of various kinds. It is understood that the descendants of a cell grown in culture may not be completely identical (i.e., morphologically, genetically, or phenotypically) to the parent cell.
[0024] A "composition" is intended to mean a combination of active polypeptides and another compound or composition, inert (e.g. a detectable label) or active (e.g. a delivery vehicle) alone or in combination with a carrier which can in one embodiment be a simple carrier like saline or pharmaceutically acceptable or a solid support as defined below.
[0025] A "pharmaceutical composition" is intended to include the combination of an active polypeptide, polynucleotide or antibody with a carrier, inert or active such as a solid support, making the composition suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo.
[0026] As used herein, the term "pharmaceutically acceptable carrier" encompasses any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see Martin (1975) Remington's Pharm. Sci., 15th Ed. (Mack Publ. Co., Easton).
[0027] A "subject," "individual," or "patient" is used interchangeably herein, and refers to a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murines, rats, rabbits, simians, bovines, ovines, porcines, canines, felines, farm animals, sport animals, pets, equines, and primates, particularly humans. [0028] The terms "disease" and "disorder" are used inclusively and refer to any condition associated with improper function of the immune system of a subject.
[0029] As used herein, the term "egg" refers to either embryonated or nonembryonated, viable T. suis ova.
[0030] Applicants have provided herein the polypeptide and/or polynucleotide sequences for use in gene and protein transfer and expression techniques described below. It should be understood, although not always explicitly stated that the sequences provided herein can be used to provide the expression product as well as substantially identical sequences that produce a protein that has the same biological properties. These "biologically equivalent" or "biologically active" polypeptides are encoded by equivalent polynucleotides. They may possess at least 60%, or alternatively, at least 65%, or alternatively, at least 70%, or alternatively, at least 75%, or alternatively, at least 80%, or alternatively at least 85%, or alternatively at least 90%, or alternatively at least 95% or alternatively at least 98%, identical primary amino acid sequence to the reference polypeptide when compared using sequence identity methods run under default conditions. Specific polypeptide sequences are provided as examples of particular embodiments. Modifications to the sequences to amino acids with alternate amino acids that have similar charge.
[0031] Additional features, advantages, and embodiments of the present disclosure may be set forth from consideration of the following detailed description, drawings, and claims. Moreover, it is to be understood that both the foregoing summary of the present disclosure and the following detailed description are exemplary and intended to provide further explanation without further limiting the scope of the present disclosure claimed.
BRIEF DESCRIPTION OF THE DRAWINGS
[0032] All of the data shown in the Figures described below were derived from experiments in the murine system.
[0033] Figure 1A depicts T. suis eggs (TSO), first stage larvae (LI) and the timeline of experimentation in which the eggs were hatched, washed thoroughly and cultured in vitro for 10-14 days during which time supernatants were harvested every two days (arrows). Figure IB is a photograph showing electrophoresis results characterizing T. suis first stage larval E/S proteins. [0034] Figure 2 is a series of graphs showing the suppression of antigen-specific CD4+ T cell cytokine production by Ts E/S, as well as modulation of dendritic cell activation in vitro. I A and B show the proliferation of ovalbumin-specific CD4+ T cells treated with decreasing volumes of Ts E/S supematants for 72 hours and (A) incubated with 3H thymidine for a further 20 hours to assess proliferation, (B) and supematants analyzed via ELISA for production of IFN-γ or IL-4. C+D. Bone marrow-derived dendritic cells (DCs) were pre- treated with Ts E/S supernatant prior to pulsing with (C) the TLR-9 agonist CpG or (D) the TLR-4 agonist LPS and DC activation was assessed via quantification of cells expressing the co-stimulatory markers CD86 and MHCII via flow cytometry and by production of the proinflammatory cytokines IL-6 and IL-12/23p40 by ELISA.
[0035] Figure 3(A)-(F) is a series of graphs and photographs showing the suppression of the development of allergic airway inflammation in mice that were sensitized by intraperitoneal injection of PBS or OVA. (A) is a line graph showing airway hyperreactivity in response to increasing concentrations of metacholine; (B) is a series of cytology results showing cellular infiltrate into bronchiolar lavage fluid; (C) is a series of bar graphs showing total cytokine concentrations in BAL; (D) is a series of bar graphs showing ELISA results of total cytokine concentrations in splenocytes following OVA restimulation; (E) is a series of photographs and of histological staining (RELM-a-specific antibody staining in the lung parenchyma; and (F) is a series of graphs showing concentration of ova-specific cytokine isotopes in serum of PBS, OVA, and Ts E/S mice.
[0036] Figure 4 is two sets of graphs showing that Ts E/S treatment induces a T. suis- specific Th2 response and antibody production. Figure 2(A) is a series of six bar graphs showing Ts E/S-specific cytokine production assessment via ELISA in culture supematants of splenocytes following restimulation with Ts E/S ex vivo for 24 hours. Figure 2(B) is a series of three plots showing Ts E/S-specific antibody titres in the serum.
[0037] Figure 5 shows that Ts E/S amelioration of allergic airway disease is not associated with increased Treg frequencies but is partially dependent on IL-10 production. (A) is a quantification of FoxP3 expression in splenocytes in vitro stimulated with ConA +/- Ts E/S or Hp E/S or heat inactivated E/S proteins. (B) is a quantification of CD25+ FoxP3+ regulatory T cells as a frequency of the CD4+ T cell compartment in the spleen and peribroncheolar lymph nodes (PBLN) by flow cytometry, and (C) is a quantification of FoxP3+ cells in the lung parenchyma by immunohistochemistry staining. (D) is an experiment schematic for antibody neutralization of IL-10R in OVA-induced allergic airway hyperreactive mice. (E) is a quantification and differentiation of cell infiltrate in the BAL. (F) is a quantification of IL- 5 and IL-13 in BAL fluid via ELISA. (G) is a quantification of OVA-specific antibody titers in the serum of OVA-induced allergic airway hyperreactive mice +/- alL-lOR treatment.
[0038] Figure 6 shows PCR amplification of SEQ ID NO: 1. Agarose gel electrophoresis of SEQ ID NO: 1 amplification. Lane 1, Marker (M; 2-log DNA Ladder); lane 2, empty; lane 3, SEQ ID NO: 1 (804 bp).
[0039] Figure 7 shows secretion of recombinant polypeptide SEQ ID NO: 1. Coomassie staining of SEQ ID NO: 1 transfected L. tarentolae cells. Lane 1, Marker (M, 7-175 kDa); Lane 2, lysed L. tarentolae cells (Ctrl); Lane 3, TCA precipitation of 5 ml SEQ ID NO: 1 transfected L. tarentolae cell culture supernatant (Culture media).
[0040] Figure 8 shows purification of recombinant polypeptide SEQ ID NO: 1. Coomassie stained SDS-PAGE of His-tagged polypeptide SEQ ID NO: 1 HPLC purification showing representative aliquots of wash (Lane 2, Ctrl) and elution (Lane 3, HisTrap Elution) fractions.
[0041] Figure 9 shows that SEQ ID NO: 1 suppresses antigen-specific proliferation of T cells co-cultured with Ova pulsed DCs. Purified CD4+ T cells from Ova-tg D01 1.10 mice were labeled with CFSE cell proliferation dye and co-cultured with Ova-peptide pulsed DCs in the absence (+ Ctrl.) or presence of recombinant SEQ ID NO: 1 or heat-inactivated SEQ ID NO: 1. CFSE dilution was analyzed 5 days after beginning of co-culture. **p=0.0045, n=6.
[0042] Figure 10 shows that recombinant SEQ ID NO: 1 increases IL-10 secretion in CD4+ T cell - DC co-cultures. Supernatant of CD4+ T cell - DC co-cultures was analyzed for IFNg, IL-12 and IL-10 cytokine levels by ELISA. *p=0.0335, n=3.
DESCRIPTION OF SEQUENCES
SEQ ID NO: 1 is the full length amino acid sequence of a T. suis Locus_7131_l/l protein.
SEQ ID NO: 2 is the full length amino acid sequence of a T. suis Locus_1063_l/1 T. suis protein. SEQ ID NO: 3 is the full length amino acid sequence of a T. suis Locus_1815_l/l protein.
SEQ ID NO: 4 is the amino acid sequence of a T. suis protein homolog of T. muris accession number TMC00067 1.
SEQ ID NO: 5 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 1 (i.e., SEQ ID NO: 1 minus the signal peptide sequence).
SEQ ID NO: 6 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 2 (i.e., SEQ ID NO: 2 minus the signal peptide sequence).
SEQ ID NO: 7 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 3 (i.e., SEQ ID NO: 3 minus the signal peptide sequence).
SEQ ID NOs: 8-10 and 30 are exemplary cDNA sequences encoding the full length protein of SEQ ID NO: 1.
SEQ ID NOs: 11-13 and 31 are exemplary cDNA sequences encoding the full length protein of SEQ ID NO: 2.
SEQ ID NOs: 14-16 and 32 are exemplary cDNA sequences encoding the full length protein of SEQ ID NO: 3.
SEQ ID NOs: 17-19 and 33 are exemplary cDNA sequences encoding the protein of SEQ ID NO: 4.
SEQ ID NOs: 20-22 and 34 are exemplary cDNA sequences encoding the mature protein of SEQ ID NO: 5.
SEQ ID NOs: 23-25 and 35 are exemplary cDNA sequences encoding the mature protein of SEQ ID NO: 6.
SEQ ID NOs: 26-28 and 36 are exemplary cDNA sequences encoding the mature protein of SEQ ID NO: 7.
SEQ ID NO: 29 is the amino acid sequence of a Trichuris muris protein.
SEQ ID NO: 37 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide. SEQ ID NO: 46 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 37. SEQ ID NO: 38 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide. SEQ ID NO: 47 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 38.
SEQ ID NO: 39 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide. SEQ ID NO: 48 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 39.
SEQ ID NO: 40 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide. SEQ ID NO: 49 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 40.
SEQ ID NO: 41 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide. SEQ ID NO: 50 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 41.
SEQ ID NO: 42 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide. SEQ ID NO: 51 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 42.
SEQ ID NO: 43 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide. SEQ ID NO: 52 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 43.
SEQ ID NO: 44 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide. SEQ ID NO: 53 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 44.
SEQ ID NO: 45 is the full length amino acid sequence of a T. suis excretory/secretory polypeptide. SEQ ID NO: 54 is the amino acid sequence of the mature form of the protein of SEQ ID NO: 45.
DETAILED DESCRIPTION
[0043] The present disclosure demonstrates for the first time that E/S products derived from the larval stage of T. suis (Ts E/S) have potent immunomodulatory effects using both in vitro and in vivo murine model systems of human disease. Administration of soluble products derived from helminths is advantageous as it negates the need to expose patients to live parasites. Excretory/Secretory Polypeptides
[0044] Immunomodulatory potential of excretory/secretory supernatants (E/S) was derived from the first stage larvae of T. suis (Ts E/S) - thus, mirroring the expected situation in patients undergoing transient exposure to larval stages during multiple applications of TSO. Ts E/S was found to have potent immunomodulatory capacity using both in vitro systems and in vivo murine model of disease. In particular, Ts E/S was found to significantly reduce disease severity and immunopathology in murine models of asthma (allergic airway hyperreactivity). Ts E/S was used in the experiments described herein.
[0045] Proteomic analysis of Ts E/S identified 4 T. suis proteins by LC-MS/MS spectrometry and subsequent database searches. Three of the identified proteins are specific for T. suis and could only be found in the T. suis transcriptome data base (www.helmdb.org) in contrast to the available Trichuris EST database "NEMBASE4" (www.nematodes,org) comprising of data from Trichuris muris, Trichineiia spiralis and Trichuris vulpis or the NCBI database. HelmDB comprises 1288 protein sequences translated from transcriptomic data of T. suis. In particular 2 out of the 3 identified proteins are not only novel proteins but being identified with a high degree of confidence (Mascot Score 542 and 172) and identified by more than 2 matching peptides. The fourth protein was identified based on NemBase4 matching a Trichuris muris sequence. Molecular cloning and production of these specific immunomodulatory components from the clinically therapeutic helminth T. suis will allow for the development of a refined therapy without need for patient exposure to live parasites.
[0046] Polypeptides comprising, or alternatively consisting essentially of, or yet further consisting of, the amino acid sequences of the invention (SEQ ID NOs: 1-7 and 37-54 or a fragment or variant thereof) can be prepared by expressing polynucleotides encoding the polypeptide sequences of this invention in an appropriate host cell. This can be accomplished by methods of recombinant DNA technology known to those skilled in the art, employing, for example, a cDNA having a sequence selected from the group consisting of SEQ ID NOs: 8- 28 and 30-36. Accordingly, this invention also provides methods for recombinantly producing the polypeptides of this invention in a eukaryotic or prokaryotic host cell, which in one aspect is further isolated from the host cell. The amino acid sequences of SEQ ID NOs: 1-3 and 37-45 are full length proteins, i.e., they include a secretion signal peptide. Accordingly, the nucleotide sequences encoding SEQ ID NOs: 1-3 and 37-45 include a portion that encodes the secretion signal peptide. The protein of any of SEQ ID NOs: 1-3 and 37-45 minus the secretion signal peptide is referred to as the mature protein. The mature proteins of the invention comprise or consist of an amino acid sequence selected from the group consisting of SEQ ID NO: 5 (the mature protein of SEQ ID NO: 1), SEQ ID NO: 6 (the mature protein of SEQ ID NO: 2), SEQ ID NO: 7 (the mature protein of SEQ ID NO: 3), SEQ ID NO: 46 (the mature protein of SEQ ID NO: 37), SEQ ID NO: 47 (the mature protein of SEQ ID NO: 38), SEQ ID NO: 48 (the mature protein of SEQ ID NO: 39), SEQ ID NO: 49 (the mature protein of SEQ ID NO: 40), SEQ ID NO: 50 (the mature protein of SEQ ID NO: 41), SEQ ID NO: 51 (the mature protein of SEQ ID NO: 42), SEQ ID NO: 52 (the mature protein of SEQ ID NO: 43), SEQ ID NO: 53 (the mature protein of SEQ ID NO: 44), and SEQ ID NO: 54 (the mature protein of SEQ ID NO: 45). In some embodiments, a polynucleotide sequence encoding a mature protein having a sequence selected from SEQ ID NOs: 5-7 and 46-54 may be linked to a polynucleotide sequence encoding a particular secretion signal peptide to produce a full length nucleotide sequence for expression/secretion in a particular cell type.
[0047] The proteins and peptide fragments of this invention also can be obtained by chemical synthesis using a commercially available automated peptide synthesizer such as those manufactured by Perkin Elmer/Applied Biosystems, Inc., Model 430A or 431A, Foster City, CA, USA. The synthesized protein or polypeptide can be precipitated and further purified, for example by high performance liquid chromatography (HPLC). Accordingly, this invention also provides a process for chemically synthesizing the proteins of this invention by providing the sequence of the protein and reagents, such as amino acids and enzymes and linking together the amino acids in the proper orientation and linear sequence.
Compositions for Therapy
[0048] One or more of the above polypeptides, or polynucleotides encoding these compositions, can be further combined with a carrier, a pharmaceutically acceptable carrier or medical device which is suitable for use of the compositions in diagnostic or therapeutic methods. Thus, the compositions comprise, or alternatively consist essentially of, or yet further consists of, one or more of the above compositions described above in combination with a carrier, a pharmaceutically acceptable carrier or medical device. [0049] The carrier can be a liquid phase carrier or a solid phase carrier, e.g., bead, gel, microarray, or carrier molecule such as a liposome. The composition can optionally further comprise at least one further compound, protein or composition.
[0050] Additional examples of "carriers" includes therapeutically active agents such as another peptide or protein (e.g., an Fab' fragment). For example, an antibody of this invention, derivative or fragment thereof can be functionally linked (e.g., by chemical coupling, genetic fusion, noncovalent association, or otherwise) to one or more other molecular entities, such as another antibody (e.g., to produce a bispecific or a multispecific antibody), a cytotoxin, a cellular ligand, or an antigen. Accordingly, this invention encompasses a large variety of antibody conjugates, bi- and multispecific molecules, and fusion proteins, whether or not they target the same epitope as the antibodies of this invention.
[0051] Yet additional examples of carriers are organic molecules (also termed modifying agents) or activating agents, that can be covalently attached, directly or indirectly, to an antibody of this invention. Attachment of the molecule can improve pharmacokinetic properties (e.g., increased in vivo serum half-life). Examples of organic molecules include, but are not limited to a hydrophilic polymeric group, a fatty acid group or a fatty acid ester group. As used herein, the term "fatty acid" encompasses mono-carboxylic acids and di- carboxylic acids. A "hydrophilic polymeric group," as the term is used herein, refers to an organic polymer that is more soluble in water than in octane.
Methods of Use of Polypeptides and Their Compositions
[0052] Applicants have discovered that the polypeptides of this invention modulate a Thl- or Th2-mediated immune response in a subject and regulate expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN-γ. In one aspect, the Thl- or Th2-mediated response is allergic airway hyperreactivity or inflammation. Thus, methods to achieve such in vitro or in vivo responses are provided by contacting or administering an effective amount of the polypeptide of this invention to a subject in need of such treatment. Administration can be by any suitable method and effective amounts can be empirically determined by a treating physician. [0053] An "effective amount" is an amount sufficient to effect beneficial or desired results. An effective amount can be administered in one or more administrations, applications or dosages. Such delivery is dependent on a number of variables including the time period for which the individual dosage unit is to be used, the bioavailability of the therapeutic agent, the route of administration, etc. It is understood, however, that specific dose levels of the therapeutic agents of the present invention for any particular subject depends upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, and diet of the subject, the time of administration, the rate of excretion, the drug combination, and the severity of the particular disorder being treated and form of administration. Treatment dosages generally may be titrated to optimize safety and efficacy. Typically, dosage-effect relationships from in vitro and/or in vivo tests initially can provide useful guidance on the proper doses for patient administration. In some embodiment, an "effective amount" corresponds to an amount of polypeptide administered to prevent or reduce an inflammatory response. In some embodiments, an "effective amount" corresponds to the amount of polypeptide given in a pre-challenge.
[0054] The "therapeutically effective amount" will vary depending on the polypeptide, the Thl- or Th2-mediated disorder and its severity and the age, weight, etc., of the patient to be treated all of which is within the skill of the attending clinician. It is contemplated that a therapeutically effective amount of a polypeptide described herein will decrease levels of inflammation in the patient as compared to the levels of inflammation in the absence of treatment. A therapeutically effective amount is distinguishable from an amount having a biological effect (a "biologically effective amount"). A biological effect, however, may not result in any clinically measurable therapeutically effect as described above as determined by methods within the skill of the attending clinician.
[0055] Administration in vivo can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell, solid tumor or cancer being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents can be found below. [0056] The pharmaceutical compositions can be administered orally, intranasally, parenterally, injection, orally and may take the form of tablets, lozenges, granules, capsules, pills, ampoules, suppositories or aerosol form. They may also take the form of suspensions, solutions and emulsions of the active ingredient in aqueous or nonaqueous diluents, syrups, granulates or powders. In addition to an agent of the present invention, the pharmaceutical compositions can also contain other pharmaceutically active compounds or a plurality of compounds of the invention.
[0057] More particularly, an agent of the present invention also referred to herein as the active ingredient, may be administered for therapy by any suitable route including oral, rectal, nasal, topical (including transdermal, aerosol, buccal and sublingual), vaginal, intraperitoneal, parenteral (including subcutaneous, intramuscular, intravenous and intradermal) and pulmonary. It will also be appreciated that the preferred route will vary with the condition and age of the recipient, and the disease being treated.
[0058] Ideally, the agent should be administered to achieve peak concentrations of the active compound at sites of disease. This may be achieved, for example, by the intravenous injection of the agent, optionally in saline, or orally administered, for example, as a tablet, capsule or syrup containing the active ingredient. Desirable blood levels of the agent may be maintained by a continuous infusion to provide a therapeutic amount of the active ingredient within disease tissue. The use of operative combinations is contemplated to provide therapeutic combinations requiring a lower total dosage of each component agent than may be required when each individual therapeutic compound or drug is used alone, thereby reducing adverse effects.
[0059] While it is possible for the agent to be administered alone, it is preferable to present it as a pharmaceutical formulation comprising at least one active ingredient, as defined above, together with one or more pharmaceutically acceptable carriers therefor and optionally other therapeutic agents. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
[0060] Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets, each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
[0061] A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder (e.g., povidone, gelatin, hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (e.g., sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose) surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
[0062] Formulations suitable for parenteral administration include aqueous and nonaqueous isotonic sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents, and liposomes or other microparticulate systems which are designed to target the compound to blood components or one or more organs. The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
[0063] It should be understood that in addition to the ingredients particularly mentioned above, the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example, those suitable for oral administration may include such further agents as sweeteners, thickeners and flavoring agents. It also is intended that the agents, compositions and methods of this invention be combined with other suitable compositions and therapies.
[0064] The present invention is further illustrated by the following examples, which should not be construed as limiting in any way.
EXAMPLES
Example 1; Determination and Characterization of 71 suis Secretome
[0065] In order to demonstrate the efficacy of helminthic secretory proteins in combating Thl- and Th2-mediated disorders, an exemplary T. suis secretome is first obtained. Ova of Trichuris suis, were incubated, hatched into larvae, maintained as larvae, and induced to secrete products, which were then collected, as described below.
Harvesting of larval (LI) Trichuris suis Excretory/Secretory supernatants (Ts E/S)
[0066] 250,000 purified T. suis ova (Ovamed GmbH) were resuspended in 8 ml PBS mixed with 12 ml 6% Hypochloride solution and incubated for 45 minutes at 37°C, 5% CC^ with periodical agitation. T. suis ova were washed via multiple rounds of centrifugation at 2000 RPM (425 x g) for 10 minutes and resuspended in serum free RPMI-1640 media containing 20 mM L-glutamine, 100 U/ml penicillin and 100 μg/ml streptomycin (Invitrogen). Hatched T. suis first stage larvae were then cultured at 50,000 larvae/well in a 24 well plate in a volume of 0.5 ml serum free media. Following 24 hours 400 μΐ of supernatant was discarded and fresh media added. Supernatants were then harvested every 48 hours for 10-14 days or until the larvae were no longer viable and stored at -20°C until further use. To generate Ts E/S protein for gel visualization supernatants were precipitated via the addition of 100% TCA at a 1 :5 ratio of TCA volume:sample volume. The total dried protein pellet was resuspended in loading buffer and run on a 12% SDS gel and subsequently visualized with a commercially available silver staining kit following manufacturer's instructions (Thermo Scientific). Alternatively, protein pellet was used for LC-MS/MS spectrometry to identify the protein content. To generate Ts E/S supernatants for use in in vitro and in vivo experiments, supernatants were concentrated 20x with centrifugal concentrators with a molecular weight cut off of 5000 PES (Vivaspin 6; Viva products GmbH) and used as indicated below. Final Endotoxin levels were detected by a Limulus amoebocyte lysate test (Cambrex) and consistently found to be ≤0.2 pg/μΐ. E/S containing supernatants were then concentrated/precipitated and the concentrated E/S was analysed via silver staining (Fig IB). LI T. suis E/S (Ts E/S) contained at least 7 distinct protein bands.
Tryptic digestion ofTs E/S proteins for LC-MS/MS
[0067] TCA-precipitated dried protein pellets of Ts E/S were suspended in 25 μΐ of 0.1M ammonium bicarbonate buffer. For reduction and alkylation of cysteine residues 4 μΕ dithiotreitol (45 mM in 0.1 M ammonium bicarbonate) was added and the mixture was incubated at 56°C for 45 min. After cooling to room temperature 4 μΕ iodoacetamide solution (100 mM in 0.1 M ammonium biocarbonate) was applied and the sample was kept in the dark for 30 min. Then the solution was diluted with 95 μΐ of 0.1 M ammonium bicarbonate. Trypsin was dissolved in 50 mM acedic acid und diluted to a final concentration of 0.1 μg/μl and kept at -80°C until use. In-solution digests were performed with 0.5 or Ι μΕ of trypsin (0.1 μg/μL) for 16-18 h at 37°C. The reaction was stopped with 2.5 μΐ of 20% (v/v) trifluoroacetic acid (TFA) in water and samples were dried in a vacuum centrifuge.
[0068] For a first characterization of the protein composition of Ts E/S, 1 D gel electrophoresis and silver staining were performed (Fig IB). Using this approach at least 7 distinct Ts E/S protein bands were visualized. For identification of these proteins, E/S containing supernatants were precipitated and analysed by LC-MS/MS.
LC-MALDI MS/MS ofTs E/S
[0069] LC-MS/MS analyses were performed on a 4700 proteomics Analyzer (ABSCIEX, Framingham, MS) off-line coupled with a Thermo Scientific Dionex UltiMate 3000 RSLC system and Probot fractionation device (Thermo Scientific, Idstein, Germany). Dried peptides were reconstituted in 12 μΐ of 0.1% (v/v) TFA in water. Ten μΐ of the sample were concentrated on a trap column (Acclaim-PepMap CI 8, 3 μιη, 2 cm x 75 μιη i.d. Thermo Scientific). LC separations were performed on an analytical column (Acclaim-PepMap CI 8, 2 μιη, 150 mm x 75 μιη, Thermo Scientific) at a flow rate of 300 nl/min. Mobile phase (A) was 0.1% (v/v) TFA in water and (B) was 80:20 (v/v) acetonitrile/water containing 0.08% (v/v) TFA. The gradient was 3-15% B in 2 min and 15-60% B in 60 min. Column effluent was continuously mixed with MALDI matrix (5 mg/ml a-cyano-4-hydroxycinnamic acid in 70:30 (v/v) acetonitrile/water containing 0.1% (v/v) TFA, 1 μΐ/min) and spotted at ten-second intervals on 26 x 12 spot arrays on MALDI steel targets (ABSCIEX). Mass spectra were acquired in a data-dependent mode. The MS spectra were recorded in the mass range of mlz 80C 000 and with the accumulation of 2000 subspectra. MS/MS spectra were measured from the five most intensive precursor ions (S/N > 30). 5000-10,000 laser shots were accumulated.
Identified peptides
[0070] In principle, the mass spectra (MS) obtained by peptide fragments produces a mass list that can be used to interrogate a database of known proteins that has to be theoretically digested with trypsin. In case of MS/MS the resulting data are further supported by sequence information of the peptide fragmentation spectrum. Mascot software used for protein identification of T. suis LI E/S compared the observed MS spectra to databases containing known nematode proteins ( CBI, NemBase4, and HelmDB). Peptides that identified LI E/S proteins with a significant level of confidence are listed in Table 1. Peptides are grouped by the protein they match. The first column of Table 1 lists the "database" where the matching protein sequence information was obtained. The second column is the respective database specific "accession ID," followed by internal "description and species." For each identified protein, the software calculates an overall protein score (Mascot score). The column "Mascot score" reflects the combined scores of all observed mass spectra that can be matched to amino acid sequences within that protein. The higher the Mascot score, the more confident is the match. The column "Peptide overage" indicates the percent of protein sequence information that is covered by the sum of all matched peptides. The next two columns list peptide mass specific information, by showing the observed peptide mass obtained from the spectra (Peptide mass exp) and the calculated mass (Peptide mass calc) resulting from theoretical tryptic digests of the database sequence information. The "peptide score" is a measure of how well the observed MS/MS spectra match to the stated peptide. The following "Identity score" is a probability score that comprises the peptide length, the trypsin target sites and the possibility of incomplete proteolytic cleavage. Notably, the peptide score should be higher than the identity score to significantly identify a protein. Finally, the "p- value" indicates the probability that the observed match between spectra and peptide sequence has been found by chance. In the last column the matching peptide "sequences" are listed. [0071] The tryptic digested LI E/S was analyzed by LC-MS/MS spectrometry as described above. The resulting masses of peptide fragments (MS spectra) and the sequence data obtained for the specific peptides (MS/MS spectra) were matched to theoretical fragmentation pattern of different protein databases. Database searches using NCBI, NemBase4 and HelmDB identified a number of distinct LI E/S proteins with a significant Mascot Score (Table 1 and Table 2) only in HelmDB. The protein annotated as Locus_7131_l/l (T. suis transcriptome, HelmDB) has a molecular weight of 32510 Da and was identified based on 8 individual matched peptides with a Mascot Score of 447, covering together 25% of the protein sequence (Table 2). Further database searches with the full Locus_7131_l/l sequence identified no homology to any known sequences and no gene ontologies. Three individual peptides matched the protein sequence of Locus_1063_l/1 (T. suis transcriptome, HelmDB, 26.5% sequence coverage) with a Mascot Score of 128. This locus describes a protein with an estimated mass of 15737 Da. Homology searches with the full sequence information revealed homology to an unknown protein from Trichinella spiralis. Locus_1815_l/l (40146 Da) was identified with a Mascot score of 83 by two different peptides covering 6% sequence information (T. suis transcriptome, HelmDB). Further database search predicted the gene ontology terms: serine-type endopeptidase activity; proteolysis.
Table 1:
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
a modified amino acids: Ccarb=carbadidomethylated cysteine; pyrQ=pyroglutamat
Processing of MS data
[0072] MS and MS/MS peaklists were generated by the "Peak to Mascot" tool of the 4000er Series Expolerer v 3.6. For MS/MS data analysis MASCOT server (version 2.2, Matrixscience, London, UK) was used. Data base searches were performed using NCBI (31.601.460 protein sequences), Helm database (1288 protein sequences translated from transcriptome data of T. suis, (www.helmdb.org), and Trichuris EST database (in-house database composed of 12.895 cDNA sequences from Trichuris muris, Trichinella spiralis und Trichuris vulpis, made from www.nematodes.org/nembase4/overview.shtml). The following parameters were set: enzyme: trypsin with two missed cleavages or no enzyme, variable modifications: carbamidomethylation (C), oxidation (M), pyro-glu (Q), mass tolerances for MS and MS/MS: 80 ppm and 0.3 Da. Proteins were accepted as identified if at least two MS/MS spectra provided a MASCOT score for identity (p<0.01). MS/MS spectra which did not fulfill these criteria were inspected manually and their identity was verified, when the fragment pattern was comparable with those of synthetic analogs. These reference peptides were synthesized in-house using Fmoc solid phase chemistry.
[0073] Protein identification revealed a number of protein matches based on NemBase4, HelmDB and NCBI. Table 2 lists all proteins centered information according to an internal ranking (first column). In addition, Table 2 lists the expected protein weight in Da, which is calculated based on the available sequence information. The last column illustrates the complete protein sequence obtained from the indicated database. The respective peptides matching to each protein are underlined.
TABLE 2
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
[0074] In order to clone and express the sequences identified above, snap frozen T. suis LI larvae will be used for isolation of RNA. For that purpose T. suis LI (lmg) will be ground in liquid nitrogen before being homogenized with the QIAshredder column (Quiagen). RNA will be isolated from the lysate using RNeasy Mini Kit (Quiagen) according to manufacturer's instruction. Integrity of the total RNA will be proofed by gel electrophoresis in 1% agarose, which have to yield two bands corresponding to 28 S and 18 S ribosomal RNA showing no smear confirming the integrity of the total RNA. Concentration of total RNA will be measured using a Nanodrop Spectrophotometer (PeqLab). 1 unit of adsorbance of RNA at OD26o = 4(^g/ml RNA. About 0.5 to 5μg of total T. suis LI RNA will be used to reverse transcribe messenger RNA (mRNA) into cDNA using Superscript III Reverse Transcriptase (Invitrogen), 0.5 μg Oligo dT primer and lOmM dNTPs. The synthesized cDNA will be used as template for PCR reactions to amplify respective protein sequences.
[0075] PCR amplified cDNA of the respective proteins will be cloned into pGEM-T Easy vector (Promega). Subsequently, for expression in E. coli the cDNA will be subcloned into pET 28 (Merck Millipore) with a 6-His tag at the N- and C-terminal end of the sequence. Alternatively, pQE 30 (Quiagen) will be used for only N-terminal 6-His tag expression, depending on keeping or eliminating the stop codon within the cloning strategy. Recombinant proteins will be purified by affinity chromatography using a Ni-NTA column and dialysed against PBS. Removal of endotoxin contaminations will be done by EndoTrap columns (Profos) and final endotoxin concentrations will be detected by Limulus amoebocyte lysate test (Cambrex).
Characterization of LI Ts E/S
[0076] The first larval stages (LI) of Trichuris suis is likely to be the most clinical relevant stage of the life cycle in the context of therapeutic application in humans as it has been shown that the development to sexually mature adult stages is absent in TSO treated humans and efficacious treatment requires repeat applications of high doses of embryonated TSO suggesting that helminth establishment is transient in the human host. As such, the proteins of excretory/secretory (E/S) products derived from the LI T. suis in culture supernatants were determined. T. suis eggs were hatched, washed thoroughly and cultured in vitro for 10-14 days and supernatants were harvested every two days (Figure 1A). Example 2; 71 suis Excretory/Secretory Proteins Have Immunomodulatory Properties in vitro
[0077] In order to ascertain the immunomodulatory potential of larval Ts E/S, a well characterized antigen-specific in vitro assay was performed.
[0078] Whole splenocytes from TcR restricted DO 11.10 mice were cultured for two days with the cognate antigen Ovalbumin (OVA) in the presence or absence of differing amounts of Ts E/S, or heat inactivated Ts E/S supernatant (HI). DO 1 1.10 splenocytes cultured in the presence of OVA showed a significant proliferation as determined by H3 Thymidine incorporation, which was not significantly altered in the presence of Ts E/S (Fig. 2A). Similarly, OVA cultured splenocytes produced significant levels of both Thl -associated IFN- γ and Th2-associated IL-4 during culture in comparison to cells cultured in the absence of antigen (Fig 2B). In contrast treatment of OVA cultured splenocytes with Ts E/S showed a highly significant suppression of both IFN-γ and IL-4 production that was partially dose dependent and that could be reversed following heat inactivation of Ts E/S (HI) (Fig 2B).
[0079] To further determine the nature of the modulatory activity of Ts E/S, an assessment of the ability of Ts E/S to alter dendritic cell (DC) activation by TLR ligands in vitro was performed. DCs stimulated with the TLR-9 agonist CpG upregulated expression of the co- stimulatory molecule CD86 and MHC class II in comparison to unstimulated controls and produced high levels of IL-6 and IL-12/23p40 (Fig 2C). In contrast, DCs pre-treated for 4 hours with Ts E/S prior to CpG stimulation failed to upregulate expression of CD86 and MHC class II and had significantly suppressed production of IL-6, but not IL-12/23p40 (Fig 2C). Conversely, Ts E/S was unable to significantly modulate DC maturation in response to the TLR-4 agonist LPS, although a mild decrease in IL-6 levels was consistently seen (Fig 2D).
[0080] For all cytokine and antibody ELISA experiments herein, IFN-γ, IL-4, IL-5, IL-6, IL-10, IL-12/IL-23p40, IL-13 and IL-17A were determined using Cytometric Bead Assay (CBA, BD Biosciences) as per manufacturer's instructions or commercially available ELISA antibody pairs (BD Biosciences, eBioscience) following manufacturer's instructions. For detection of antigen-specific antibodies ELISA plates were coated with either 50μg/ml OVA or 50 μΐ Ts E/S in bicarbonate buffer at 4°C overnight along with serial dilutions of recombinant IgM, IgG, or IgE (SantCruz Biotech). Plates were blocked with 3% BSA for one hour, washed in PBS 0.05% Tween and incubated for 2 hours with murine serum at multiple dilutions. Plate-bound antibody was then detected by probing with HRP-conjugated anti-mouse IgM, IgGl, or IgE secondary antibody and developed with TMB substrate (R&D Systems).
[0081] For all flow cytometry experiments herein, single cell suspensions were surface stained with fluorescent conjugated antibodies against CD1 lc, CD86, MHCII, CD3, CD4 and CD25 (eBioscience). Intracellular stains for FoxP3, IL-17A or isotype controls were performed following a fixation and permeabilisation protocol (eBioscience). Samples were acquired using LSR II (BD Biosciences) and analysed using FlowJo software (Treestar).
Example 3; 71 suis Excretory/Secretory Proteins Have Immunomodulatory Properties in vivo
Ts E/S suppresses the onset of OVA-speciflc allergic airway inflammation
[0082] To confirm whether Ts E/S has therapeutic effects and/or is sufficient to modulate disease in a characterized murine model of Th2 driven allergic airway inflammation, female C57BL/6 mice and DO 11.10 transgenic mice were purchased from Charles River GmbH and Female BALB/c were purchased from Harlan Olac Ltd. Allergic airway hyperreactivity was induced as described in Schn511er et al. J. Immunol. 2008). Briefly, 8 week old female BALB/c mice were sensitized three times (day 0, 14 and 21) via intraperitoneal injection of 20μg OVA (grade VI, Sigma- Aldrich) or an equal volume of PBS (control) emulsified in 2mg of Alum (Pierce). For Ts E/S treatment mice received ΙΟΟμΙ concentrated Ts E/S supernatant i.p. on day 0, 7, 14 and 21 days post-sensitization. In some experiments mice received 0.5mg anti-IL-lOR (clone 1B1, a kind gift from the DRFZ, Berlin) i.p. or rat IgG (control) on day 21, 23 and 26 post-sensitization. All mice were subsequently challenged via intranasal administration of 50μg OVA in sterile PBS and sacrificed two days later. Airway hyperreactivity was assessed on day 30 via whole-body plethysomography in unrestrained mice in response to increasing doses of metacholine (MCh, Sigma-Aldrich) as described previously (Schn511er et al. 2008, Hamelmann et al. 1997). All mice were maintained in the animal house of the University in accordance with the national animal protection guidelines and approved by the German animal ethics committee. [0083] Lymphocytes from the lung-draining peribroncheolar lymph node (PBLN) and spleen were isolated via manual disruption through a 70 μιη nylon mesh and cultured in RPMI 1640 media containing 10% fetal calf serum (FCS), 20mM L-glutamine, 100 U/ml penicillin and 100 μg/ml streptomycin. Cells were plated at lxlO6 cells/ml in a 96 well plate and restimulated with 50μg/ml Ovalbumin, 20μ1 TsE/S or ^g aCD3 and aCD28 mAb for 48 hours at 37 °C/5% CO2 and supernatants assessed for cytokine production via ELISA. Intracellular cytokine staining was performed by stimulating 5xl06 cells/well with ^g/ml PMA and ^g/ml Ionomycin for 30 minutes followed by the addition of Brefeldin A (5μg/ml) (Sigma Aldrich) and a further 3 hour incubation. Cellular proliferation was analyzed via a further 20-hour culture in fresh media containing 1 μθί methyl- [3H] -thymidine (Amersham Pharmacia) and subsequent detection using a β-counter (Perkin-Elmer). De novo regulatory T cell induction assays were performed as previously described by Grainger et al. 2010.
[0084] Lung tissue was degassed and fixed with 3.7% paraformaldehyde and embedded in paraffin. 2μιη sections were cut, deparaffinised and stained with hematoxylin and eosin (H&E) or Periodic Acid Schiff s reagent to visualize goblet cells. In some experiments sections of lung were stained with an antibody against RELM-a (R&D systems) to visualize alternatively activated macrophages. Goblet cells and RELM-a+ cells were quantified by counting number of cells in 10 HPF (400x magnification). Broncheolar lavage fluid (BAL) cells were differentiated via centrifugation onto frosted slides and differentiated via H&E staining. A minimum of 200 lymphocytes were counted per slide and the percentage of each cell type used to calculate total BAL cell numbers.
[0085] All experiments described herein were performed with 5 animals per group, unless indicated otherwise and expressed as mean ± SEM. Statistical analysis was performed using GraphPad Prism software using one way ANOVA or two tailed Mann- Whitney test for other analyses. p<0.05 (*), p<0.01 (**) and p<0.001 (***) values were considered to be statistically significant.
[0086] Mice that were sensitized and challenged intranasally with ovalbumin (OVA) developed pronounced airway hyperreactivity (AHR) were compared to mice that were sensitised with PBS vehicle only prior to OVA challenge (Fig 3A). In contrast, mice that were sensitized and challenged with OVA, but which received Ts E/S treatment i.p. during the sensitization period (Ts E/S) had significantly reduced airway hyperreactivity (Fig 3A; area under the curve analysis - PBS 0.13; OVA 5.8; Ts E/S 1.6). In line with a reduced airway hyperreactivity the local inflammatory infiltrate in the bronchoalveolar lavage fluid (BAL) was found to be ameliorated in Ts E/S treated mice (Fig 3B). In comparison to disease control mice, Ts E/S treated mice had significantly reduced total cell numbers in the BAL, as well as a complete reduction in lymphocyte and eosinophil infiltration (Fig 3B). Furthermore, whereas the Th2 cytokines IL-4, IL-5, IL-10 and IL-13 were found to be elevated in the BAL of disease control mice, IL-4, IL-5 and IL-13 (but not IL-10) were suppressed in the Ts E/S treated group (Fig 3C). In contrast, no significant effect of Ts E/S treatment was detected on the systemic OVA-specific response, assessed via restimulation of splenocytes (Fig 3D), suggesting the effects of Ts E/S were restricted to the local inflammatory response in the lung. In addition to reduced local Th2 responses and cellular infiltration into the lung, Ts E/S treated mice exhibited significantly fewer mucus secreting bronchiolar goblet cells as well as reduced numbers of RELM-a+ alternatively activated macrophages in the lung tissue in comparison to OVA control mice (Fig 3E). Furthermore, Ts E/S treatment resulted in significant reductions in OVA-specific IgM and IgE levels (Fig 3F), and a trend towards reduced OVA-specific IgGl (Fig 3F).
Ts E/S treatment elicits a Ts E/S-speciflc Th2 response and antibody production
[0087] Ts E/S treatment during the priming phase of OVA-specific airway inflammation resulted in a significant reduction in disease-associated inflammation, local cytokine production and OVA-specific antibody production. However, nematode-derived proteins also elicited potent immune responses. Restimulation of splenocytes with Ts E/S led to the antigen-specific production of IL-4, IL-5, and IL-13 and a significant induction of IL-10 in Ts E/S treated mice (Fig 4A), whereas little cytokine production was detected following restimulation of PBS and OVA control groups that had not received Ts E/S treatment (Fig 4A). In line with a Th2 skewed immune response to Ts E/S we were unable to detect IFN-γ or IL-17A in Ts E/S restimulated cultures (Fig 4A). Furthermore, Ts E/S treatment elicited significant titres of Ts E/S-specific IgM, IgGl and IgE, which could not be detected in control groups (Fig 4B). These results suggest that in addition to modulating responses to bystander antigens (i.e. OVA), Ts E/S treatment also elicits a host Th2 response and production of antibodies directed against these helminth-derived products.
Ts E/S suppression of airway inflammation is partially mediated through IL-10
[0088] Immunomodulatory helminth E/S products have previously been shown to ameliorate inflammation via the induction of regulatory immune cell types, particularly CD4+CD25+FoxP3+ regulatory T cells (Treg). In particular, E/S products derived from the murine helminth Heligmosomoides polygyrus bakeri (Hp) have been reported to contain TGF-P-like activity and to directly induce de novo Treg differentiation in vitro and in vivo (Grainger et al. 2010). In order to determine whether Ts E/S has a similar potential to induce Treg differentiation whole splenocytes with Ts E/S or Hp E/S were cultured in the presence or absence of the potent T cell stimulating mitogen, ConA (Fig 5A). In contrast to Hp E/S, Ts E/S was unable to induce FoxP3 expression in stimulated and sorted CD4+Foxp3- T cells in vitro (Fig 5A). Furthermore, suppression of allergic airway inflammation was not associated with increased frequencies of Tregs in the spleen, lung draining lymph node (PBLN) or in the lung parenchyma (Fig 5B). Nematodes commonly elicit a so-called "modified Th2 response" in their host, which is associated with the production of high levels of IL-10 leading to the persistence of the nematode infection, but also to suppression of bystander immune responses to unrelated antigens. Although, no significant increases in local production of IL-10 in the lung or systemic OVA-specific IL-10 production was detected in Ts E/S treated mice in the model of airway inflammation (Fig 3), a significant IL- 10 production in response to Ts E/S itself was not detected (Fig 4). Thus, to further interrogate the mechanism through which Ts E/S suppressed allergic airway inflammation the immunoregulatory cytokine, IL-10, was inhibited via administration of anti-IL-10 receptor (alL-lOR) neutralizing antibody (Fig 5D). Neutralization of IL-10R significantly ablated the ability of Ts E/S to suppress inflammatory infiltration into the BAL - as indicated by a significant increase in total cell numbers, lymphocyte and eosinophil numbers to levels comparable with OVA disease controls (Fig 5E). Levels of IL-13 in the BAL were also dramatically increased in the BAL in Ts E/S treated mice administered with alL-lOR antibody when compared to Ts E/S treated mice, although this did not reach statistical significance (Fig 5E). In contrast blocking IL-10R had little to no effect on Ts E/S driven suppression of IL-5 in the BAL (Fig 5E), and also failed to restore OVA-specific antibody titres to control levels (Fig 5F). This data suggests Ts E/S induction of IL-10 is at least partially responsible for the suppression of local airway inflammation, although IL-10 independent mechanisms may also contribute to suppression, particularly in regard to antibody production.
Example 4; Recombinant Expression and Functional Testing of SEQ ID NO; 1
[0089] Snap frozen T. suis worms were used for isolation of RNA. For that purpose, worm material was ground in liquid nitrogen before homogenization with the QIAshredder column (Qiagen). RNA was isolated from homogenized lysate using Innu PREP RNA Mini Kit (Analytik Jena AG) according to Manufacturer's instruction. Concentration of total RNA was measured using NanoDrop Spectrophotometer (PeqLab). 1 μg of total T. suis RNA was used to reverse transcribe messenger RNA (mRNA) into cDNA with the Transcriptor First Strand cDNA Synthesis Kit (Roche), containing reverse transcriptase, oligo dT primer and dNTPs. The herein synthesized cDNA was used as template for PCR reactions to amplify SEQ ID NO: 1 nucleotide sequence (Fig. 6).
[0090] PCR amplified cDNA of SEQ ID NO: 1 was cloned into pGEM-T Easy vector (Promega) and transformed into competent E. coli cells (JM109). For protein expression, SEQ ID NO: 1 cDNA was subcloned into a eukaryotic expression system of of Leishmania tarentolae (LEXSY, Jena Bioscience), a unicellular-flagellated protozoa. Genome integrated cloning, a Leishmanian secretion system and a C-terminal His-tag enabled recombinant polypeptide purification of SEQ ID NO: 1 from liquid culture media (Fig. 7).
[0091] Recombinant polypeptide SEQ ID NO: 1 was purified by affinity chromatography using HisTrap excel columns (GE Healthcare) prepacked with Ni Sepharose excel affinity media for capture and purification of histidine-tagged proteins secreted into eukaryotic cell culture supernatants (Fig. 8).
[0092] Recombinant polypeptide SEQ ID NO: 1 was dialysed against PBS and endotoxin concentrations, detected by Limulus amoebocyte lysate test (Cambrex), were consistently lower than 0.6pg LPS^g protein.
[0093] In order to ascertain the immunomodulatory potential of recombinant polypeptide SEQ ID NO: 1, a well characterized antigen-specific in vitro assay was performed. Sorted and CFSE-labeled CD4+ T cells from TcR restricted DO 1 1.10 mice were cultured for 5 days with cognate antigen Ovalbumin pulsed bone-marrow derived dendritic cells (DCs) in the presence or absence of recombinant polypeptide SEQ ID NO: 1 or heat inactivated (hi) recombinant polypeptide SEQ ID NO: 1. Preliminary data showed that in the presence of recombinant polypeptide SEQ ID NO: 1, CD4+ T cell proliferation, as determined by CFSE- dilution, was significantly suppressed in comparison to cells cultured in the absence of recombinant polypeptide SEQ ID NO: 1 (Fig. 9).
[0094] In addition, treatment of CD4+ T cell - DC co-cultures with recombinant polypeptide SEQ ID NO: 1 showed a significant increase of the anti-inflammatory cytokine IL-10, that could be reversed following heat inactivation of SEQ ID NO: 1 (Fig. 10). In contrast, IL-12 and IFNg secretion were not affected.
[0095] The foregoing description of illustrative embodiments has been presented for purposes of illustration and of description. It is not intended to be exhaustive or limiting with respect to the precise form disclosed, and modifications and variations are possible in light of the above teachings or may be acquired from practice of the disclosed embodiments. It is intended that the scope of the invention be defined by the claims appended hereto and their equivalents.
SEQ ID NO: 1
>Locus_7131_aa_full length
MHTFLAVAFLALSELCAVHPWDGSPYTLEQLATRPRPRVSFPWFVLIKTPRNKCVGTILR ADDSHKTPLVLAPSECLLTADISVNDTSQGQNETRLEIPSMVEIVGSNFSSVAKNLYLSV KTGYPVVFIKLEEPYRGPTVLVDRAVYFTGNVTKCILVRFSSSETQKRQLLSNDDSPVRF REEEEICRNSTASETGILDGQGLTCKAGDGLWTFYGFCGEEGFESIYVPVQPLIKLITYA AREMELYKKDEVLKGRKISYKHSFLHGQEVWTVTRGRQTKIYYREPVL
SEQ ID NO: 2
>Locus_l 063_aa_ full length
LTMKMNPCMLISILALLVLRAASECVTHKDCRSAQSLCFDGYCLLATAVPGKCTTSADCR TQLSVMQNAGRGCRN ICYEI ITNKLCTGHVSCDDAQVCIRNHC PS A SAKCHVGALC AAGQRCLGGICYEPLEPSRVLETDNSR
SEQ ID NO: 3
>Locus_1815_aa_full length
XTPMQWFLITALVIVAFSDARFHFRNKYKVHQKGKQPAPAAEEWSYSDDDDSVCGYINIT KTEKEMLHQRKKLPHTLPFIAYVVKRHRHNYQPVCVATILPDADDESSSIVLTLTTCIES PQLLDHYKIFTGRKLPVPLNKTRKMYDIEEVLVDKKSPNLQYANAMVVKLSGSIPIDAKR QPICLPKPNAEMPKKSECILSVVRTDGRQQPYLIEVEPKSNKDCKKMHGESIQIDNKMEI CGLDYRDGSFISRGGSLICEGEGGRWIQYGIYTHASHISDRMADRRSVSRIQKKGRPSVY INVANFTKLMQTSQNLERLSRINLMKLRXSKMYAKNDVRSYVNTPQTFYT
SEQ ID NO: 4
>Tsui7117105_aa
MDRLPLVSDPKYVHKNSARLRSARRPKSNRRFGKFPAKRSHHQCADIAAS NSAAAEGTTSRIVILSTAKWTYAMITAKSVRCQNCQHLGCGYAFINEKLK QCFTRPDTPSYVRLGYRKMFESIPKKHCIEKDGMCKCCCGDYEPNEAGTE CVKPPAHDCPAFGPPSEWSECLGSQ
SEQ ID NO: 5
>Locus_7131_aa_mature
WDGSPYTLEQLATRPRPRVSFPWFVLIKTPRNKCVGTILRADDSHKTPLVLAPSECLLTA DISVNDTSQGQNETRLEIPSMVEIVGSNFSSVAKNLYLSVKTGYPVVFIKLEEPYRGPTV LVDRAVYFTGNVTKCILVRFSSSETQKRQLLSNDDSPVRFREEEEICRNSTASETGILDG QGLTCKAGDGLWTFYGFCGEEGFESIYVPVQPLIKLITYAAREMELYKKDEVLKGRKISY KHSFLHGQEVWTVTRGRQTKIYYREPVL
SEQ ID NO: 6
>Locus_l 063_aa_mature
ECVTHKDCRSAQSLCFDGYCLLATAVPGKCTTSADCRTQLSVMQNAGRGCRNNICYEIITNK LC GHVSCDDAQVCIRNHC PS A SAKCHVGALCAAGQRCLGGICYEPLEPSRVLETDNSR
SEQ ID NO: 7
>Locus_1815_aa_mature
VHQKGKQPAPAAEEWSYSDDDDSVCGYINITKTEKEMLHQRKKLPHTLPFIAYVVKRHRHNY QPVCVATILPDADDESSSIVLTLTTCIESPQLLDHYKIFTGRKLPVPLNKTRKMYDIEEVLV DKKPNLQYANAMVVKLSGSIPIDAKRQPICLPKPNAEMPKKSECILSVVRTDGRQQPYLIEV EPKSNKDCKKMHGESIQIDNKMEICGLDYRDGSFISRGGSLICEGEGGRWIQYGIYTHASHI SDRMADRSVSRIQKKGRPSVYINVANFTKLMQTSQNLERLSRINLMKLRXSKMYAKNDVRSY VNTPQTFYT
SEQ ID NO: 8
ATGCATACCTTTCTGGCGGTGGCGTTTCTGGCGCTGAGCGAACTGTGCGCGGTGCATCCG TGGGATGGCAGCCCGTATACCCTGGAACAGCTGGCGACCCGCCCGCGCCCGCGCGTGAGC TTTCCGTGGTTTGTGCTGATTAAAACCCCGCGCAACAAATGCGTGGGCACCATTCTGCGC GCGGATGATAGCCATAAAACCCCGCTGGTGCTGGCGCCGAGCGAATGCCTGCTGACCGCG GATATTAGCGTGAACGATACCAGCCAGGGCCAGAACGAAACCCGCCTGGAAATTCCGAGC ATGGTGGAAATTGTGGGCAGCAACTTTAGCAGCGTGGCGAAAAACCTGTATCTGAGCGTG AAAACCGGCTATCCGGTGGTGTTTATTAAACTGGAAGAACCGTATCGCGGCCCGACCGTG CTGGTGGATCGCGCGGTGTATTTTACCGGCAACGTGACCAAATGCATTCTGGTGCGCTTT AGCAGCAGCGAAACCCAGAAACGCCAGCTGCTGAGCAACGATGATAGCCCGGTGCGCTTT CGCGAAGAAGAAGAAATTTGCCGCAACAGCACCGCGAGCGAAACCGGCATTCTGGATGGC CAGGGCCTGACCTGCAAAGCGGGCGATGGCCTGTGGACCTTTTATGGCTTTTGCGGCGAA GAAGGCTTTGAAAGCATTTATGTGCCGGTGCAGCCGCTGATTAAACTGATTACCTATGCG GCGCGCGAAATGGAACTGTATAAAAAAGATGAAGTGCTGAAAGGCCGCAAAATTAGCTAT AAACATAGCTTTCTGCATGGCCAGGAAGTGTGGACCGTGACCCGCGGCCGCCAGACCAAA ATTTATTATCGCGAACCGGTGCTG
SEQ ID NO: 9
>Locus_7131_nucleotide_full length
ATGCACACGTTCTTGGCCGTTGCTTTTCTTGCTTTAAGCGAATTATGTGCAGTGCACCCA TGGGATGGTTCACCATATACACTTGAGCAGCTGGCAACTAGGCCCAGACCTCGCGTCAGT TTTCCCTGGTTTGTACTGATTAAAACACCGAGGAACAAATGTGTTGGAACCATTCTACGA GCAGACGACTCCCACAAGACTCCACTAGTGTTAGCGCCATCAGAATGCTTGCTAACGGCG GATATCAGTGTTAATGACACGAGTCAGGGACAGAATGAAACGCGTCTGGAAATTCCCAGT ATGGTGGAAATAGTTGGCAGCAATTTTTCGTCTGTGGCAAAGAACCTGTATTTATCTGTG AAAACCGGCTATCCAGTGGTTTTTATTAAACTGGAAGAACCCTACAGAGGTCCAACTGTC TTGGTAGACCGTGCTGTCTACTTCACTGGCAACGTGACGAAGTGCATACTGGTGCGCTTC TCTTCTTCAGAAACCCAGAAACGGCAACTGCTGAGCAACGATGATTCTCCTGTAAGATTC CGGGAGGAAGAGGAGATTTGTAGAAACAGTACGGCGAGTGAAACAGGCATACTTGACGGA CAAGGCTTGACTTGTAAGGCTGGAGACGGTCTGTGGACTTTCTACGGCTTTTGCGGTGAA GAAGGGTTTGAGAGCATATACGTGCCAGTCCAGCCGCTTATCAAACTGATTACATACGCA GCTAGAGAGATGGAGTTGTACAAAAAGGATGAAGTTCTCAAAGGGCGAAAAATTTCTTAT AAACACTCTTTTCTTCATGGACAAGAAGTATGGACCGTCACGAGGGGTCGCCAAACGAAA ATTTATTACAGGGAACCTGTTTTG
SEQ ID NO: 10
ATGCATACTTTTCTTGCTGTTGCTTTTCTTGCTCTTTCTGAACTTTGTGCTGTTCATCCT TGGGATGGTTCTCCTTATACTCTTGAACAACTTGCTACTCGTCCTCGTCCTCGTGTTTCT TTTCCTTGGTTTGTTCTTATTAAAACTCCTCGTAATAAATGTGTTGGTACTATTCTTCGT GCTGATGATTCTCATAAAACTCCTCTTGTTCTTGCTCCTTCTGAATGTCTTCTTACTGCT GATATTTCTGTTAATGATACTTCTCAAGGTCAAAATGAAACTCGTCTTGAAATTCCTTCT ATGGTTGAAATTGTTGGTTCTAATTTTTCTTCTGTTGCTAAAAATCTTTATCTTTCTGTT AAAACTGGTTATCCTGTTGTTTTTATTAAACTTGAAGAACCTTATCGTGGTCCTACTGTT CTTGTTGATCGTGCTGTTTATTTTACTGGTAATGTTACTAAATGTATTCTTGTTCGTTTT TCTTCTTCTGAAACTCAAAAACGTCAACTTCTTTCTAATGATGATTCTCCTGTTCGTTTT CGTGAAGAAGAAGAAATTTGTCGTAATTCTACTGCTTCTGAAACTGGTATTCTTGATGGT CAAGGTCTTACTTGTAAAGCTGGTGATGGTCTTTGGACTTTTTATGGTTTTTGTGGTGAA GAAGGTTTTGAATCTATTTATGTTCCTGTTCAACCTCTTATTAAACTTATTACTTATGCT GCACGAGAGATGGAACTCTATAAAAAGGATGAAGTTCTTAAAGGTCGTAAAATTTCTTAT AAGCATTCTTTTCTTCATGGACAAGAAGTTTGGACTGTAACTCGTGGCAGTCAAACTAAA ATATATTATCGTGAACCTGTTCTA
SEQ ID NO: 11
CTTACTATGAAAATGAATCCTTGTATGCTTATTTCTATTCTTGCTCTTCTTGTTCTTCGT GCTGCTTCTGAATGTGTTACTCATAAAGATTGTCGTTCTGCTCAATCTCTTTGTTTTGAT GGTTATTGTCTTCTTGCTACTGCTGTTCCTGGTAAATGTACTACTTCTGCTGATTGTCGT ACTCAACTTTCTGTTATGCAAAATGCTGGTCGTGGTTGTCGTAATAATATTTGTTATGAA ATTATTACTAATAAACTTTGTACTGGTCATGTTTCTTGTGATGATGCTCAAGTTTGTATT CGTAATCATTGTGTTCCTTCTGTTGCTACTTCTGCTAAATGTCATGTTGGTGCTCTTTGT GCTGCTGGTCAACGTTGTCTTGGTGGTATTTGTTATGAACCTCTTGAACCTTCTCGTGTT CTTGAAACTGATAATTCTCGT
SEQ ID NO: 12
>Locus_l 063_nucleotide_full length
TTAACAATGAAGATGAATCCATGCATGCTTATTTCCATTCTGGCCCTCCTAGTACTAAGA GCCGCAAGCGAATGTGTGACGCATAAAGACTGCCGTTCCGCACAATCACTCTGTTTCGAC GGATATTGCTTACTAGCAACTGCAGTGCCTGGGAAATGCACCACGTCTGCAGACTGCAGA ACGCAACTTTCAGTCATGCAAAACGCCGGAAGAGGGTGTAGAAACAACATATGCTACGAA ATTATAACTAACAAGTTGTGTACTGGTCACGTGAGTTGCGATGACGCGCAAGTGTGCATT AGAAACCATTGCGTTCCTTCAGTTGCCACCTCCGCGAAATGCCATGTCGGGGCACTGTGC GCCGCTGGCCAGCGATGTTTGGGCGGAATTTGCTATGAGCCGCTTGAACCTTCGCGGGTA TTAGAAACCGACAATTCTCGT
SEQ ID NO: 13
TTGACGATGAAAATGAACCCCTGTATGCTTATTTCTATTCTTGCTCTTCTTGTTCTTCGT GCTGCTTCTGAATGTGTTACTCATAAAGATTGTCGTTCTGCTCAATCTCTTTGTTTTGAT GGTTATTGTCTTCTTGCTACTGCTGTTCCTGGTAAATGTACTACTTCTGCTGATTGTCGT ACTCAACTTTCTGTTATGCAAAATGCTGGTCGTGGTTGTCGTAATAATATTTGTTATGAA ATTATTACTAATAAACTTTGTACTGGTCATGTTTCTTGTGATGATGCTCAAGTTTGTATT CGTAATCATTGTGTTCCTTCTGTTGCTACTTCTGCTAAATGTCATGTTGGTGCTCTTTGT GCGGCAGGTCAACGTTGTCTTGGTGGTATTTGTTATGAACCTCTTGAACCTTCTCGTGTT TTAGAAACTGATAACAGCAGG
SEQ ID NO: 14
TTTACTCCTATGCAATGGTTTCTTATTACTGCTCTTGTTATTGTTGCTTTTTCTGATGCT CGTTTTCATTTTCGTAATAAATATAAAGTTCATCAAAAAGGTAAACAACCTGCTCCTGCT GCTGAAGAATGGTCTTATTCTGATGATGATGATTCTGTTTGTGGTTATATTAATATTACT AAAACTGAAAAAGAAATGCTTCATCAACGTAAAAAACTTCCTCATACTCTTCCTTTTATT GCTTATGTTGTTAAACGTCATCGTCATAATTATCAACCTGTTTGTGTTGCTACTATTCTT CCTGATGCTGATGATGAATCTTCTTCTATTGTTCTTACTCTTACTACTTGTATTGAATCT CCTCAACTTCTTGATCATTATAAAATTTTTACTGGTCGTAAACTTCCTGTTCCTCTTAAT AAAACTCGTAAAATGTATGATATTGAAGAAGTTCTTGTTGATAAAAAATCTCCTAATCTT CAATATGCTAATGCTATGGTTGTTAAACTTTCTGGTTCTATTCCTATTGATGCTAAACGT CAACCTATTTGTCTTCCTAAACCTAATGCTGAAATGCCTAAAAAATCTGAATGTATTCTT TCTGTTGTTCGTACTGATGGTCGTCAACAACCTTATCTTATTGAAGTTGAACCTAAATCT AATAAAGATTGTAAAAAAATGCATGGTGAATCTATTCAAATTGATAATAAAATGGAAATT TGTGGTCTTGATTATCGTGATGGTTCTTTTATTTCTCGTGGTGGTTCTCTTATTTGTGAA GGTGAAGGTGGTCGTTGGATTCAATATGGTATTTATACTCATGCTTCTCATATTTCTGAT CGTATGGCTGATCGTCGTTCTGTTTCTCGTATTCAAAAAAAAGGTCGTCCTTCTGTTTAT ATTAATGTTGCTAATTTTACTAAACTTATGCAAACTTCTCAAAATCTTGAACGTCTTTCT CGTATTAATCTTATGAAACTTCGTTTTTCTAAAATGTATGCTAAAAATGATGTTCGTTCT TATGTTAATACTCCTCAAACTTTTTATACT
SEQ ID NO: 15
>Locus_l 815_nucleotide_full length
XTGACTCCAATGCAATGGTTTCTCATTACGGCGCTCGTCATTGTCGCTTTTTCAGATGCG AGATTTCACTTCAGAAACAAATATAAAGTACATCAAAAGGGCAAGCAACCGGCGCCTGCA GCAGAAGAATGGTCATATAGTGATGACGATGACAGCGTTTGCGGATACATAAATATAACA AAAACAGAGAAAGAGATGCTTCATCAACGCAAGAAACTGCCCCATACGCTCCCATTTATT GCCTACGTGGTGAAGAGACATCGACATAACTACCAACCGGTTTGTGTGGCTACAATTTTG CCAGATGCGGATGACGAGTCGAGCAGTATTGTGCTCACGCTTACGACATGCATCGAATCG CCACAACTGTTGGACCACTATAAAATTTTCACCGGAAGAAAACTTCCGGTGCCACTGAAT AAAACAAGGAAGATGTACGATATTGAAGAGGTTCTGGTCGATAAAAAGAGCCCAAATTTA CAGTATGCTAATGCAATGGTTGTGAAATTGAGCGGTTCGATACCAATTGATGCAAAGCGT CAGCCAATCTGCCTACCAAAACCAAACGCGGAAATGCCTAAGAAGTCTGAATGTATTCTT TCAGTTGTTCGAACCGATGGCAGACAGCAACCCTATCTGATTGAGGTTGAACCCAAGTCA AACAAGGACTGTAAGAAGATGCACGGAGAGAGCATCCAAATTGACAATAAAATGGAAATA TGCGGGCTCGACTATCGAGATGGATCATTTATCTCCAGAGGTGGATCACTGATTTGCGAA GGCGAAGGGGGCAGGTGGATACAGTACGGCATTTACACGCATGCTTCCCACATTTCTGAC AGAATGGCAGACAGACGGAGCGTATCCAGAATTCAAAAGAAAGGACGCCCCAGCGTGTAC ATCAATGTTGCCAACTTCACAAAGCTAATGCAAACGTCACAAAACCTGGAAAGACTATCA CGCATAAACTTAATGAAACTTCGCTGATCGAAAATGTACGCAAAAAATGATGTCCGTTCA TACGTGAATACTCCGCAAACTTTCTACACT
SEQ ID NO: 16
ATGACACCGATGCAGTGGTTTCTTATTACAGCACTAGTAATTGTTGCTTTTTCTGATGCT CGTTTTCATTTTCGTAATAAATATAAAGTTCATCAAAAAGGTAAACAACCTGCTCCTGCT GCTGAAGAATGGTCTTATTCTGATGATGATGATTCTGTTTGTGGTTATATTAATATTACT AAAACTGAAAAAGAAATGCTTCATCAACGTAAAAAACTTCCTCATACTCTTCCTTTTATT GCTTATGTTGTTAAACGTCATCGTCATAATTATCAACCTGTTTGTGTTGCTACTATTCTT CCTGATGCTGATGATGAAAGCTCTTCTATTGTACTTACTCTTACTACTTGTATTGAATCT CCTCAACTTCTTGATCATTATAAAATTTTTACTGGTCGTAAACTTCCTGTTCCTCTTAAT AAAACTCGTAAAATGTATGATATTGAAGAAGTTCTTGTTGATAAAAAATCTCCTAATCTT CAATATGCTAATGCTATGGTTGTTAAACTTTCTGGTTCTATTCCTATTGATGCTAAACGT CAACCTATTTGTCTTCCTAAACCTAATGCTGAAATGCCTAAAAAATCTGAATGTATTCTT TCTGTTGTCCGTACCGATGGTCGTCAACAACCTTATCTTATTGAAGTTGAACCTAAATCT AATAAAGATTGTAAAAAAATGCATGGTGAATCTATTCAAATTGATAATAAAATGGAAATT TGTGGTCTTGATTATCGTGATGGTTCTTTTATTTCTCGTGGTGGTTCTCTTATTTGTGAA GGTGAGGGTGGTCGTTGGATCCAATATGGTATATATACTCATGCTTCTCATATTTCTGAT CGTATGGCTGATCGTCGTTCTGTTTCTCGTATTCAAAAAAAAGGTCGTCCTTCTGTTTAT ATTAATGTTGCTAATTTTACTAAACTTATGCAAACTTCTCAAAATCTTGAACGTCTTTCT CGTATTAATCTTATGAAACTTCGTTTTTCTAAAATGTATGCTAAAAATGATGTTCGTTCT TATGTAAATACTCCTCAAACGTTTTACACA
SEQ ID NO: 17
>Sequence_ID_Tsui7117105_nucleotide
ATGGATCGTCTTCCTCTTGTTTCTGATCCTAAATATGTTCATAAAAATTCTGCTCGTCTT CGTTCTGCTCGTCGTCCTAAATCTAATCGTCGTTTTGGTAAATTTCCTGCTAAACGTTCT CATCATCAATGTGCTGATATTGCTGCTTCTAATTCTGCTGCTGCTGAAGGTACTACTTCT CGTATTGTTATTCTTTCTACTGCTAAATGGACTTATGCTATGATTACTGCTAAATCTGTT CGTTGTCAAAATTGTCAACATCTTGGTTGTGGTTATGCTTTTATTAATGAAAAACTTAAA CAATGTTTTACTCGTCCTGATACTCCTTCTTATGTTCGTCTTGGTTATCGTAAAATGTTT GAATCTATTCCTAAAAAACATTGTATTGAAAAAGATGGTATGTGTAAATGTTGTTGTGGT GATTATGAACCTAATGAAGCTGGTACTGAATGTGTTAAACCTCCTGCTCATGATTGTCCT GCTTTTGGTCCTCCTTCTGAATGGTCTGAATGTCTTGGTTCTCAA
SEQ ID NO: 18
T. suis counterpart of the T. muris protein TMC00067_1 >Sequence_ID_Tsui7117105_nucleotide
ATGCATTTCACTCGGAAGCTGCAATAGCCGCACTTTTCCGGTATGTGGAT TCCGGGCGGAGTTGCAACGGGCGGTCGGTAACCGTCCGCGTCCGGATGAA CGTGACAATGATCTTCCACGTGCTTGACAAGATCTTTCATTGGGAACCAA GGCATTCGCTCCATTCGCTTGGTGGTCCAAAGGCGGGGCAATCGTGCGCC GGCGGCTTGACGCATTCAGTGCCTGCCTCGTTCGGCTCGTAGTCTCCGCA GCAACACTTGCACATTCCGTCCTTCTCGATGCAATGCTTTTTCGGGATAC TTTCGAACATTTTTCTATAGCCAAGTCGTACGTAGGATGGCGTATCCGGT CGAGTGAAGCATTGCTTCAATTTTTCGTTTATGAAAGCGTAACCGCATCC CAAATGTTGGCAATTTTGGCAGCGAACAGATTTGGCCGTGATCATGGCAT ACGTCCACTTGGCCGTCGAGAGGATAACAATCCTTGACGTTGTCCCTTCG GCGGCAGCGGAATTTGAAGCTGCAATGTCCGCACATTGGTGGTGGCTTCT CTTCGCCGGGAATTTCCCAAAACGCCGGTTAGATTTCGGTCGGCGTGCAG ACCGCAGGCGTGCGCTATTTTTGTGTACATATTTTGGGTCGGATACCAAA GGCAGTCGGTCCATGGTTTCCAATCTTTCCAAGCTGGACAGTGCTCGTGG TGGTCAGCCAACGAGAAACCAAACGTCAGCAGCAGCAGCGAAACGAACGG CAGTAGCATCGCAGAGATCGGAAGAGCGTCGTGTAGGGAAAGAGTGTAGA TCTCGGTGGTCGCCGTATCA
SEQ ID NO: 19
T. suis counterpart of the T. muris protein TMC00067_1
ATGGACAGACTTCCTCTTGTAAGCGATCCTAAATATGTTCATAAAAATAGCGCTCGTCTT
CGTAGCGCTCGTCGTCCTAAAAGCAATCGTCGTTTTGGTAAATTCCCTGCTAAACGATCT
CATCATCAATGTGCTGATATTGCTGCTAGCAATTCTGCTGCTGCTGAAGGTACTACTTCT
CGTATTGTTATTCTTTCTACTGCTAAATGGACTTATGCTATGATTACTGCTAAATCTGTT
CGTTGTCAAAATTGTCAACATCTTGGTTGTGGTTATGCTTTTATTAATGAAAAACTTAAA
CAATGTTTTACTCGTCCTGATACTCCTTCTTATGTTCGTCTTGGTTATCGTAAAATGTTT
GAAAGCATTCCCAAAAAACATTGTATTGAAAAAGATGGGATGTGTAAATGTTGTTGTGGT
GATTATGAACCTAATGAAGCTGGTACTGAATGTGTTAAACCTCCAGCGCATGATTGTCCA
GCATTTGGTCCTCCTAGCGAATGGTCTGAATGTCTTGGTTCTCAA SEQ ID NO: 20
TGGGATGGCAGCCCGTATACCCTGGAACAGCTGGCGACCCGCCCGCGCCCGCGCGTGAGC TTTCCGTGGTTTGTGCTGATTAAAACCCCGCGCAACAAATGCGTGGGCACCATTCTGCGC GCGGATGATAGCCATAAAACCCCGCTGGTGCTGGCGCCGAGCGAATGCCTGCTGACCGCG GATATTAGCGTGAACGATACCAGCCAGGGCCAGAACGAAACCCGCCTGGAAATTCCGAGC ATGGTGGAAATTGTGGGCAGCAACTTTAGCAGCGTGGCGAAAAACCTGTATCTGAGCGTG AAAACCGGCTATCCGGTGGTGTTTATTAAACTGGAAGAACCGTATCGCGGCCCGACCGTG CTGGTGGATCGCGCGGTGTATTTTACCGGCAACGTGACCAAATGCATTCTGGTGCGCTTT AGCAGCAGCGAAACCCAGAAACGCCAGCTGCTGAGCAACGATGATAGCCCGGTGCGCTTT CGCGAAGAAGAAGAAATTTGCCGCAACAGCACCGCGAGCGAAACCGGCATTCTGGATGGC CAGGGCCTGACCTGCAAAGCGGGCGATGGCCTGTGGACCTTTTATGGCTTTTGCGGCGAA GAAGGCTTTGAAAGCATTTATGTGCCGGTGCAGCCGCTGATTAAACTGATTACCTATGCG GCGCGCGAAATGGAACTGTATAAAAAAGATGAAGTGCTGAAAGGCCGCAAAATTAGCTAT AAACATAGCTTTCTGCATGGCCAGGAAGTGTGGACCGTGACCCGCGGCCGCCAGACCAAA ATTTATTATCGCGAACCGGTGCTG
SEQ ID NO: 21
>Locus_7131_nucleotide_mature
TGGGATGGTTCACCATATACACTTGAGCAGCTGGCAACTAGGCCCAGACCTCGCGTCAGT TTTCCCTGGTTTGTACTGATTAAAACACCGAGGAACAAATGTGTTGGAACCATTCTACGA GCAGACGACTCCCACAAGACTCCACTAGTGTTAGCGCCATCAGAATGCTTGCTAACGGCG GATATCAGTGTTAATGACACGAGTCAGGGACAGAATGAAACGCGTCTGGAAATTCCCAGT ATGGTGGAAATAGTTGGCAGCAATTTTTCGTCTGTGGCAAAGAACCTGTATTTATCTGTG AAAACCGGCTATCCAGTGGTTTTTATTAAACTGGAAGAACCCTACAGAGGTCCAACTGTC TTGGTAGACCGTGCTGTCTACTTCACTGGCAACGTGACGAAGTGCATACTGGTGCGCTTC TCTTCTTCAGAAACCCAGAAACGGCAACTGCTGAGCAACGATGATTCTCCTGTAAGATTC CGGGAGGAAGAGGAGATTTGTAGAAACAGTACGGCGAGTGAAACAGGCATACTTGACGGA CAAGGCTTGACTTGTAAGGCTGGAGACGGTCTGTGGACTTTCTACGGCTTTTGCGGTGAA GAAGGGTTTGAGAGCATATACGTGCCAGTCCAGCCGCTTATCAAACTGATTACATACGCA GCTAGAGAGATGGAGTTGTACAAAAAGGATGAAGTTCTCAAAGGGCGAAAAATTTCTTAT AAACACTCTTTTCTTCATGGACAAGAAGTATGGACCGTCACGAGGGGTCGCCAAACGAAA ATTTATTACAGGGAACCTGTTTTG
SEQ ID NO: 22
TGGGATGGTTCTCCTTATACTCTTGAACAACTTGCTACTCGTCCTCGTCCTCGTGTTTCT TTTCCTTGGTTTGTTCTTATTAAAACTCCTCGTAATAAATGTGTTGGTACTATTCTTCGT GCTGATGATTCTCATAAAACTCCTCTTGTTCTTGCTCCTTCTGAATGTCTTCTTACTGCT GATATTTCTGTTAATGATACTTCTCAAGGTCAAAATGAAACTCGTCTTGAAATTCCTTCT ATGGTTGAAATTGTTGGTTCTAATTTTTCTTCTGTTGCTAAAAATCTTTATCTTTCTGTT AAAACTGGTTATCCTGTTGTTTTTATTAAACTTGAAGAACCTTATCGTGGTCCTACTGTT CTTGTTGATCGTGCTGTTTATTTTACTGGTAATGTTACTAAATGTATTCTTGTTCGTTTT TCTTCTTCTGAAACTCAAAAACGTCAACTTCTTTCTAATGATGATTCTCCTGTTCGTTTT CGTGAAGAAGAAGAAATTTGTCGTAATTCTACTGCTTCTGAAACTGGTATTCTTGATGGT CAAGGTCTTACTTGTAAAGCTGGTGATGGTCTTTGGACTTTTTATGGTTTTTGTGGTGAA GAAGGTTTTGAATCTATTTATGTTCCTGTTCAACCTCTTATTAAACTTATTACTTATGCT GCACGAGAGATGGAACTCTATAAAAAGGATGAAGTTCTTAAAGGTCGTAAAATTTCTTAT AAGCATTCTTTTCTTCATGGACAAGAAGTTTGGACTGTAACTCGTGGCCGCCAAACGAAA ATATATTATCGTGAACCTGTTCTA
SEQ ID NO: 23
GAATGTGTTACTCATAAAGATTGTCGTTCTGCTCAATCTCTTTGTTTTGATGGTTATTGT CTTCTTGCTACTGCTGTTCCTGGTAAATGTACTACTTCTGCTGATTGTCGTACTCAACTT TCTGTTATGCAAAATGCTGGTCGTGGTTGTCGTAATAATATTTGTTATGAAATTATTACT AATAAACTTTGTACTGGTCATGTTTCTTGTGATGATGCTCAAGTTTGTATTCGTAATCAT TGTGTTCCTTCTGTTGCTACTTCTGCTAAATGTCATGTTGGTGCTCTTTGTGCTGCTGGT CAACGTTGTCTTGGTGGTATTTGTTATGAACCTCTTGAACCTTCTCGTGTTCTTGAAACT GATAATTCTCGT
SEQ ID NO: 24
>Locus_l 063_nucleotide_mature
GAATGTGTGACGCATAAAGACTGCCGTTCCGCACAATCACTCTGTTTCGACGGATATTGC TTACTAGCAACTGCAGTGCCTGGGAAATGCACCACGTCTGCAGACTGCAGAACGCAACTT TCAGTCATGCAAAACGCCGGAAGAGGGTGTAGAAACAACATATGCTACGAAATTATAACT AACAAGTTGTGTACTGGTCACGTGAGTTGCGATGACGCGCAAGTGTGCATTAGAAACCAT TGCGTTCCTTCAGTTGCCACCTCCGCGAAATGCCATGTCGGGGCACTGTGCGCCGCTGGC CAGCGATGTTTGGGCGGAATTTGCTATGAGCCGCTTGAACCTTCGCGGGTATTAGAAACC GACAATTCTCGT
SEQ ID NO: 25
GAGTGCGTAACACATAAAGATTGTCGTTCTGCTCAATCGCTGTGCTTTGATGGTTATTGT CTGTTAGCTACTGCTGTTCCTGGTAAATGTACTACTTCTGCTGATTGTCGTACTCAATTG AGCGTTATGCAAAATGCTGGTCGTGGATGTCGTAATAATATTTGTTATGAAATTATTACA AATAAGCTATGTACTGGTCATGTTAGCTGTGATGATGCTCAAGTTTGTATTCGTAATCAC TGTGTACCTAGCGTTGCTACTTCTGCTAAATGTCATGTTGGTGCTCTTTGTGCTGCTGGT CAAAGATGCCTAGGTGGTATTTGTTATGAACCTCTTGAACCTTCTCGTGTTCTTGAAACT GATAACAGCCGG
SEQ ID NO: 26
GTTCATCAAAAAGGTAAACAACCTGCTCCTGCTGCTGAAGAATGGTCTTATTCTGATGAT GATGATTCTGTTTGTGGTTATATTAATATTACTAAAACTGAAAAAGAAATGCTTCATCAA CGTAAAAAACTTCCTCATACTCTTCCTTTTATTGCTTATGTTGTTAAACGTCATCGTCAT AATTATCAACCTGTTTGTGTTGCTACTATTCTTCCTGATGCTGATGATGAATCTTCTTCT ATTGTTCTTACTCTTACTACTTGTATTGAATCTCCTCAACTTCTTGATCATTATAAAATT TTTACTGGTCGTAAACTTCCTGTTCCTCTTAATAAAACTCGTAAAATGTATGATATTGAA GAAGTTCTTGTTGATAAAAAACCTAATCTTCAATATGCTAATGCTATGGTTGTTAAACTT TCTGGTTCTATTCCTATTGATGCTAAACGTCAACCTATTTGTCTTCCTAAACCTAATGCT GAAATGCCTAAAAAATCTGAATGTATTCTTTCTGTTGTTCGTACTGATGGTCGTCAACAA CCTTATCTTATTGAAGTTGAACCTAAATCTAATAAAGATTGTAAAAAAATGCATGGTGAA TCTATTCAAATTGATAATAAAATGGAAATTTGTGGTCTTGATTATCGTGATGGTTCTTTT ATTTCTCGTGGTGGTTCTCTTATTTGTGAAGGTGAAGGTGGTCGTTGGATTCAATATGGT ATTTATACTCATGCTTCTCATATTTCTGATCGTATGGCTGATCGTTCTGTTTCTCGTATT CAAAAAAAAGGTCGTCCTTCTGTTTATATTAATGTTGCTAATTTTACTAAACTTATGCAA ACTTCTCAAAATCTTGAACGTCTTTCTCGTATTAATCTTATGAAACTTCGTTTTTCTAAA ATGTATGCTAAAAATGATGTTCGTTCTTATGTTAATACTCCTCAAACTTTTTATACT SEQ ID NO: 27
>Locus_l 815_nucleotide_mature
GTACATCAAAAGGGCAAGCAACCGGCGCCTGCA
GCAGAAGAATGGTCATATAGTGATGACGATGACAGCGTTTGCGGATACATAAATATAACA AAAACAGAGAAAGAGATGCTTCATCAACGCAAGAAACTGCCCCATACGCTCCCATTTATT GCCTACGTGGTGAAGAGACATCGACATAACTACCAACCGGTTTGTGTGGCTACAATTTTG CCAGATGCGGATGACGAGTCGAGCAGTATTGTGCTCACGCTTACGACATGCATCGAATCG CCACAACTGTTGGACCACTATAAAATTTTCACCGGAAGAAAACTTCCGGTGCCACTGAAT AAAACAAGGAAGATGTACGATATTGAAGAGGTTCTGGTCGATAAAAAGAGCCCAAATTTA CAGTATGCTAATGCAATGGTTGTGAAATTGAGCGGTTCGATACCAATTGATGCAAAGCGT CAGCCAATCTGCCTACCAAAACCAAACGCGGAAATGCCTAAGAAGTCTGAATGTATTCTT TCAGTTGTTCGAACCGATGGCAGACAGCAACCCTATCTGATTGAGGTTGAACCCAAGTCA AACAAGGACTGTAAGAAGATGCACGGAGAGAGCATCCAAATTGACAATAAAATGGAAATA TGCGGGCTCGACTATCGAGATGGATCATTTATCTCCAGAGGTGGATCACTGATTTGCGAA GGCGAAGGGGGCAGGTGGATACAGTACGGCATTTACACGCATGCTTCCCACATTTCTGAC AGAATGGCAGACAGACGGAGCGTATCCAGAATTCAAAAGAAAGGACGCCCCAGCGTGTAC ATCAATGTTGCCAACTTCACAAAGCTAATGCAAACGTCACAAAACCTGGAAAGACTATCA CGCATAAACTTAATGAAACTTCGCTGATCGAAAATGTACGCAAAAAATGATGTCCGTTCA TACGTGAATACTCCGCAAACTTTCTACACT
SEQ ID NO: 28
GTCCATCAGAAAGGAAAGCAACCTGCGCCCGCAGCGGAAGAGTGGAGCTATTCTGATGAT GATGATAGCGTTTGTGGTTATATTAATATTACAAAAACTGAAAAAGAAATGCTTCATCAA CGTAAAAAACTTCCTCATACTCTTCCATTTATTGCTTACGTTGTTAAACGTCATCGTCAT AATTACCAGCCTGTTTGTGTTGCTACGATTCTTCCTGATGCTGATGATGAATCTAGCAGC ATTGTTCTTACTCTTACTACTTGTATTGAAAGCCCTCAACTTCTTGATCATTACAAGATT TTTACTGGTAGAAAACTTCCTGTGCCCCTTAATAAAACTCGTAAAATGTATGATATTGAG GAGGTTCTTGTTGATAAAAAGCCTAATCTTCAATATGCTAATGCCATGGTTGTTAAACTA TCAGGTTCGATTCCTATTGATGCCAAACGTCAACCTATTTGTCTTCCTAAACCTAATGCC GAAATGCCTAAAAAATCCGAATGTATTCTTAGCGTTGTTCGTACGGACGGTCGTCAACAA CCCTATCTTATTGAAGTAGAACCTAAAAGCAATAAAGATTGTAAAAAAATGCATGGCGAA TCTATTCAGATAGACAATAAGATGGAAATTTGCGGTCTGGATTATCGTGATGGTTCTTTC ATATCTCGTGGTGGTAGCCTTATTTGTGAGGGTGAAGGTGGTCGTTGGATTCAGTATGGT ATCTATACTCATGCTTCTCATATTTCTGATCGTATGGCTGATCGTAGCGTTTCTCGAATA CAGAAGAAGGGTCGGCCTTCTGTTTATATTAATGTTGCGAATTTTACTAAACTTATGCAG ACTTCTCAAAATCTTGAGCGTCTTTCTCGTATTAATCTGATGAAGCTTCGTTTTTCTAAG ATGTATGCGAAGAATGATGTGCGTTCTTATGTGAATACTCCGCAAACTTTTTATACG
SEQ ID NO: 29
MLVLFFPLLLTVGLSTAGHVKCPDFGDWKPWTDCLWYPPQHMYSKLSHACGMHAHRNLTG VMDLPHGHKTPPPCGHCSFKFRCRRRPNTEGCYPLDGEVEVCHDHSDICTLPKLPHLGCG YAFINEKLKQCFTRPDTPSYVRLGYRKMFESIPKKHCIEKDGMCKCCCGDYEPNESGTEC IKPPAHDCPAYGPPSEWSECLWFPLKNIVSHVYDHCHVHKEPDGYEPHSVAPANVHIPEK CGFCSFRVKCMKRDKKDGCFPLKLGKKSCGKDDCPTCGDICTLDKINGSCAFPRVMKEKI WDDFTATSKEKHMPHWKRDGYAKMLMQLPYSNCKEVGDKCKCCCHPYEPNKDGTACVVKE YCKRVHELHHHDHHGHGEEHHKSSSSESKEHHHH
SEQ ID NO: 30
ATGCATACATTCCTAGCTGTTGCCTTTCTCGCTCTTTCCGAACTTTGTGCCGTTCACCCT TGGGATGGTTCTCCCTACACTCTCGAACAACTTGCCACTCGTCCTCGCCCTCGCGTTTCT TTTCCCTGGTTCGTCCTTATTAAAACTCCTCGCAATAAATGTGTCGGCACTATTCTTCGC GCCGATGATTCTCACAAAACTCCTCTCGTTCTTGCCCCTTCTGAATGTCTTCTTACCGCC GACATTTCTGTCAATGATACTTCTCAAGGCCAAAACGAAACCCGTCTCGAAATTCCTTCC ATGGTCGAAATCGTTGGTTCTAACTTTTCTTCTGTCGCCAAAAATCTCTATCTTTCCGTC AAAACCGGCTATCCTGTCGTTTTTATCAAACTTGAAGAACCTTATCGTGGCCCCACTGTT CTTGTCGATCGTGCTGTTTACTTTACTGGTAACGTTACTAAATGCATTCTTGTTCGTTTC TCTTCTAGCGAAACCCAAAAACGCCAACTTCTTTCTAATGACGATTCTCCTGTTCGTTTT CGTGAAGAAGAAGAAATCTGTCGTAATTCTACCGCTTCTGAAACCGGCATTCTTGACGGC CAAGGCCTTACTTGTAAAGCCGGTGATGGTCTCTGGACTTTTTACGGCTTTTGTGGCGAA GAAGGCTTCGAATCTATTTACGTCCCTGTTCAACCTCTTATCAAACTTATTACCTATGCT GCCCGTGAAATGGAACTTTACAAAAAAGACGAAGTTCTTAAAGGTCGTAAAATTTCTTAC AAACATTCTTTCCTTCATGGCCAAGAAGTTTGGACCGTCACTCGTGGTCGTCAAACCAAA ATCTACTATCGTGAACCCGTTCTT
SEQ ID NO: 31
CTGACAATGAAAATGAATCCATGTATGCTAATATCTATACTTGCACTTCTAGTTCTTAGA GCAGCATCTGAATGTGTAACACATAAAGATTGTCGTTCTGCACAAAGCCTATGTTTTGAT GGATATTGTCTACTAGCAACAGCAGTACCAGGAAAATGTACAACATCTGCAGATTGTAGA ACACAACTATCTGTAATGCAAAATGCAGGAAGAGGATGTAGAAATAATATATGTTATGAA ATAATAACAAATAAACTATGTACAGGACATGTTTCATGTGATGATGCACAAGTATGTATA AGAAATCATTGTGTACCTTCTGTAGCTACTTCTGCAAAATGTCATGTAGGAGCACTATGT GCAGCTGGTCAACGTTGTCTAGGAGGAATATGTTATGAACCACTAGAACCTTCTCGAGTA CTAGAAACAGACAATTCACGA
SEQ ID NO: 32
ATGACACCAATGCAATGGTTTCTAATAACTGCACTAGTAATAGTAGCATTTTCTGATGCA CGATTTCATTTTCGAAATAAATATAAAGTACATCAAAAAGGAAAACAACCTGCACCAGCA GCAGAAGAATGGTCTTATTCTGATGATGATGATTCTGTATGTGGATATATAAATATAACA AAAACAGAAAAAGAAATGCTACATCAACGAAAAAAACTACCACATACACTACCATTTATA GCATATGTTGTAAAACGTCATCGACATAATTATCAACCAGTATGTGTAGCAACAATACTA CCAGATGCAGATGATGAATCTTCATCAATTGTTCTTACACTAACAACATGTATTGAATCA CCACAACTTCTAGATCATTATAAAATATTTACAGGTCGAAAACTACCAGTACCACTAAAT AAAACTCGAAAAATGTATGATATAGAAGAAGTACTAGTAGATAAAAAAACTCCAAATCTA CAATATGCAAATGCAATGGTAGTAAAACTTTCTGGTTCTATTCCAATAGATGCAAAACGA CAACCTATATGTCTACCAAAACCAAATGCAGAAATGCCAAAAAAATCTGAATGTATACTA AGCGTAGTACGAACAGATGGACGTCAACAACCATATCTAATAGAAGTAGAACCAAAATCT AATAAAGATTGTAAAAAAATGCATGGAGAATCTATACAAATAGATAATAAAATGGAAATA TGTGGACTAGATTATAGAGATGGTTCTTTTATTTCTCGAGGAGGTTCTCTAATATGTGAA GGAGAAGGAGGTAGATGGATACAATATGGAATATATACACATGCTTCTCATATTTCTGAT AGAATGGCAGATCGACGATCTGTATCTCGAATACAAAAAAAAGGACGACCATCAGTATAT ATTAATGTTGCTAATTTTACTAAACTTATGCAAACTTCTCAAAATCTTGAACGTCTTTCT CGTATTAATCTTATGAAACTTCGTTTTTCTAAAATGTATGCAAAAAATGATGTACGTTCA TATGTAAATACACCACAAACATTTTATACA
SEQ ID NO: 33
>T . suis counterpart of the T. muris protein TMC00067_1
ATGGATCGACTACCACTAGTTTCTGATCCAAAATATGTACATAAAAATTCTGCACGACTA
AGATCTGCAAGACGACCAAAATCTAATAGACGATTTGGAAAATTTCCAGCAAAACGATCT
CATCATCAATGTGCAGATATAGCAGCTTCTAATTCTGCAGCAGCAGAAGGAACAACATCA
CGAATAGTTATTCTTTCTACAGCAAAATGGACATATGCAATGATAACAGCAAAATCTGTA
CGTTGTCAAAATTGTCAACATCTAGGATGTGGATATGCATTTATAAATGAAAAACTAAAA
CAATGTTTTACACGTCCAGATACACCATCTTATGTACGTCTAGGATATCGTAAAATGTTT
GAATCTATACCAAAAAAACATTGTATAGAAAAAGATGGAATGTGTAAATGTTGTTGTGGA
GATTATGAACCAAATGAAGCAGGAACAGAATGTGTAAAACCACCAGCTCATGATTGTCCA
GCATTTGGACCACCTTCTGAATGGTCTGAATGTCTAGGTTCTCAA
SEQ ID NO: 34
TGGGACGGTAGCCCATATACACTTGAACAACTAGCTACTCGACCACGACCACGAGTATCT TTTCCATGGTTTGTACTTATTAAAACTCCACGTAATAAATGTGTAGGAACTATTCTACGT GCAGATGATTCTCATAAAACACCTCTAGTTCTAGCACCTTCTGAATGTCTTCTAACAGCA GATATTTCTGTAAATGATACATCTCAAGGACAAAATGAAACACGTCTAGAAATACCATCT ATGGTAGAAATAGTTGGTTCTAATTTTTCTTCTGTAGCAAAAAATCTATATCTTTCTGTA AAAACAGGATATCCTGTAGTATTTATTAAACTTGAAGAACCATATCGAGGACCTACTGTA CTTGTTGATCGTGCTGTTTATTTTACTGGTAATGTTACTAAATGTATTCTTGTTAGATTT TCTAGCTCTGAAACTCAAAAACGTCAACTTCTATCAAATGATGATTCTCCTGTAAGATTT CGAGAAGAAGAAGAAATATGTCGAAATTCAACTGCTTCAGAAACTGGAATTCTAGATGGA CAAGGACTTACATGTAAAGCAGGAGATGGACTTTGGACATTTTATGGATTTTGTGGAGAA GAAGGTTTTGAATCAATTTATGTTCCAGTACAACCACTAATTAAACTAATTACATATGCA GCACGAGAAATGGAACTATATAAAAAAGATGAAGTACTAAAAGGTCGAAAAATTTCTTAC AAACACTCTTTTCTACATGGACAAGAAGTATGGACAGTAACTCGAGGTCGTCAAACAAAA ATATATTATCGAGAACCAGTATTA
SEQ ID NO: 35
GAATGTGTAACCCATAAAGATTGTCGATCAGCTCAATCTCTATGTTTTGATGGATATTGT CTTCTAGCAACAGCAGTACCAGGAAAATGTACAACATCTGCAGATTGTCGAACACAACTA TCTGTAATGCAAAATGCAGGACGAGGATGTCGAAATAATATATGTTATGAAATAATAACA AATAAACTATGTACAGGACATGTATCATGTGATGATGCACAAGTATGTATACGAAATCAT TGTGTACCTTCTGTAGCAACATCAGCAAAATGTCATGTAGGAGCACTATGTGCAGCAGGA CAACGATGTCTAGGAGGAATATGTTATGAACCACTAGAACCATCTCGAGTACTAGAAACA GATAACTCTAGG
SEQ ID NO: 36
GTTCACCAGAAAGGAAAACAACCAGCACCAGCAGCAGAAGAATGGTCATATTCTGATGAT GATGATTCAGTATGTGGATATATAAATATAACAAAAACAGAAAAAGAAATGCTACATCAA CGAAAAAAACTACCACATACACTACCATTTATAGCATATGTAGTAAAAAGACATAGACAT AATTATCAACCAGTATGTGTAGCAACAATACTACCAGATGCAGATGATGAATCATCATCA ATAGTACTAACACTAACAACATGTATAGAATCACCACAACTACTAGATCATTATAAAATA TTTACAGGACGAAAACTACCAGTACCACTAAATAAAACACGAAAAATGTATGATATAGAA GAAGTACTAGTAGATAAAAAACCAAATCTACAATATGCAAATGCAATGGTAGTAAAACTA TCAGGATCAATTCCAATAGATGCAAAACGACAACCAATATGTCTACCAAAACCAAATGCA GAAATGCCAAAAAAATCAGAATGTATACTTTCAGTAGTACGAACAGATGGAAGACAACAA CCATATCTTATAGAAGTAGAACCAAAATCAAATAAAGATTGTAAAAAAATGCATGGAGAA TCAATTCAAATAGATAATAAAATGGAAATTTGTGGACTTGATTATCGAGATGGTTCATTT ATATCACGAGGAGGTTCTCTAATATGTGAAGGAGAAGGTGGACGATGGATTCAATATGGA ATATATACACATGCTTCACATATTTCAGATCGAATGGCAGATCGTTCAGTATCTCGAATA CAAAAAAAAGGACGACCTTCTGTATATATTAATGTAGCAAATTTTACAAAACTAATGCAA ACATCACAAAATCTTGAAAGGCTATCACGAATAAATCTAATGAAACTACGATTTTCTAAA ATGTATGCAAAAAATGATGTTCGTTCTTATGTTAATACACCTCAAACATTTTATACA
SEQ ID NO: 37
MKMNPCMLISILALLVLRAASECVTHKDCRSAQSLCFDGYCLLATAVPGKCTTSADCRTQLS VMQNAGRGCRN ICYEI ITNKLCTGHVSCDDAQVCIRNHCVPS ATSAKCHVGALCAAGQRC
LGGICYEPLEPSRVLETDNSPVQSFY
SEQ ID NO: 38
MTPMQWFLITALVIVAFSDARFHFRNKYKVHQKGKQPAPAAEEWSYSDDDDSVCGYINITKT EKEMLHQRKKLPHTLPFIAYVVKRHRHNYQPVCVATILPDADDESSSIVLTLTTCIESPQLL DHYKIFTGRKLPVPLNKTRKMYDIEEVLVDKKSPNLQYANAMVVKLSGSIPIDAKRQPICLP KPNAEMPKKSECILSWRTDGRQQPYLIEVEPKSNKDCKKMHGESIQIDNKMEICGLDYRDG SFISRGGSLICEGEGGRWIQYGIYTHASHISDRMADRRSVSRIQKKGRPSVYINVANFTKLM QTSQNLERLSRINLMKLR
SEQ ID NO: 39
MKTAILIFLATILTLKPKAKCAAESGARSVCNTVRRRSAGMLRKDSQDASDEKYVRGCYFTN WAQYRPGNGKYNPEHYQANLCEYIFYAFAKLNDDFTVDQFEWNDIDVLYPGVMKQKSSQPDL KVLLSLGGWNAGTATFKKMAATYSNRAKFISSLVSFLQQNKFDGFDLDWEYPESSDKENYLL LCQEILAKFEEVAKCTSTSRLLFTAAVSANPKTVDAGYDVPALAKVLDFVNLMCYDFHGAWE TQTGINSPLYSRKEDSSEFKMWNVEQSSKYWSDKGMPKKQIIIGLPTYGRGWTLSDASKTDI GAPAQGSSTATEYLREAGVISYYEVCQKLSSGAKRVWDDESKTPYLVQGNQWFSYDDVESMK AKINWIKQENYGGAFVWTLDYDDFLGSFCTEHNGKKYPLISLMQEILGGGYVPPSTESTTSQ VTTTPSTTTSTTSPAGAFQCPSDGLFPDPESCSNFYQCAGGTAYKMKCPTGLMFNPKTSTCD YPSNVDCQEKTIT
SEQ ID NO: 40
MYTASAFVGISYLLLATLVAEGRHVPPEQAAKYPCGVSKFPITAPGNRISKGWEAAKHSLPW HVHLMMYDSPQERRGEQCGGSLIRIKPANVTDLVLTAAHCVKNPKTLMDHPVNRIAVTVGIH DRTDPHRTKVRVKKFVTADYRGDMSGKENDIALLRLKSAIPYTDFTRPVCLPSKGDVLPVGQ ECYVSGHGATFMLDPGSTSNKLLMVDVNVLTADQCSRDMDPGKKFNHSTQFCAGTNDRSARA GDSGGPLVCKKGDRFVQYGIVSFGSNDPRYFDRSGKYTFVPPFVDWIEKMDKQLPHSDESLS EDAIHEYLTQNELAQIERNRTLSALQKPTRVAKKPPVLSLSEYKCGMPAPDYPILLSPTGMN RIVNGWEAEKHSLPWMVLVNSPDPYNKAVTQSCGGSLIQLYPGNRTDLVLTAAHCFKKTADS PDKKPSDITVFVGVHHQYADDALRKEVRVKALKLHEKYDAKQFHNDIALLRLAQAVPYTDIT RPICLPEQGEQVQVGTVCLIAGWGKTSHHATMLESELKMTIAPVAEFNECNQNLPITVYEDQ VLCTKPEARRGFCYGDSGGPLICKKGDQWFQYGIVSFSPQRCATTFDKYAKVSSYINWIVKQ DKYFPATTEEFPYEDLVEYRSPMEIVTGTRKPSSSNLVQKPLTPQSSLPTKPSLTSPSKIVT GGLPGRPSALPSRFAQPGPTVSSNIGRRPAAFSRFLS
SEQ ID NO: 41
MKFVLILYLLYTISSQRCTADTNDPNEAKGANTLGTGVAEIKKPTMADKEEILTTVSVVDRQ MRTVEANETDTKEENPLQKGEERNLTETVPKKENHTEEEKMEENKEATKNKRSVGIGDNVGT TGQDRMVIEEANDPQNPAMTITRGEDTGRPIRRNINRRRNFDGSSSEEGNPEHGHHHGKNES SSSSSSSEEEHRPHPNCTGKPPPPEGHNVTNYDNIWEELRTVFGFSDVHIDKYKAFMDKLRK YRWAKLSEKEREMVTHLKSNQCATRYSQLPLQIVDYRNINLGGAEGNPLLYEGDILLSENQI DILNKVIDEDINGTRTLDLAAIASNLASRWTTTIYYTISGVDSNVIEAGLQKWRETTCLKFI RNDQYTGDRIQYIKGSGCYSSVGRLGGKQQVSIGTGCERVGTVCHETSHALGVWHEQSRTDR DNYISINYNNIIASAVGNFERLTSNIMYLDQVPYDFGSDMHYPPKSFAINYNQYVITTKDSK YQYTIGQRDYPSFNDAKVLNLAYCGSICAPRSCQNKGYADPKNCNICRCPEGLAGDSCERSK SQHSHYHKCNKSRLLSSGGRLQTTVKAFSFNCQETCTGSYFGIQAGSDIGNSVARLCCSRPG VITSTGSSILVIYQGRQNNGVTFEVKQALEPY TQ
SEQ ID NO: 42
MQTVRTLRLVFLCFSVAYCGRTRIPTDEEVEMAYTLLNKVFPPTRGPVKVVHRIDQVTYSGE EDKYTLKFMAVDSTCQTLLAGMDQKDCAPVSYPTKENFKYFIGNIKKTGSGYEVLGGSFRNP EAVEFLEVDR
SEQ ID NO: 43
MGWCQRFLVILSLALYWRSSQGQNLETFTATERTQIVSSHNKFRGMLEGGNEQCITGYDTSL QAIAESVAVNCSIQDVRDRVDQYGLAFYFKLLPDTRPTMEEFVEDIYTGVNSYDYTAANCID EIPCHSFLAFSWFENSRVGCALATCASVKAAEDENSQQYFGVCAYNYKADLTARPYVAPPEC
QFCPSNAKICTNHLCCPYDLTAMTCPEGAGAGVPASLKSLYRVHHTRANTVLTTNETEVDQL VQAGGTSAGIIGRIATGNDSSCSFLKPVHHIYSPHFKSNYYMTDDYLFKVRMKEGYENKGII GYAVQGVHSCNATIAIYDFFRPTDGITHIPNSTEVQNYMNYKSGYIYHGVSFALWA
SEQ ID NO: 44
MRMLWQLLFSQLFLIAPVMPYGFTLWVNGKQVTDPAQLEKFFQRYPQFKPPNWGGTGQSVKP TSTAKPVTPKPQQSSPSPTETNTNVNNEAKETQTQLSSTAPGAISLSANVIKFALEEHNNYR KKVAKGQVTNMPAATAMNELAWSNAIAANASAWAAKCSMTHTPSGYYCGLEGYDNIGENLAA STTSSSGLSSDEQLKSAFQSAFKSWFDEYKDYNFNTRACSRVCGHFTQMIKDSSEKLGCGIA LCKNGISGFILGRPSYI I CNYGPGN FSNKPAYETAKGTECPQVRPIRKDDLCTGSGKRGP KNCVDNTSYCSV KNEGKCELCNNPYYNWMAGECPRTCGICS SEQ ID NO: 45
MNLRLVYIQILLLWCVTIVHTAPLQCKNAAKADISWAVFYVVPQKQAARAIYAAPAPAWEQQ EVDLTPVDGALGELFTPFFTAQTKPTYNLIAYSNFPPLFRAPVSKSSPAKGVLGYTDGNGWW LVHSIEQWPDMTGTAFTAPAAGSAGLIVCLSIPFTSLKEWATVLNFEDPVVYYYHSSTPPAA TSIGNIKELNDLTKKSSPWYPPFVKSMTFTTVGASGALTTFVGKAAKAFIDIYSKHLANKV LKQSLIVWSDQTKGAKLASYCSGKYKVENVKPGQTIKVDSVDVKRSEDTSNWAVSKDAGATA VFCTSDAVRTTAWKTVPGGAVCLQQQQLQTLFSAVANTAEIEKCPSSG
SEQ ID NO: 46
ECVTHKDCRSAQSLCFDGYCLLATAVPGKCTTSADCRTQLSVMQNAGRGCRNNICYEIITNK LCTGHVSCDDAQVCIRNHCVPSVATSAKCHVGALCAAGQRCLGGICYEPLEPSRVLETDNSP
VQSFY
SEQ ID NO: 47
RFHFRNKYKVHQKGKQPAPAAEEWSYSDDDDSVCGYINITKTEKEMLHQRKKLPHTLPFIAY VVKRHRHNYQPVCVATILPDADDESSSIVLTLTTCIESPQLLDHYKIFTGRKLPVPLNKTRK MYDIEEVLVDKKSPNLQYANAM VKLSGSIPIDAKRQPICLPKPNAEMPKKSECILSWRTD GRQQPYLIEVEPKSNKDCKKMHGESIQIDNKMEICGLDYRDGSFISRGGSLICEGEGGRWIQ
YGIYTHASHISDRMADRRSVSRIQKKGRPSVYINVANFTKLMQTSQNLERLSRINLMKLR
SEQ ID NO: 48
AAESGARSVCNTVRRRSAGMLRKDSQDASDEKYVRGCYFTNWAQYRPGNGKYNPEHYQANLC EYIFYAFAKLNDDFTVDQFEWNDIDVLYPGVMKQKSSQPDLKVLLSLGGWNAGTATFKKMAA TYSNRAKFISSLVSFLQQNKFDGFDLDWEYPESSDKENYLLLCQEILAKFEEVAKCTSTSRL LFTAAVSANPKTVDAGYDVPALAKVLDFV LMCYDFHGAWETQTGINSPLYSRKEDSSEFKM WNVEQSSKYWSDKGMPKKQIIIGLPTYGRGWTLSDASKTDIGAPAQGSSTATEYLREAGVIS YYEVCQKLSSGAKRVWDDESKTPYLVQGNQWFSYDDVESMKAKINWIKQENYGGAFVWTLDY DDFLGSFCTEHNGKKYPLISLMQEILGGGYVPPSTESTTSQVTTTPSTTTSTTSPAGAFQCP SDGLFPDPESCSNFYQCAGGTAYKMKCPTGLMFNPKTSTCDYPSNVDCQEKTIT
SEQ ID NO: 49
RHVPPEQAAKYPCGVSKFPITAPGNRISKGWEAAKHSLPWHVHLMMYDSPQERRGEQCGGSL
IRIKPANVTDLVLTAAHCVKNPKTLMDHPVNRIAVTVGIHDRTDPHRTKVRVKKFVTADYRG DMSGKENDIALLRLKSAIPYTDFTRPVCLPSKGDVLPVGQECYVSGHGATFMLDPGSTSNKL LMVDVNVLTADQCSRDMDPGKKFNHSTQFCAGTNDRSARAGDSGGPLVCKKGDRFVQYGIVS FGSNDPRYFDRSGKYTFVPPFVDWIEKMDKQLPHSDESLSEDAIHEYLTQNELAQIERNRTL SALQKPTRVAKKPPVLSLSEYKCGMPAPDYPILLSPTGMNRIVNGWEAEKHSLPWMVLVNSP DPYNKAVTQSCGGSLIQLYPGNRTDLVLTAAHCFKKTADSPDKKPSDITVFVGVHHQYADDA LRKEVRVKALKLHEKYDAKQFHNDIALLRLAQAVPYTDITRPICLPEQGEQVQVGTVCLIAG WGKTSHHATMLESELKMTIAPVAEFNECNQNLPITVYEDQVLCTKPEARRGFCYGDSGGPLI CKKGDQWFQYGIVSFSPQRCATTFDKYAKVSSYINWIVKQDKYFPATTEEFPYEDLVEYRSP MEIVTGTRKPSSSNLVQKPLTPQSSLPTKPSLTSPSKIVTGGLPGRPSALPSRFAQPGPTVS SNIGRRPAAFSRFLS SEQ ID NO: 50
DTNDPNEAKGANTLGTGVAEIKKPTMADKEEILTTVSVVDRQMRTVEANETDTKEENPLQKG EERNLTETVPKKENHTEEEKMEENKEATKNKRSVGIGDNVGTTGQDRMVIEEANDPQNPAMT ITRGEDTGRPIRRNINRRRNFDGSSSEEGNPEHGHHHGKNESSSSSSSSEEEHRPHPNCTGK PPPPEGHNVTNYDNIWEELRTVFGFSDVHIDKYKAFMDKLRKYRWAKLSEKEREMVTHLKSN QCATRYSQLPLQIVDYRNINLGGAEGNPLLYEGDILLSENQIDILNKVIDEDINGTRTLDLA AIASNLASRWTTTIYYTISGVDSNVIEAGLQKWRETTCLKFIRNDQYTGDRIQYIKGSGCYS SVGRLGGKQQVSIGTGCERVGTVCHETSHALGVWHEQSRTDRDNYISINYNNIIASAVGNFE RLTSNIMYLDQVPYDFGSDMHYPPKSFAINYNQYVITTKDSKYQYTIGQRDYPSFNDAKVLN LAYCGSICAPRSCQNKGYADPKNCNICRCPEGLAGDSCERSKSQHSHYHKCNKSRLLSSGGR LQTTVKAFSFNCQETCTGSYFGIQAGSDIGNSVARLCCSRPGVITSTGSSILVIYQGRQNNG VTFEVKQALEPY TQ
SEQ ID NO: 51
GRTRIPTDEEVEMAYTLLNKVFPPTRGPVKVVHRIDQVTYSGEEDKYTLKFMAVDSTCQTLL AGMDQKDCAPVSYPTKENFKYFIGNIKKTGSGYEVLGGSFRNPEAVEFLEVDR
SEQ ID NO: 52
SQGQNLETFTATERTQIVSSHNKFRGMLEGGNEQCITGYDTSLQAIAESVAVNCSIQDVRDR VDQYGLAFYFKLLPDTRPTMEEFVEDIYTGVNSYDYTAANCIDEIPCHSFLAFSWFENSRVG CALATCASVKAAEDENSQQYFGVCAYNYKADLTARPYVAPPECQFCPSNAKICTNHLCCPYD
LTAMTCPEGAGAGVPASLKSLYRVHHTRANTVLTTNETEVDQLVQAGGTSAGIIGRIATGND SSCSFLKPVHHIYSPHFKSNYYMTDDYLFKVRMKEGYENKGIIGYAVQGVHSCNATIAIYDF FRPTDGITHIPNSTEVQNYMNYKSGYIYHGVSFALWA
SEQ ID NO: 53
YGFTLWVNGKQVTDPAQLEKFFQRYPQFKPPNWGGTGQSVKPTSTAKPVTPKPQQSSPSPTE TNTNVNNEAKETQTQLSSTAPGAISLSANVIKFALEEHNNYRKKVAKGQVTNMPAATAMNEL AWSNAIAANASAWAAKCSMTHTPSGYYCGLEGYDNIGENLAASTTSSSGLSSDEQLKSAFQS AFKSWFDEYKDYNFNTRACSRVCGHFTQMIKDSSEKLGCGIALCKNGISGFILGRPSYIIVC NYGPGNNFSNKPAYETAKGTECPQVRPIRKDDLCTGSGKRGPKNCVDNTSYCSVWKNEGKCE LCNNPYYNWMAGECPRTCGICS
SEQ ID NO: 54
APLQCKNAAKADISWAVFYVVPQKQAARAIYAAPAPAWEQQEVDLTPVDGALGELFTPFFTA QTKPTYNLIAYSNFPPLFRAPVSKSSPAKGVLGYTDGNGWWLVHSIEQWPDMTGTAFTAPAA GSAGLIVCLSIPFTSLKEWATVLNFEDPVVYYYHSSTPPAATSIGNIKELNDLTKKSSPWY PPFVKSMTFTTVGASGALTTFVGKAAKAFIDIYSKHLANKVLKQSLIVWSDQTKGAKLASYC SGKYKVENVKPGQTIKVDSVDVKRSEDTSNWAVSKDAGATAVFCTSDAVRTTAWKTVPGGAV CLQQQQLQTLFSAVANTAEIEKCPSSG

Claims

WHAT IS CLAIMED IS:
1. An isolated helminthic excretory/secretory polypeptide, said polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: I, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, or a biologically active or immunogenic fragment thereof.
2. The polypeptide of claim 1 isolated from Trichuris suis.
3. The polypeptide of claim 1 having an amino acid sequence of SEQ ID NO: 4.
4. The polypeptide of claim 1 having an amino acid sequence of SEQ ID NO: 5.
5. The polypeptide of claim 1 having an amino acid sequence of SEQ ID NO: 6.
6. The polypeptide of claim 1 having an amino acid sequence of SEQ ID NO: 7.
7. A recombinant polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, or a biologically active or immunogenic fragment thereof.
8. A recombinant polypeptide of claim 7 obtained by a process comprising: culturing a cell transformed with a polynucleotide comprising a nucleic acid sequence encoding an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-7 under conditions suitable for polypeptide expression, and
recovering the polypeptide so expressed.
9. A pharmaceutical composition comprising a therapeutically effective amount of a recombinant polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, or a biologically active or immunogenic fragment thereof; and a pharmaceutically acceptable carrier.
10. A composition comprising one or more isolated Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, said one or more Ts E/S polypeptides having a molecular weight ranging from about 20 kD and about 80 kD.
1 1. The composition of claim 10 in which the one or more polypeptides are recombinant.
12. The composition of claim 10 in which the one or more polypeptides have an amino acid sequence selected from the group consisting of SEQ ID NO: I, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, or a biologically active or immunogenic fragment thereof.
13. The composition of claim 10 in which the one or more polypeptides have a molecular weight of about 58 kD.
14. The composition of claim 10 in which the one or more polypeptides have a molecular weight of about 50 kD.
15. The composition of claim 10 in which the one or more polypeptides have a molecular weight of about 45 kD.
16. The composition of claim 10 in which the one or more polypeptides have a molecular weight of about 35 kD.
17. The composition of claim 10 in which the one or more polypeptides have a molecular weight of about 20 kD.
18. The composition of claim 10 in which the polypeptides were obtained from in vitro hatched T. suis first-stage larvae.
19. The composition of claim 10 in which the polypeptides are obtained by a process comprising:
culturing in vitro hatched T. suis first-stage larvae in the presence of a culture medium;
harvesting a portion of the culture medium while replenishing an amount of culture medium harvested; and
isolating the one or more polypeptides from the harvested culture medium.
20. A method of modulating a Thl- or Th2-mediated immune response in a subject comprising administering to a subject whose Thl- or Th2-mediated immune response is in need of modulation an effective amount of one or more isolated polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, a recombinant version thereof, or a biologically active or immunogenic fragment of the foregoing.
21. A method of modulating a Thl- or Th2-mediated immune response in a subject comprising contacting one or more T regulatory cells of a subject with a composition comprising one or more isolated Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, each having a molecular weight ranging from about 20 kD and about 80 kD, and a carrier.
22. A method of regulating expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN-γ in a subject comprising administering to a subject whose expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN-γ are in need of regulation an effective amount of a composition comprising one or more isolated Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, each having a molecular weight ranging from about 20 kD and about 80 kD, and a carrier.
23. A method of treating allergic airway hyperreactivity or inflammation in a subject, said method comprising administering to a subject in need thereof an effective amount of a composition comprising one or more isolated Trichuris suis (Ts) excretory/secretory (E/S) polypeptides, each having a molecular weight ranging from about 20 kD and about 80 kD and a carrier.
24. A composition comprising a mixture of isolated Trichuris suis (Ts) excretory/secretory (E/S) polypeptides obtained by culturing Ts first stage larvae in an aqueous medium, harvesting a portion of the medium at predetermined intervals while replenishing the medium over a period of about one to two weeks.
25. The composition of claim 24 in which 50,000 larvae/well are cultured on a 24- well plate using 0.5 mL of serum-free medium per well.
26. The composition of claim 24 in which 0.4 mL of supernatant is subsequently harvested every 48 hours with the medium being replenished over a period often days.
27. The composition of claim 24 in which 0.4 mL of supernatant is subsequently harvested every 48 hours with the medium being replenished over a period of fourteen days.
28. A protein pellet obtained by acidifying the composition of claim 24.
29. The protein pellet of claim 24 obtained by adding 100% trichloroacetic acid at a TCA volume to sample volume ratio of 1 :5.
30. A method of modulating an immune response of a subject comprising administering to a subject whose immune response is in need of modulation an effective amount of a composition comprising one or more isolated Ts E/S polypeptides.
31. The method of claim 30 in which splenocytes of a treated subject exhibit suppressed IFN-γ and IL-4 production compared to splenocytes of a control subject.
32. The method of claim 30 in which dendritic cells of a treated subject exhibit no upregulation in their expression of CD86 and MHC class II molecules compared to dendritic cells of a control subject.
33. The method of claim 32 in which dendritic cells of a treated subject exhibit suppressed IL-6 production compared to dendritic cells of a control subject.
34. A method of alleviating allergic airway hyperreactivity or inflammation comprising administering to a subject in need thereof an effective amount of a composition comprising one or more isolated Ts E/S polypeptides.
35. The method of claim 34 in which treated subjects exhibit reduced lymphocyte and eosinophil infiltration compared to a control subject.
36. The method of claim 34 in which treated subjects exhibit suppressed IL-4, IL- 5 and IL-13 production compared to a control subject.
37. An isolated polypeptide comprising an amino acid sequence at least 95% identical to a sequence selected from the group consisting of SEQ ID NOs: 1-7.
38. An isolated polynucleotide comprising a nucleic acid sequence that encodes an amino acid sequence at least 95% identical to a sequence selected from the group consisting of SEQ ID NOs: 1-7.
39. An isolated helminthic excretory/secretory polypeptide, said polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 37-54, or a biologically active or immunogenic fragment thereof.
40. A recombinant polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 37-54, or a biologically active or immunogenic fragment thereof.
41. A recombinant polypeptide of claim 40 obtained by a process comprising: culturing a cell transformed with a polynucleotide comprising a nucleic acid sequence encoding an amino acid sequence selected from the group consisting of SEQ ID NOs: 37-54 under conditions suitable for polypeptide expression, and
recovering the polypeptide so expressed.
42. A pharmaceutical composition comprising a therapeutically effective amount of a recombinant polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 37-54, or a biologically active or immunogenic fragment thereof; and a pharmaceutically acceptable carrier.
43. The composition of claim 10 in which the one or more polypeptides have an amino acid sequence selected from the group consisting of SEQ ID NOs: 37-54, or a biologically active or immunogenic fragment thereof.
44. A method of modulating a Thl- or Th2-mediated immune response in a subject comprising administering to a subject whose Thl- or Th2-mediated immune response is in need of modulation an effective amount of one or more isolated polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOs: 37-54, a recombinant version thereof, or a biologically active or immunogenic fragment of the foregoing.
45. An isolated polypeptide comprising an amino acid sequence at least 95% identical to a sequence selected from the group consisting of SEQ ID NOs: 37-54.
46. An isolated polynucleotide comprising a nucleic acid sequence that encodes an amino acid sequence at least 95% identical to a sequence selected from the group consisting of SEQ ID NOs: 37-54.
47. A method of modulating a Thl- or Th2-mediated immune response in a subject comprising administering to a subject whose Thl- or Th2-mediated immune response is in need of modulation a composition comprising Ts E/S supernatants but substantially free of Ts eggs and first stage larvae.
48. A method of modulating a Thl- or Th2-mediated immune response in a subject comprising contacting one or more T regulatory cells of a subject with a composition comprising Ts E/S supernatants but substantially free of Ts eggs and first stage larvae.
49. A method of regulating expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN-γ in a subject comprising administering to a subject whose expression levels of one or more of IL-4, IL-6, IL-10, IL-13 and IFN-γ are in need of regulation a composition comprising Ts E/S supernatants but substantially free of Ts eggs and first stage larvae.
50. A method of treating allergic airway hyperreactivity or inflammation in a subject, said method comprising administering to a subject in need thereof a composition comprising Ts E/S supernatants but substantially free of Ts eggs and first stage larvae.
PCT/US2014/059440 2013-10-07 2014-10-07 T. suis excretory/secretory products and methods of their use WO2015054210A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361887900P 2013-10-07 2013-10-07
US61/887,900 2013-10-07
US201461940166P 2014-02-14 2014-02-14
US61/940,166 2014-02-14

Publications (2)

Publication Number Publication Date
WO2015054210A2 true WO2015054210A2 (en) 2015-04-16
WO2015054210A3 WO2015054210A3 (en) 2015-06-18

Family

ID=52813730

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/059440 WO2015054210A2 (en) 2013-10-07 2014-10-07 T. suis excretory/secretory products and methods of their use

Country Status (1)

Country Link
WO (1) WO2015054210A2 (en)

Also Published As

Publication number Publication date
WO2015054210A3 (en) 2015-06-18

Similar Documents

Publication Publication Date Title
US11000566B2 (en) Compositions and methods for modulating the immune system
Aharoni et al. Bystander suppression of experimental autoimmune encephalomyelitis by T cell lines and clones of the Th2 type induced by copolymer 1
JP2023029565A (en) Use of low dose il-2 for treating autoimmune-related or inflammatory disorders
Deeg Ocular immunology in equine recurrent uveitis
EP2268669B1 (en) Bovine Colostrum comprising anti-insulin antibodies for treating Metabolic Disorder diseases.
WO2018102597A1 (en) Methods of treating inflammatory conditions
US20210253737A1 (en) Methods and compositions for treating disease-related cachexia
KR102468907B1 (en) Conjugate vaccines targeting in vivo proteins that cause disease
WO2017044979A2 (en) Lactoferrin compositions and methods for modulation of t cell subtypes and treatment of autoimmune diseases
EP1664122B1 (en) Therapeutic humanised antibodies against cd45 isoforms
KR20180095037A (en) Prevention and treatment of cancer by inhibition of water-soluble tumor necrosis factor
KR20020001865A (en) Use of soluble costimulatory molecules to enhance immune responses
RU2598247C2 (en) Modulation of antigen immunogenicity by deleting epitopes recognized by nkt-cells
WO2012062697A1 (en) Combination therapy for type 1 diabetes
US20180086802A1 (en) Peptide
WO2015054210A2 (en) T. suis excretory/secretory products and methods of their use
CA3051826A1 (en) Prophylactic and/or therapeutic agent for infectious disease or inflammatory disease
US20210061886A1 (en) Modified anti-inflammatory proteins and method of use
US20230348562A1 (en) Proinsulin Peptides for Type I Diabetes
US7598223B2 (en) Methods for producing target cell reactive lymphocytes
WO2023159140A2 (en) Anti-asgr1 polypeptides and methods of use for immune tolerance
Samara et al. Prothymosin Alpha and Immune
WO2013059322A2 (en) Amyloid beta peptide as a therapy for inflammation
Hamour The Role of IL-17 in the Pathogenesis of Glomerulonephritis and ANCA-associated Vasculitis
Mattapallil et al. Interleukin 22 ameliorates neuropathology and protects from central

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14852640

Country of ref document: EP

Kind code of ref document: A2