WO2007025977A2 - Anti-tnf alpha producing lactic acid bacteria for the treatment of chronic enterocolitis - Google Patents

Anti-tnf alpha producing lactic acid bacteria for the treatment of chronic enterocolitis Download PDF

Info

Publication number
WO2007025977A2
WO2007025977A2 PCT/EP2006/065803 EP2006065803W WO2007025977A2 WO 2007025977 A2 WO2007025977 A2 WO 2007025977A2 EP 2006065803 W EP2006065803 W EP 2006065803W WO 2007025977 A2 WO2007025977 A2 WO 2007025977A2
Authority
WO
WIPO (PCT)
Prior art keywords
nano3f
lactic acid
organism
micro
disease
Prior art date
Application number
PCT/EP2006/065803
Other languages
French (fr)
Other versions
WO2007025977A8 (en
WO2007025977A3 (en
Inventor
Pieter Rottiers
Klaas Vandenbroucke
Original Assignee
Actogenix Nv
Ablynx N.V.
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 Actogenix Nv, Ablynx N.V. filed Critical Actogenix Nv
Priority to AU2006286563A priority Critical patent/AU2006286563B2/en
Priority to JP2008528509A priority patent/JP5049973B2/en
Priority to ES06778386.0T priority patent/ES2626610T3/en
Priority to CA2619748A priority patent/CA2619748C/en
Priority to EP06778386.0A priority patent/EP1948206B1/en
Priority to US12/065,335 priority patent/US9017662B2/en
Publication of WO2007025977A2 publication Critical patent/WO2007025977A2/en
Publication of WO2007025977A3 publication Critical patent/WO2007025977A3/en
Publication of WO2007025977A8 publication Critical patent/WO2007025977A8/en

Links

Classifications

    • 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
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/06Fungi, e.g. yeasts
    • A61K36/062Ascomycota
    • A61K36/064Saccharomycetales, e.g. baker's yeast
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®

Definitions

  • the present invention relates to a novel treatment of chronic enterocolitis. More specifically, the invention relates to the production of a medicament comprising anti-TNF ⁇ producing lactic acid bacteria, and the use of this medicament in the treatment of chronic enterocolitis.
  • IBD Inflammatory bowel disease
  • UC ulcerative colitis
  • CD Crohn's disease
  • IBD diseases appear to result from the unrestrained activation of an inflammatory response in the intestine.
  • UC and CD are associated with many symptoms and complications, including growth retardation in children, rectal prolaps, blood in stools, wasting, iron deficiency and anemia.
  • UC refers to a chronic, non-specific, inflammatory and ulcerative disease having manifestations primarily in the colonic mucosa. It is frequently characterized by bloody diarrhea, abdominal cramps, blood and mucus in the stoll, malaise, fever, anemia, anorexia, weight loss, leukocytosis, hypoalbuminemia and an elevated erythrocyte sedimentation rate.
  • Crohn's disease shares many features in common with ulcerative colitis. Crohn's disease is distinguishable in that lesions tend to be sharply demarcated from adjacent normal bowel, in contrast to the lesions of ulcerative colitis which are fairly diffuse. Additionally, Crohn's disease predominantly afflicts the ileum (ileitis) and the ileum and colon (ileocolitis). In some cases, the colon alone is diseased (granulomatous colitis) and sometimes the entire small bowel is involved. Colon cancer is a known complication of chronic IBD. It is increasingly common in those patients who have IBD for many years. The risk for cancer begins to rise significantly after eight to ten years of IBD, making a fast and efficient treatment of IBD even more important.
  • the most commonly used medication to treat IBD includes anti-inflammatory drugs such as corticosteroids and sulicilates such as sulphasalazine and its derivatives.
  • anti-inflammatory drugs such as corticosteroids and sulicilates such as sulphasalazine and its derivatives.
  • immunosuppressive drugs such as cyclosporine A, mercaptopurin and azathropine are used.
  • these medicaments all have serious side effects.
  • a recent, successful development in the treatment of IBD consists in the use of compounds, blocking the working of TNF or its receptor. In that respect is the use of TNF antibodies one of the most promising new therapies.
  • Tumor necrosis factor ⁇ is a cytokine produced by numerous cell types, including monocytes and macrophages, that was originally identified based on its capacity to induce the necrosis of certain mouse tumors (see e.g., Old, L. (1985) Science 230:630-632). TNF ⁇ has been implicated in the pathophysiology of a variety of other human diseases and disorders, including sepsis, infections, autoimmune diseases, transplant rejection and graft-versus-host disease (see e.g. Moeller, A. et al. (1990) Cytokine2: 162-169; U.S. Patent No. 5,231 ,024 to Moeller et al ; European Patent Publication No.
  • Infliximab is a chimeric human-mouse monoclonal antibody of the IgGI K subclass, which specifically targets and irreversibly binds to TNF ⁇ on cell membranes and in blood.
  • Single intravenous doses, ranging from 5 to 20 mg/kg of the antibody infliximab resulted in a drastic clinical improvement in active Crohn's disease; it has been launched on the market to treat Crohn's disease in 1998.
  • the human monoclonal TNF ⁇ adalimumab was developed, which is currently tested in phase III clinical trials for the treatment of Crohn's.
  • Celltech developed Certolizumab pegol, which is a humanized monoclonal pegylated anti- TNF ⁇ antibody, which is currently also tested in phase III clinical trials for the treatment of Crohn's.
  • the antibodies are applied in a systemic way, mainly by subcutaneous injection.
  • Systemic administration of anti TNF- ⁇ antibody may result in rather serious unwanted effects, including headache, abscess, upper respiratory tract infection and fatigue.
  • the unwanted effects associated with systemic delivery could be solved by local delivery on the place of the inflammation.
  • a promising system for delivery of biological active compounds in the intestine has been disclosed in WO97/14806 whereby non-invasive gram positive bacteria such as lactic acid bacteria are used to deliver biological active compounds in the gut.
  • WO00/23471 discloses that this system can be used to deliver IL-10 to the ileum, whereby this strain can be used to treat IBD.
  • WO01/98461 discloses an alternative method for intestinal delivery using yeast.
  • a first aspect of the invention is the use of a genetically modified micro-organism, producing an anti TNF- ⁇ antibody, for the preparation of a medicament to treat IBD.
  • the term antibody includes, but is not limited to conventional antibodies, chimeric antibodies, single chain antibodies, bifunctional antibodies, bivalent antibodies, diabodies, and camelid antibodies, including antibody fragments such as VHHs; Preferably said antibody is a camelid antibody fragment (VHH; further called nanobody), even more preferably said antibody is a bivalent anti-TNF ⁇ nanobody.
  • said genetically modified micro-organisms is a lactic acid bacterium or a yeast. Delivery of biologically active polypeptides into the animal body by lactic acid bacteria has been disclosed in WO9714806; intestinal delivery of peptides by yeast has been described in WO0198461. However, none of these documents mention the delivery of antibodies or nanobodies in the intestine. Production, secretion and delivery in vivo of biological active antibodies or nanobodies is far from evident, as a correct folding and secretion of the antibody is required, and sufficient antibody is required to obtain a neutralizing activity.
  • said genetically modified micro-organism is a Lactococcus lactis strain, preferably said genetically a Lactococcus lactis ThyA mutant.
  • a specially preferred embodiment is the use of a Lactococcus lactis ThyA mutant, whereby the gene encoding the TNF- ⁇ antibody has been used to disrupt the THYA gene.
  • a yeast is be used to deliver the anti TNF- ⁇ antibody.
  • said yeast is Saccharomyces cerevisiae, even more preferably said yeast is Saccharomyces cerevisiae subsp. Boulardii.
  • IBD includes but is not limited to chronic colitis, ulcerative colitis and Crohn's disease.
  • IBD is chronic colitis.
  • Another aspect of the invention is a pharmaceutical composition for oral administration, comprising at least one genetically modified anti-TNF ⁇ VHH producing micro-organism.
  • said anti-TNF ⁇ VHH is a bivalent antibody.
  • the pharmaceutical composition may be liquid, comprising biological active micro-organisms, or it may be solid, comprising dried micro-organisms that can be reactivated when put in a suitable environment. Micro-organisms may be dried by any system, including freeze drying and spray drying.
  • Anti-TNF ⁇ VHH producing as used here doesn't imply that the micro-organism is producing the VHH in the pharmaceutical composition, but it means that the micro-organism is viable and can produce the VHH when placed in a suitable environment.
  • Micro-organisms may be coated to facilitate the delivery into the gastro-intestinal tract. Such coating are known to the person skilled in that art and was, amongst others, described by Huyghebaert et al. (2005).
  • the pharmaceutical composition may further comprise agents to improve the viability of the micro-organisms, such as, but not limited to trehalose.
  • the micro-organisms are selected from the group consisting of lactic acid bacteria and yeasts.
  • One preferred embodiment is a pharmaceutical composition, whereby the VHH producing micro-organism is a Lactococcus lactis, preferably a ThyA mutant.
  • Another preferred embodiment is a pharmaceutical composition, whereby the VHH producing micro-organism is a Lactobacillus sp. preferably a ThyA mutant.
  • said ThyA mutants are obtained by gene disruption, using the VHH encoding construct as insert.
  • Still another preferred embodiment is a pharmaceutical composition whereby the VHH producing micro-organism is Saccharomyces cerevisiae, preferably S. cerevisiae subspecies boulardii.
  • Another aspect of the invention is a method of preventing, treating and/or alleviating at least one disease or disorder of the gastro-intestinal tract, comprising administering to the gastro- intestinal tract an effective amount of an anti-TNF ⁇ VHH producing micro-organism.
  • said anti-TNF ⁇ VHH is a bivalent antibody.
  • the way of administering can be any way known to the person skilled in the art, and includes, but is not limited to oral and rectal administration.
  • the way of administering is oral administration.
  • said disease or disorder is a disease or disorder characterized by an imbalance in TNF ⁇ production, and can be treated by TNF ⁇ inactivating compounds such as TNF ⁇ antibodies.
  • said disease is an irritable bowel disease, including but not limited to chronic colitis, ulcerative colitis and Crohn's disease. Most preferably, said disease or disorder is chronic colitis.
  • said genetically modified micro-organisms is a lactic acid bacterium or a yeast.
  • said genetically modified micro-organism is a Lactococcus lactis strain, preferably said genetically a Lactococcus lactis ThyA mutant.
  • a specially preferred embodiment is a Lactococcus lactis ThyA mutant, whereby the gene encoding the TNF- ⁇ antibody has been used to disrupt the THYA gene.
  • genetically modified micro-organism is a Lactobacillus sp strain, preferably said genetically a Lactobacillus ThyA mutant.
  • a specially preferred embodiment is a Lactobacillus ThyA mutant, whereby the gene encoding the TNF- ⁇ antibody has been used to disrupt the THYA gene.
  • a yeast is the anti TNF- ⁇ antibody producing microorganism.
  • said yeast is Saccharomyces cerevisiae, even more preferably said yeast is Saccharomyces cerevisiae subsp. Boulardii. BRIEF DESCRIPTION OF THE FIGURES
  • Fig . 1 Time course of heterologous monovalent and bivalent nanobody NANO3F production by GM L. lactis (LL-NANO3F and LL-NANO3F-3F).
  • LL-pTREX1 vector control
  • LL-mlL10 L. lactis strain secreting murine interleukin-10.
  • A Western blot analysis of proteins secreted from the various strains revealed by anti-Myc Ab. Each lane on the blot represents 250 ⁇ l_ of L. lactis culture supernatant obtained after different periods of growth (2 x 10 7 CFU at time zero).
  • E. coli purified monovalent NANO3F (+) and bivalent NANO3F-3F (++) were used as positive control.
  • B Concentrations of secreted heterologous Myc-tagged proteins in culture supematants of LL-pTREX1 (O), LL-NANO3F ( ⁇ ), LL-NANO3F-3F (A) and LL-mlL10 (O), as determined by ELISA.
  • L. lactis secreted or E. coli purified monovalent and bivalent nanobody NANO3F are able to efficiently neutralize soluble and membrane bound TNF.
  • soluble TNF (20 IU/ml) was neutralized by NANO3F and NANO3F-3F in a 16 hour cytotoxicity assay using the WEHI 164 cl 13 cells in the presence of 1 ⁇ g/ml actinomycin D.
  • B L. lactis secreted and E. coli purified nanobodies NANO3F and NANO3F-3F were able to inhibit the cytotoxic effects of L929 expressing uncleavable membrane-bound TNF.
  • the gray bars represent wells where purified NANO3F or NANO3F-3F was added (total concentration of 250 ng/mL).
  • the black bars represent wells where 50 ⁇ l_ of filtered (0.22 ⁇ m) lactococcal supernatant was added.
  • the final concentration of LL-NANO3F and LL-NANO3F-3F was 250 ng/ml in each setting.
  • FIG. 3 Analysis of morbidity in chronic DSS colitis.
  • A-E Representative histology of the middle colon from healthy control mice (A) and mice with chronic DSS colitis either mock- treated (B) or treated with LL-pTREX1 (C), LL-NANO3F-3F (D) or LL-mlL10 (E).
  • F Statistical evaluation of the histological score of the middle colon. Bars represent the mean + SEM. White bar represent the healthy control group. Mice with DSS-induced chronic colitis were either mock-treated (hatched bars) or received LL-pTREX1 (black bar), LL-NANO3F-3F (red bar) or LL-mll_10 (gray bar). * * * and * * represent statistical significant differences in comparison with the mock-treated and the vector control groups of P ⁇ 0.001 and P ⁇ 0.01 , respectively.
  • Fig . 4 Analysis of morbidity in 20 weeks old 129Sv/Ev IL-10 " ' " mice. Each group received daily for 14 days 2 x 10 9 CFU LL-pTREX1 (vector control), LL-NANO3F-3F or LL-mlL10, except the mock treated group.
  • A-D Representative histology of the middle colon from I L- 10 " ' " mice which were mock-treated (A) or treated with LL-pTREX1 (B), LL-NANO3F-3F (C) or LL-mlL10 (D) (hematoxylin and eosin staining).
  • Fig. 7 Statistical evaluation of the histological score of the distal colon in mice with chronic DSS colitis. Bars represent the mean + SEM. White bars represent the healthy control group. Mice with DSS-induced chronic colitis received different L. lactis cultures for 28 days whereafter the mice were killed and analyzed immediately. The black bar represents the vector control LL-pTREX1 treated group, the hatched bar those that received LL-NANO3F (L. lactis secreting monovalent 3F), the gray bar those that received LL-NANO3F-3F (L. lactis secreting bivalent 3F-3F-. * * represents a statistical significant difference in comparison with the vector control LL-pTREX1 and the LL-NANO3F treated groups of P ⁇ 0.01.
  • Fig. 8 L. lactis secreted monovalent and bivalent Nanobody 3E (anti-human TNF Nanobody) are able to efficiently neutralize soluble human TNF. Soluble human TNF (different concentrations ranging from 30 till 3.3 Ill/ml) was neutralized by 3E and 3E-3E in a 16 hour cytotoxicity assay using L929s cells in the presence of 1 ⁇ g/ml actinomycin D. Cell survival was calculated relative to healthy cells The white bar represents wells where 50 ⁇ L of filtered (0.22 ⁇ m) lactococcal supernatant of the vector control LL-pTREX1 was added.
  • Black and gray bars represent wells where 50 ⁇ L of filtered (0.22 ⁇ m) lactococcal supernatant was added; containing L. lactis secreted 3E or 3E-3E, respectively.
  • the final concentration of L. lactis secreted 3E or 3E-3E was 250 ng/ml in each setting.
  • Bacteria and plasm ids The L. lactis strain MG 1363 was used throughout this study. Bacteria were cultured in GM 17 medium, i.e. M17 (Difco Laboratories, Detroit, Ml) supplemented with 0.5% glucose. Stock suspensions of all strains were stored at -20 0 C in 50% glycerol in GM 17. For intragastric inoculations, stock suspensions were diluted 200-fold in fresh GM 17 and incubated at 30 0 C. They reached a saturation density of 2 x 10 9 colony-forming units (CFU) per ml_ within 16 hours. Bacteria were harvested by centrifugation and concentrated 10-fold in BM9 medium. (Schotte, Steidler et al. 2000). For treatment, each mouse received 100 ⁇ l_ of this suspension daily by intragastric catheter.
  • GM 17 medium i.e. M17 (Difco Laboratories, Detroit, Ml) supplemented with 0.5% glucose.
  • Stock suspensions of all strains
  • Nanobody NANO3F (MW-15 kDa) was isolated.
  • the murine TNF specific Nanobody was converted into a bivalent format (coded NANO3F-3F, MW ⁇ 30 kDa) using the 12 aminoterminal residues of the llama lgG2a upper hinge sequence as a spacer.
  • MG1363 strains transformed with plasmids carrying the NANO3F or NANO3F-3F coding sequence were designated LL-NANO3F and LL-NANO3F-3F respectively.
  • LL-pTREX1 which is MG1363 containing the empty vector pTREXI , served as control.
  • Myc-tagged LL-NANO3F and LL-NANO3F-3F were quantified by direct adsorption of crude L. lactis supematants to ELISA plates (Maxisorp F96, Nunc, Rochester, NY) and subsequent detection with a specific mouse mAb against the Myc epitope (Sigma, St. Louis, MO).
  • ELISA plates Maxisorp F96, Nunc, Rochester, NY
  • a specific mouse mAb against the Myc epitope Sigma, St. Louis, MO
  • 3F-3F nanobodies secreted in vivo in colon tissue the entire colon was homogenized in PBS containing 1% BSA and sonicated. The 3F-3F nanobodies were measured in the colon supernatant with the nanobody quantification protocol.
  • the plate was washed 5 times and detection was performed by incubation with rabbit-polyclonal-anti-mouse- immunoglobulin-HRP (DAKO, 3, 000-fold diluted) for one hour at RT, and after washing plates were stained with ABTS/H2O2. The OD405nm was measured.
  • Anti-soluble and membrane-bound TNF bioassay DAKO, 3, 000-fold diluted
  • the inhibitory effect of the NANO3F and NANO3F-3F nanobodies on soluble mTNF (20 IU/mL) was measured in a 16 hour cytotoxicity assay using the mouse fibroblast WEHI 164 cl 13 cells in the presence of 1 ⁇ g/ml actinomycin D, as described. (Espevik and Nissen-Meyer 1986)
  • the effect of NANO3F and NANO3F-3F to counteract the cytotoxic effect of membrane-bound TNF was determined on the WEHI 164 cl 13 cells after adding L929 cells, expressing uncleavable, membrane-bound TNF to the cell culture (Decoster et al. 1998).
  • MF4/4 macrophages (Desmedt et al. 1998) were incubated with NANO3F-3F (100 ⁇ g/ml). After 1 hour cells were extensively washed (3X) in a sufficient volume of PBS to completely remove all nanobody present in solution. The cells were resuspended and incubated in the presence or absence of LPS for 4 hours. The cells were washed (1X) in PBS and after 4 hours of incubation, the supematans and cells were separated by centrifugation. To measure the soluble TNF release, the WEHI 164 cl 13 cells bioassay was used.
  • mice 1 1-week old female BALB/c mice were obtained form Charles River Laboratories (Sulzfeld, Germany). They were housed under SPF conditions. IL-10 knockout mice (129Sv/Ev IL-10 " ' " ) (Kuhn, Lohler et al. 1993) were housed and bred under SPF conditions. The IL-10 ' ' mice were used at 20 weeks of age, at which time chronic colitis had fully developed. All mice were fed standard laboratory feed and tap water ad libitum. The animal studies were approved by the Ethics Committee of the Department for Molecular Biomedical Research, Ghent University (File No. 04/02).
  • mice weighing approximately 21 g were induced to chronic colitis by four cycles of administration of 5% (w/v) DSS (40 kDa, Applichem, Darmstadt, Germany) in the drinking water, alternating with 10-day periods of recovery with normal drinking water. (Okayasu, Hatakeyama et al. 1990; Kojouharoff, Hans et al. 1997) Treatment was arbitrarily initiated at day 21 after the fourth cycle of DSS.
  • MPO activity in the middle colon tissue was measured as described (Bradley, Priebat et al. 1982). Pure human MPO was used as a standard (Calbiochem, San Diego, CA). Data are expressed as ⁇ g MPO/mm 2 colon tissue. Histological analysis
  • the colon was removed, cleaned and opened longitudinally. A segment of 1 cm was taken from the middle part of the colon, embedded in paraffin and sectioned longitudinally. Three sections of 4 ⁇ m were cut at 200 ⁇ m intervals and stained with hematoxylin/eosin. Colon sections were numbered randomly and interpreted semiquantitative ⁇ in a blinded manner by a pathologist. The histological score is the sum of the epithelial damage and lymphoid infiltration, each ranging from 0 to 4 as described (Kojouharoff, Hans et al. 1997).
  • Example 1 Anti-TNF- ⁇ VHH production by L. lactis in vitro L. lactis was transformed with the plasmids encoding NANO3F and NANO3F-3F. The production of the antibodies was checked by Western blot and ELISA, using a strain transformed with the empty plasmid pTREX and an IL10 producing strain as reference. The results are shown in Figure 1. NANO3F-3F is produced by L. lactis in similar or higher amounts than NANO3F. The amount produced is significantly higher than for IL10.
  • Example 2 LL-NANO3F-3F is bioactive and inhibits both soluble and membrane bound
  • NANO3F and NANO3F-3F nanobodies produced by L. lactis on soluble mTNF was measured in a cytotoxicity assay using the mouse fibroblast WEHI 164 cl 13 cells as described by Espevik and Nissen-Meyer (1986).
  • E. coli produced NANO3F and NANO3F-3F was used as a positive reference. Both the purified nonobodies as well as the nanobodies produced by L. lactis can neutralize the soluble TNF.
  • Example 3 LL-NANO3F-3F effect in vivo on established DSS induced chronic colitis Chronic colitis was induced by DSS as described in materials and methods. Mice were daily treated with 2 10 9 colony forming units (cfu) of either LL-pTREX1 , LL-NANO3F-3F, or LL- mlL10. A mock treatment, and healthy mice ("watercontrol") were used as additional control. The effect of the nano3F-3F nanobody deliverd by L. lactis is comparable to the protection obtained by the in situ produced IL-10 ( Figure 3).
  • Example 4 LL-NANO3F-3F effect in vivo on established IL-10 v enterocolitis
  • Each group received daily for 14 days 2 x 10 9 CFU of either LL- pTREXI (vector control), LL-NANO3F-3F or LL-mlL10, except the mock treated group.
  • the results are summarized in Figure 4. Both the myelperoxidase assay as well as the histological score indicate a significant protection in the LL-NANO3F-3F treated mice.
  • mice were treated intragastically over a period of 14 days with LL-NANO3F-3F, using intraperitoneal injection of purified nanobody as control. Anti-Nanobody antibody levels were measured in the mouse seum. The results are shown in Figure 5. While interperitoneal injection of NANO3F-3F is giving a clear immune response, the treatment with LL-NANO3F-3F is not immunogenic and proofs to be safe in that respect.
  • Example 6 effect of NANO3F-3F on LPS induction of proinflammatory cytokines
  • MF4/4 macrophages (Desmedt et al. 1998) were incubated with NANO3F-3F. The cells were washed and then incubated with LPS. Soluble TNF release was measured using the WEHI 164 cl 13 cell toxicity assay. The results are shown in Figure 6. Pretreatment of the macrophages with NANO3F-3F naobody gives a clear protection against LPS induced soluble TNF production.
  • Example 7 bivalent antibodies perform surprisingly better than monovalent antibodies Although bivalent antibodies are larger than monovalent ones, it doesn't affect the production in Lactococcus. The production of bivalent antibodies is at least as good if not better than for monovalent antibodies (Fig. 1 ). However, even more important is the efficacy of the bivalent antibodies. From the in vivo experiments, it is obvious that a monovalent anti-TNF antibody results only in a marginal, non-significant improvement of the histological score, whereas administration of a L. lactis secreting bivalent antibodies results in a significant improvement (Figure 7). Indeed, the neutralizing effect of bivalent antibodies is, for a comparable concentration of protein, more pronounced than that of monovalent antibodies. As long as no complete neutralization is reached, the improvement is more than a factor 2, indicating the effect is not purely due to the double valence of the nanobody ( Figure 8) REFERENCES
  • Lactococcus lactis Enzyme Microb Technol 27(10): 761-765. van Asseldonk, M., G. Rutten, et al. (1990). "Cloning of usp45, a gene encoding a secreted protein from Lactococcus lactis subsp. lactis MG1363.” Gene 95(1 ): 155-60. Waterfield, N. R., R. W. Le Page, et al. (1995). "The isolation of lactococcal promoters and their use in investigating bacterial luciferase synthesis in Lactococcus lactis.” Gene

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Mycology (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Microbiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Botany (AREA)
  • General Chemical & Material Sciences (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Biotechnology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medical Informatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention relates to a novel treatment of chronic enterocolitis. More specifically, the invention relates to the production of a medicament comprising anti-TNFalpha antibody producing lactic acid bacteria, and the use of this medicament in the treatment of chronic enterocolitis.

Description

NOVEL TREATMENT OF CHRONIC ENTEROCOLITIS
The present invention relates to a novel treatment of chronic enterocolitis. More specifically, the invention relates to the production of a medicament comprising anti-TNFα producing lactic acid bacteria, and the use of this medicament in the treatment of chronic enterocolitis. Inflammatory bowel disease (IBD) refers to a group of gastrointestinal disorders characterized by a chronic non-specific inflammation of portions of the gastrointestinal tract. The most prominent examples of IBD in humans are ulcerative colitis (UC) and Crohn's disease (CD). The etiology or etiologies of IBD are unclear. IBD diseases appear to result from the unrestrained activation of an inflammatory response in the intestine. This inflammatory cascade is thought to be perpetuated through actions of proinflammatory cytokines and selective activation of lymphocyte subsets. UC and CD are associated with many symptoms and complications, including growth retardation in children, rectal prolaps, blood in stools, wasting, iron deficiency and anemia. UC refers to a chronic, non-specific, inflammatory and ulcerative disease having manifestations primarily in the colonic mucosa. It is frequently characterized by bloody diarrhea, abdominal cramps, blood and mucus in the stoll, malaise, fever, anemia, anorexia, weight loss, leukocytosis, hypoalbuminemia and an elevated erythrocyte sedimentation rate.
Crohn's disease shares many features in common with ulcerative colitis. Crohn's disease is distinguishable in that lesions tend to be sharply demarcated from adjacent normal bowel, in contrast to the lesions of ulcerative colitis which are fairly diffuse. Additionally, Crohn's disease predominantly afflicts the ileum (ileitis) and the ileum and colon (ileocolitis). In some cases, the colon alone is diseased (granulomatous colitis) and sometimes the entire small bowel is involved. Colon cancer is a known complication of chronic IBD. It is increasingly common in those patients who have IBD for many years. The risk for cancer begins to rise significantly after eight to ten years of IBD, making a fast and efficient treatment of IBD even more important. The most commonly used medication to treat IBD includes anti-inflammatory drugs such as corticosteroids and sulicilates such as sulphasalazine and its derivatives. For people that not respond to these drugs, immunosuppressive drugs such as cyclosporine A, mercaptopurin and azathropine are used. However, these medicaments all have serious side effects. A recent, successful development in the treatment of IBD consists in the use of compounds, blocking the working of TNF or its receptor. In that respect is the use of TNF antibodies one of the most promising new therapies. Tumor necrosis factor α (TNFα) is a cytokine produced by numerous cell types, including monocytes and macrophages, that was originally identified based on its capacity to induce the necrosis of certain mouse tumors (see e.g., Old, L. (1985) Science 230:630-632). TNFα has been implicated in the pathophysiology of a variety of other human diseases and disorders, including sepsis, infections, autoimmune diseases, transplant rejection and graft-versus-host disease (see e.g. Moeller, A. et al. (1990) Cytokine2: 162-169; U.S. Patent No. 5,231 ,024 to Moeller et al ; European Patent Publication No. 260 610 (B1 ) by Moeller, A. et al.; Vasilli. P.(1992) Annu. Rev.lmmunol. 10:41 1-452; Tracey, K.J. and Cerami, A. (1994) Annu. Rev. Med. 45:491-503). Because of the harmful role of human TNFα (hTNFα) in a variety of human disorders, therapeutic strategies have been designed to inhibit or counteract hTNFα activity. In particular, antibodies that bind to, and neutralize, hTNFα have been sought as a means to inhibit hTNFα activity.
Several antibody preparations have been tested for the treatment of IBD. Although polyclonal antibodies have been tested in phase Il clinical tests, monoclonal antibodies are clearly preferred. Infliximab is a chimeric human-mouse monoclonal antibody of the IgGI K subclass, which specifically targets and irreversibly binds to TNFα on cell membranes and in blood. Single intravenous doses, ranging from 5 to 20 mg/kg of the antibody infliximab resulted in a drastic clinical improvement in active Crohn's disease; it has been launched on the market to treat Crohn's disease in 1998. To solve possible problems linked to chimeric antibodies, the human monoclonal TNFα adalimumab was developed, which is currently tested in phase III clinical trials for the treatment of Crohn's. To improve the half-life time of the antibody in patients, Celltech developed Certolizumab pegol, which is a humanized monoclonal pegylated anti- TNFα antibody, which is currently also tested in phase III clinical trials for the treatment of Crohn's.
However, in all those cases, the antibodies are applied in a systemic way, mainly by subcutaneous injection. Systemic administration of anti TNF-α antibody may result in rather serious unwanted effects, including headache, abscess, upper respiratory tract infection and fatigue. The unwanted effects associated with systemic delivery could be solved by local delivery on the place of the inflammation. A promising system for delivery of biological active compounds in the intestine has been disclosed in WO97/14806 whereby non-invasive gram positive bacteria such as lactic acid bacteria are used to deliver biological active compounds in the gut. WO00/23471 discloses that this system can be used to deliver IL-10 to the ileum, whereby this strain can be used to treat IBD. WO01/98461 discloses an alternative method for intestinal delivery using yeast. However, although the delivery of biologically active compounds is described, these documents do not teach the delivery of antibodies in the intestine. The in situ production of active antibodies in the intestine is far from straightforward, as both folding and secretion of the antibody are critical. Especially, the stabilization of the structure by sulfur bridges may cause problems for the production of antibodies in bacteria or yeasts. Moreover, whereas cytokines like IL-10 fulfill a catalytic function, TNF antibodies need to be produced in a sufficient amount to inactivate the endogenous produced TNF. Surprisingly, we found that the local delivery of anti TNF-α antibody by a genetically engineered micro-organism can be used in an efficient way to treat IBD.
A first aspect of the invention is the use of a genetically modified micro-organism, producing an anti TNF-α antibody, for the preparation of a medicament to treat IBD. The term antibody, as used here, includes, but is not limited to conventional antibodies, chimeric antibodies, single chain antibodies, bifunctional antibodies, bivalent antibodies, diabodies, and camelid antibodies, including antibody fragments such as VHHs; Preferably said antibody is a camelid antibody fragment (VHH; further called nanobody), even more preferably said antibody is a bivalent anti-TNFα nanobody. Bivalent antibodies have the advantage to inhibit TNF binding to its receptor, in a significantly more efficient fashion than does monovalent (EC50 = 16 pM and 6.7 nM, respectively). Surprisingly we found that the production of bivalent antibodies in Lactococcus is as high or even higher than that of monovalent antibodies. Preferably, said genetically modified micro-organisms is a lactic acid bacterium or a yeast. Delivery of biologically active polypeptides into the animal body by lactic acid bacteria has been disclosed in WO9714806; intestinal delivery of peptides by yeast has been described in WO0198461. However, none of these documents mention the delivery of antibodies or nanobodies in the intestine. Production, secretion and delivery in vivo of biological active antibodies or nanobodies is far from evident, as a correct folding and secretion of the antibody is required, and sufficient antibody is required to obtain a neutralizing activity. In one preferred embodiment said genetically modified micro-organism is a Lactococcus lactis strain, preferably said genetically a Lactococcus lactis ThyA mutant. A specially preferred embodiment is the use of a Lactococcus lactis ThyA mutant, whereby the gene encoding the TNF-α antibody has been used to disrupt the THYA gene. In another preferred embodiment, a yeast is be used to deliver the anti TNF-α antibody. Preferably said yeast is Saccharomyces cerevisiae, even more preferably said yeast is Saccharomyces cerevisiae subsp. Boulardii.
IBD, as used here, includes but is not limited to chronic colitis, ulcerative colitis and Crohn's disease. Preferably, IBD is chronic colitis. Another aspect of the invention is a pharmaceutical composition for oral administration, comprising at least one genetically modified anti-TNFα VHH producing micro-organism. Preferably, said anti-TNFα VHH is a bivalent antibody. The pharmaceutical composition may be liquid, comprising biological active micro-organisms, or it may be solid, comprising dried micro-organisms that can be reactivated when put in a suitable environment. Micro-organisms may be dried by any system, including freeze drying and spray drying. "Anti-TNFα VHH producing" as used here doesn't imply that the micro-organism is producing the VHH in the pharmaceutical composition, but it means that the micro-organism is viable and can produce the VHH when placed in a suitable environment. Micro-organisms may be coated to facilitate the delivery into the gastro-intestinal tract. Such coating are known to the person skilled in that art and was, amongst others, described by Huyghebaert et al. (2005). The pharmaceutical composition may further comprise agents to improve the viability of the micro-organisms, such as, but not limited to trehalose. Preferably, the micro-organisms are selected from the group consisting of lactic acid bacteria and yeasts. One preferred embodiment is a pharmaceutical composition, whereby the VHH producing micro-organism is a Lactococcus lactis, preferably a ThyA mutant. Another preferred embodiment is a pharmaceutical composition, whereby the VHH producing micro-organism is a Lactobacillus sp. preferably a ThyA mutant. Preferably, said ThyA mutants are obtained by gene disruption, using the VHH encoding construct as insert. Still another preferred embodiment is a pharmaceutical composition whereby the VHH producing micro-organism is Saccharomyces cerevisiae, preferably S. cerevisiae subspecies boulardii.
Another aspect of the invention is a method of preventing, treating and/or alleviating at least one disease or disorder of the gastro-intestinal tract, comprising administering to the gastro- intestinal tract an effective amount of an anti-TNFα VHH producing micro-organism. Preferably, said anti-TNFα VHH is a bivalent antibody. The way of administering can be any way known to the person skilled in the art, and includes, but is not limited to oral and rectal administration. Preferably, the way of administering is oral administration. Preferably, said disease or disorder is a disease or disorder characterized by an imbalance in TNFα production, and can be treated by TNFα inactivating compounds such as TNFα antibodies. Even more preferably, said disease is an irritable bowel disease, including but not limited to chronic colitis, ulcerative colitis and Crohn's disease. Most preferably, said disease or disorder is chronic colitis. Preferably, said genetically modified micro-organisms is a lactic acid bacterium or a yeast. In one preferred embodiment said genetically modified micro-organism is a Lactococcus lactis strain, preferably said genetically a Lactococcus lactis ThyA mutant. A specially preferred embodiment is a Lactococcus lactis ThyA mutant, whereby the gene encoding the TNF-α antibody has been used to disrupt the THYA gene. In another preferred embodiment genetically modified micro-organism is a Lactobacillus sp strain, preferably said genetically a Lactobacillus ThyA mutant. A specially preferred embodiment is a Lactobacillus ThyA mutant, whereby the gene encoding the TNF-α antibody has been used to disrupt the THYA gene.
In another preferred embodiment, a yeast is the anti TNF-α antibody producing microorganism. Preferably said yeast is Saccharomyces cerevisiae, even more preferably said yeast is Saccharomyces cerevisiae subsp. Boulardii. BRIEF DESCRIPTION OF THE FIGURES
Fig . 1 . Time course of heterologous monovalent and bivalent nanobody NANO3F production by GM L. lactis (LL-NANO3F and LL-NANO3F-3F). LL-pTREX1 : vector control; LL-mlL10: L. lactis strain secreting murine interleukin-10. (A) Western blot analysis of proteins secreted from the various strains revealed by anti-Myc Ab. Each lane on the blot represents 250 μl_ of L. lactis culture supernatant obtained after different periods of growth (2 x 107 CFU at time zero).
E. coli purified monovalent NANO3F (+) and bivalent NANO3F-3F (++) were used as positive control. (B) Concentrations of secreted heterologous Myc-tagged proteins in culture supematants of LL-pTREX1 (O), LL-NANO3F (■), LL-NANO3F-3F (A) and LL-mlL10 (O), as determined by ELISA.
Fig . 2. L. lactis secreted or E. coli purified monovalent and bivalent nanobody NANO3F are able to efficiently neutralize soluble and membrane bound TNF. (A) soluble TNF (20 IU/ml) was neutralized by NANO3F and NANO3F-3F in a 16 hour cytotoxicity assay using the WEHI 164 cl 13 cells in the presence of 1 μg/ml actinomycin D. (B) L. lactis secreted and E. coli purified nanobodies NANO3F and NANO3F-3F were able to inhibit the cytotoxic effects of L929 expressing uncleavable membrane-bound TNF. The gray bars represent wells where purified NANO3F or NANO3F-3F was added (total concentration of 250 ng/mL). The black bars represent wells where 50 μl_ of filtered (0.22 μm) lactococcal supernatant was added. The final concentration of LL-NANO3F and LL-NANO3F-3F was 250 ng/ml in each setting.
Fig. 3. Analysis of morbidity in chronic DSS colitis. (A-E) Representative histology of the middle colon from healthy control mice (A) and mice with chronic DSS colitis either mock- treated (B) or treated with LL-pTREX1 (C), LL-NANO3F-3F (D) or LL-mlL10 (E). (F) Statistical evaluation of the histological score of the middle colon. Bars represent the mean + SEM. White bar represent the healthy control group. Mice with DSS-induced chronic colitis were either mock-treated (hatched bars) or received LL-pTREX1 (black bar), LL-NANO3F-3F (red bar) or LL-mll_10 (gray bar). * * * and * * represent statistical significant differences in comparison with the mock-treated and the vector control groups of P < 0.001 and P < 0.01 , respectively.
Fig . 4 Analysis of morbidity in 20 weeks old 129Sv/Ev IL-10"'" mice. Each group received daily for 14 days 2 x 109 CFU LL-pTREX1 (vector control), LL-NANO3F-3F or LL-mlL10, except the mock treated group. (A-D) Representative histology of the middle colon from I L- 10"'" mice which were mock-treated (A) or treated with LL-pTREX1 (B), LL-NANO3F-3F (C) or LL-mlL10 (D) (hematoxylin and eosin staining). Statistical evaluation of the MPO levels per mm2 colon tissue (A) and histological score of the distal colon (B). Bars represent the mean + SEM. Hatched bars represent the 129Sv/Ev I L- 107" mice that were mock treated, the black bars represent the 129Sv/EV IL-I O''' mice that received the vector control LL-pTREX1 , the red bars represent the 129Sv/EV IL-10"'" mice that received LL-NANO3F-3F and the gray bars represent the 129Sv/EV IL-107 mice that were treated with LL-mlL10. * and * * represent a statistically significant difference in comparison with the vector control group of P < 0.05 and P < 0.01 , respectively.
Fig . 5 Presence of nanobody specific antibodies was assessed by ELISA.
Fig . 6 Pretreatment with NANO3F-3F confers LPS irresponsiveness to Mf4/4 macrophages. WEHI164 cl 13 cells can still be killed with 5 IU TNF
Fig. 7 Statistical evaluation of the histological score of the distal colon in mice with chronic DSS colitis. Bars represent the mean + SEM. White bars represent the healthy control group. Mice with DSS-induced chronic colitis received different L. lactis cultures for 28 days whereafter the mice were killed and analyzed immediately. The black bar represents the vector control LL-pTREX1 treated group, the hatched bar those that received LL-NANO3F (L. lactis secreting monovalent 3F), the gray bar those that received LL-NANO3F-3F (L. lactis secreting bivalent 3F-3F-. * * represents a statistical significant difference in comparison with the vector control LL-pTREX1 and the LL-NANO3F treated groups of P < 0.01.
Fig. 8 L. lactis secreted monovalent and bivalent Nanobody 3E (anti-human TNF Nanobody) are able to efficiently neutralize soluble human TNF. Soluble human TNF (different concentrations ranging from 30 till 3.3 Ill/ml) was neutralized by 3E and 3E-3E in a 16 hour cytotoxicity assay using L929s cells in the presence of 1 μg/ml actinomycin D. Cell survival was calculated relative to healthy cells The white bar represents wells where 50 μL of filtered (0.22 μm) lactococcal supernatant of the vector control LL-pTREX1 was added. Black and gray bars represent wells where 50 μL of filtered (0.22 μm) lactococcal supernatant was added; containing L. lactis secreted 3E or 3E-3E, respectively. The final concentration of L. lactis secreted 3E or 3E-3E was 250 ng/ml in each setting.
EXAMPLES
Material and methods to the examples Bacteria and plasm ids The L. lactis strain MG 1363 was used throughout this study. Bacteria were cultured in GM 17 medium, i.e. M17 (Difco Laboratories, Detroit, Ml) supplemented with 0.5% glucose. Stock suspensions of all strains were stored at -200C in 50% glycerol in GM 17. For intragastric inoculations, stock suspensions were diluted 200-fold in fresh GM 17 and incubated at 300C. They reached a saturation density of 2 x 109 colony-forming units (CFU) per ml_ within 16 hours. Bacteria were harvested by centrifugation and concentrated 10-fold in BM9 medium. (Schotte, Steidler et al. 2000). For treatment, each mouse received 100 μl_ of this suspension daily by intragastric catheter.
Identification and formatting of an anti-muήneTNF Nanobody
The generation of anti-murine TNF nanobody was essentially carried out as described in
WO2004041862. After immunizing llamas with mTNF, subsequent cloning of the VHH repertoire and panning, Nanobody NANO3F (MW-15 kDa) was isolated. The murine TNF specific Nanobody was converted into a bivalent format (coded NANO3F-3F, MW~30 kDa) using the 12 aminoterminal residues of the llama lgG2a upper hinge sequence as a spacer. The cDNA of the NANO3F and NANO3F-3F, extended at their 3' ends with the sequence encoding the HisG and Myc-tag, were fused to the Usp45 secretion signal (van Asseldonk, Rutten et al. 1990) downstream of the lactococcal P1 promotor (Waterfield, Le Page et al. 1995) and expressed in MG 1363 (details of plasmid construction can be obtained from the authors). MG1363 strains transformed with plasmids carrying the NANO3F or NANO3F-3F coding sequence were designated LL-NANO3F and LL-NANO3F-3F respectively. LL-pTREX1 , which is MG1363 containing the empty vector pTREXI , served as control.
Quantification of nanobody in L lactis medium.
Myc-tagged LL-NANO3F and LL-NANO3F-3F were quantified by direct adsorption of crude L. lactis supematants to ELISA plates (Maxisorp F96, Nunc, Rochester, NY) and subsequent detection with a specific mouse mAb against the Myc epitope (Sigma, St. Louis, MO). For quantification of 3F-3F nanobodies secreted in vivo in colon tissue, the entire colon was homogenized in PBS containing 1% BSA and sonicated. The 3F-3F nanobodies were measured in the colon supernatant with the nanobody quantification protocol.
Measurement of anti-Nanobody antibody levels in mouse serum. Mice were injected intraperitoneal^ with 100 μg Nanobody, or intragastically with LL-NANO3F- 3F, daily over a 14 day-period and were subsequently bled. We coated Nanobody at a concentration of 10 μg/ml in microtiterplates (NUNC Maxisorb) overnight at 4°C. The plate was washed 5 times with PBS-Tween and blocked for 2 hours at RT with PBS-1 % casein. The samples were applied at a 1/50 dilution in PBS for 2 hours at RT. The plate was washed 5 times and detection was performed by incubation with rabbit-polyclonal-anti-mouse- immunoglobulin-HRP (DAKO, 3, 000-fold diluted) for one hour at RT, and after washing plates were stained with ABTS/H2O2. The OD405nm was measured. Anti-soluble and membrane-bound TNF bioassay
The inhibitory effect of the NANO3F and NANO3F-3F nanobodies on soluble mTNF (20 IU/mL) was measured in a 16 hour cytotoxicity assay using the mouse fibroblast WEHI 164 cl 13 cells in the presence of 1 μg/ml actinomycin D, as described. (Espevik and Nissen-Meyer 1986) The effect of NANO3F and NANO3F-3F to counteract the cytotoxic effect of membrane-bound TNF was determined on the WEHI 164 cl 13 cells after adding L929 cells, expressing uncleavable, membrane-bound TNF to the cell culture (Decoster et al. 1998).
Stimulation of Macrophages with LPS To measure the effect of NANO3F-3F on the induction of proinflammatory cytokines by LPS, MF4/4 macrophages (Desmedt et al. 1998) were incubated with NANO3F-3F (100 μg/ml). After 1 hour cells were extensively washed (3X) in a sufficient volume of PBS to completely remove all nanobody present in solution. The cells were resuspended and incubated in the presence or absence of LPS for 4 hours. The cells were washed (1X) in PBS and after 4 hours of incubation, the supematans and cells were separated by centrifugation. To measure the soluble TNF release, the WEHI 164 cl 13 cells bioassay was used.
Animals
1 1-week old female BALB/c mice were obtained form Charles River Laboratories (Sulzfeld, Germany). They were housed under SPF conditions. IL-10 knockout mice (129Sv/Ev IL-10"'") (Kuhn, Lohler et al. 1993) were housed and bred under SPF conditions. The IL-10 '' mice were used at 20 weeks of age, at which time chronic colitis had fully developed. All mice were fed standard laboratory feed and tap water ad libitum. The animal studies were approved by the Ethics Committee of the Department for Molecular Biomedical Research, Ghent University (File No. 04/02).
Induction of chronic colitis by DSS
Mice weighing approximately 21 g were induced to chronic colitis by four cycles of administration of 5% (w/v) DSS (40 kDa, Applichem, Darmstadt, Germany) in the drinking water, alternating with 10-day periods of recovery with normal drinking water. (Okayasu, Hatakeyama et al. 1990; Kojouharoff, Hans et al. 1997) Treatment was arbitrarily initiated at day 21 after the fourth cycle of DSS.
Myeloperoxidase (MPO) assay MPO activity in the middle colon tissue was measured as described (Bradley, Priebat et al. 1982). Pure human MPO was used as a standard (Calbiochem, San Diego, CA). Data are expressed as μg MPO/mm2 colon tissue. Histological analysis
For histological analysis, the colon was removed, cleaned and opened longitudinally. A segment of 1 cm was taken from the middle part of the colon, embedded in paraffin and sectioned longitudinally. Three sections of 4 μm were cut at 200 μm intervals and stained with hematoxylin/eosin. Colon sections were numbered randomly and interpreted semiquantitative^ in a blinded manner by a pathologist. The histological score is the sum of the epithelial damage and lymphoid infiltration, each ranging from 0 to 4 as described (Kojouharoff, Hans et al. 1997).
Statistical analysis
All data are expressed as mean + SEM Parametric data were analyzed with a 1-way analysis of variance followed by a Dunnett multiple comparisons posttest. Nonparametric data (scoring) were analyzed with a Mann-Whitney test. Example 1 : Anti-TNF-α VHH production by L. lactis in vitro L. lactis was transformed with the plasmids encoding NANO3F and NANO3F-3F. The production of the antibodies was checked by Western blot and ELISA, using a strain transformed with the empty plasmid pTREX and an IL10 producing strain as reference. The results are shown in Figure 1. NANO3F-3F is produced by L. lactis in similar or higher amounts than NANO3F. The amount produced is significantly higher than for IL10.
Example 2: LL-NANO3F-3F is bioactive and inhibits both soluble and membrane bound
TNF-α
The inhibitory effect of the NANO3F and NANO3F-3F nanobodies, produced by L. lactis on soluble mTNF was measured in a cytotoxicity assay using the mouse fibroblast WEHI 164 cl 13 cells as described by Espevik and Nissen-Meyer (1986). E. coli produced NANO3F and NANO3F-3F was used as a positive reference. Both the purified nonobodies as well as the nanobodies produced by L. lactis can neutralize the soluble TNF. (Figure 2A) The effect of NANO3F and NANO3F-3F to counteract the cytotoxic effect of membrane-bound TNF was determined on the WEHI 164 cl 13 cells after adding L929 cells, expressing uncleavable, membrane-bound TNF to the cell culture (Decoster et al. 1998). The effect of NANO3F is less pronounced, both with the purified form and the L. lactis produced form, but the effect of the NANO3F-3F nanobody is clear in both cases (Figure 2B)
Example 3: LL-NANO3F-3F effect in vivo on established DSS induced chronic colitis Chronic colitis was induced by DSS as described in materials and methods. Mice were daily treated with 2 109 colony forming units (cfu) of either LL-pTREX1 , LL-NANO3F-3F, or LL- mlL10. A mock treatment, and healthy mice ("watercontrol") were used as additional control. The effect of the nano3F-3F nanobody deliverd by L. lactis is comparable to the protection obtained by the in situ produced IL-10 (Figure 3).
Example 4: LL-NANO3F-3F effect in vivo on established IL-10 v enterocolitis To evaluate the protection in IL-10 '' enterocolitis, morbidity in 20 weeks old 129Sv/Ev IL-10 '' treated and untreated mice. Each group received daily for 14 days 2 x 109 CFU of either LL- pTREXI (vector control), LL-NANO3F-3F or LL-mlL10, except the mock treated group. The results are summarized in Figure 4. Both the myelperoxidase assay as well as the histological score indicate a significant protection in the LL-NANO3F-3F treated mice.
Example 5: immunogenicity of NANO3F-3F
To evaluate a possible adverse immunogenic effect of LL-NANO3F-3F, mice were treated intragastically over a period of 14 days with LL-NANO3F-3F, using intraperitoneal injection of purified nanobody as control. Anti-Nanobody antibody levels were measured in the mouse seum. The results are shown in Figure 5. While interperitoneal injection of NANO3F-3F is giving a clear immune response, the treatment with LL-NANO3F-3F is not immunogenic and proofs to be safe in that respect.
Example 6: effect of NANO3F-3F on LPS induction of proinflammatory cytokines To measure the effect of NANO3F-3F on the induction of proinflammatory cytokines by LPS, MF4/4 macrophages (Desmedt et al. 1998) were incubated with NANO3F-3F. The cells were washed and then incubated with LPS. Soluble TNF release was measured using the WEHI 164 cl 13 cell toxicity assay. The results are shown in Figure 6. Pretreatment of the macrophages with NANO3F-3F naobody gives a clear protection against LPS induced soluble TNF production.
Example 7: bivalent antibodies perform surprisingly better than monovalent antibodies Although bivalent antibodies are larger than monovalent ones, it doesn't affect the production in Lactococcus. The production of bivalent antibodies is at least as good if not better than for monovalent antibodies (Fig. 1 ). However, even more important is the efficacy of the bivalent antibodies. From the in vivo experiments, it is obvious that a monovalent anti-TNF antibody results only in a marginal, non-significant improvement of the histological score, whereas administration of a L. lactis secreting bivalent antibodies results in a significant improvement (Figure 7). Indeed, the neutralizing effect of bivalent antibodies is, for a comparable concentration of protein, more pronounced than that of monovalent antibodies. As long as no complete neutralization is reached, the improvement is more than a factor 2, indicating the effect is not purely due to the double valence of the nanobody (Figure 8) REFERENCES
Bradley, P. P., D. A. Priebat, et al. (1982). "Measurement of cutaneous inflammation: estimation of neutrophil content with an enzyme marker." J Invest Dermatol 78(3): 206-
9. Espevik, T. and J. Nissen-Meyer (1986). "A highly sensitive cell line, WEHI 164 clone 13, for measuring cytotoxic factor/tumor necrosis factor from human monocytes." J Immunol
Methods 95(1 ): 99-105. Huyghebaert, N., A. Vermeire, S. Neirynck, L. Steidler, E. Remaut, and J. P. Remon. (2005)
"Development of an enteric-coated formulation containing freeze-dried, viable recombinant Lactococcus lactis for the ileal mucosal delivery of human interleukin-10."
Eur J Pharm Biopharm 60(3): 349-59 Kojouharoff, G., W. Hans, et al. (1997). "Neutralization of tumour necrosis factor (TNF) but not of IL-1 reduces inflammation in chronic dextran sulphate sodium-induced colitis in mice." Clin Exp Immunol 107(2): 353-8. Kuhn, R., J. Lohler, et al. (1993). "lnterleukin-10-deficient mice develop chronic enterocolitis."
Cell 75(2): 263-74. Okayasu, I., S. Hatakeyama, et al. (1990). "A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice." Gastroenterology 98(3): 694-
702. Schotte, L., L. Steidler, et al. (2000). "Secretion of biologically active murine interleukin-10 by
Lactococcus lactis." Enzyme Microb Technol 27(10): 761-765. van Asseldonk, M., G. Rutten, et al. (1990). "Cloning of usp45, a gene encoding a secreted protein from Lactococcus lactis subsp. lactis MG1363." Gene 95(1 ): 155-60. Waterfield, N. R., R. W. Le Page, et al. (1995). "The isolation of lactococcal promoters and their use in investigating bacterial luciferase synthesis in Lactococcus lactis." Gene
165(1 ): 9-15.

Claims

1. The use of an anti-TNFα VHH producing micro-organism for the preparation of a medicament to treat IBD
2. The use according to claim 1 , whereby said anti-TNFα VHH is a bivalent antibody.
3. The use of an anti-TNFα VHH producing micro-organism according to claim 1 or 2, whereby said micro-organism is a lactic acid bacterium.
4. The use according to claim 3, whereby said lactic acid bacterium is Lactococcus lactis.
5. The use according to claim 3, whereby said lactic acid bacterium is a Lactobacillus sp.
6. The use of an anti-TNFα VHH producing micro-organism according to claim 1 or 2, whereby said micro-organism is a yeast.
7. The use of an anti-TNFα VHH producing micro-organism according to claim 6, whereby said yeast is Saccharomyces sp.
8. The use according to any of the preceding claims, whereby said IBD is chronic colitis.
9. The use according to any of the claims 1 to 7, whereby said IBD is Crohn's disease.
10. The use according to any of the claims 1 to 7, whereby said IBD is ulcerative colitis.
1 1. A pharmaceutical composition for oral administration, comprising at least one anti- TNFα VHH producing micro-organism.
12. A pharmaceutical composition for oral administration according to claim 1 1 , whereby said anti-TNFα VHH is a bivalent antibody.
13. A pharmaceutical composition according to claim 1 1 or 12, whereby said microorganism is selected from the group consisting of lactic acid bacteria and yeasts.
14. A pharmaceutical composition according to claim 13, whereby said lactic acid bacterium is Lactococcus lactis.
15. A pharmaceutical composition according to claim 13, whereby said lactic acid bacterium is Lactobacillus sp.
16. A pharmaceutical composition according to claim 13, whereby said yeast is Saccharomyces cerevisiae.
17. Method of preventing, treating and/or alleviating at least one disease or disorder of the gastro-intestinal tract, comprising administering to the gastro-intestinal tract an effective amount of an anti-TNFα VHH producing micro-organism.
18. A method according to claim 17, whereby said anti-TNFα VHH is a bivalent antibody.
19. A method according to claim 17 or 18, whereby said administering is oral administering.
20. A method according to claim any of the claims 17 - 19, whereby said disease or disorder is irritable bowel disease.
21. A method according to claim 20, whereby said irritable bowel disease is selected from the group consisting of chronic colitis, Crohn's disease and ulcerative colitis.
22. A method according to any of the claims 17-21 , whereby said micro-organism is selected from the group consisting of lactic acid bacteria and yeasts.
23. A method according to claim 22, whereby said lactic acid bacterium is Lactococcus lactis.
24. A method according to claim 22, whereby said lactic acid bacterium is Lactobacillus sp.
25. A method according to claim 22, whereby said yeast is Saccaromyces cerevisiae.
PCT/EP2006/065803 2005-08-30 2006-08-30 Anti-tnf alpha producing lactic acid bacteria for the treatment of chronic enterocolitis WO2007025977A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
AU2006286563A AU2006286563B2 (en) 2005-08-30 2006-08-30 Anti-TNF alpha producing lactic acid bacteria for the treatment of chronic enterocolitis
JP2008528509A JP5049973B2 (en) 2005-08-30 2006-08-30 Anti-TNFα producing lactic acid bacteria for the treatment of chronic enterocolitis
ES06778386.0T ES2626610T3 (en) 2005-08-30 2006-08-30 Lactic acid bacteria producing anti-TNF alpha for the treatment of inflammatory bowel disease
CA2619748A CA2619748C (en) 2005-08-30 2006-08-30 Novel treatment of chronic enterocolitis
EP06778386.0A EP1948206B1 (en) 2005-08-30 2006-08-30 Anti-tnf alpha producing lactic acid bacteria for the treatment of inflammatory bowel disease
US12/065,335 US9017662B2 (en) 2005-08-30 2006-08-30 Anti-TNF alpha producing lactic acid bacteria for the treatment of chronic enterocolitis

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP05107909 2005-08-30
EP05107909.3 2005-08-30
EP05111654.9 2005-12-02
EP05111654 2005-12-02

Publications (3)

Publication Number Publication Date
WO2007025977A2 true WO2007025977A2 (en) 2007-03-08
WO2007025977A3 WO2007025977A3 (en) 2007-06-07
WO2007025977A8 WO2007025977A8 (en) 2008-12-11

Family

ID=37335304

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2006/065803 WO2007025977A2 (en) 2005-08-30 2006-08-30 Anti-tnf alpha producing lactic acid bacteria for the treatment of chronic enterocolitis

Country Status (7)

Country Link
US (1) US9017662B2 (en)
EP (2) EP1948206B1 (en)
JP (1) JP5049973B2 (en)
AU (1) AU2006286563B2 (en)
CA (2) CA2619748C (en)
ES (1) ES2626610T3 (en)
WO (1) WO2007025977A2 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008071751A1 (en) * 2006-12-14 2008-06-19 Actogenix N.V. Delivery of binding molecules to induce immunomodulation
EP2343319A1 (en) 2010-01-07 2011-07-13 University of Ljubljana Genetically modified food grade microorganism for treatment of inflammatory bowel disease
WO2013041672A1 (en) 2011-09-23 2013-03-28 Actogenix Nv Modified gram positive bacteria and uses thereof
WO2013041673A1 (en) 2011-09-23 2013-03-28 Actogenix Nv Modified gram positive bacteria and uses thereof
US8444976B2 (en) 2008-07-02 2013-05-21 Argen-X B.V. Antigen binding polypeptides
EP2725035A1 (en) * 2007-10-02 2014-04-30 Avaxia Biologics, Inc. Antibody therapy for use in the digestive tract
CN104740631A (en) * 2008-10-29 2015-07-01 阿布林克斯公司 Formulations Of Single Domain Antigen Binding Molecules
US9079955B2 (en) 2007-10-02 2015-07-14 Avaxia Biologics, Incorporated Compositions comprising TNF-specific antibodies for oral delivery and methods of use thereof to treat inflammatory bowel disease
US9745372B2 (en) 2015-11-12 2017-08-29 Ablynx N.V. TNF binders
EP3255149A2 (en) 2006-05-02 2017-12-13 Intrexon Actobiotics NV Microbial intestinal delivery of obesity related peptides
US10633438B2 (en) 2015-03-31 2020-04-28 Vhsquared Limited Polypeptides
US10808014B2 (en) 2016-09-13 2020-10-20 Intrexon Actobiotics N.V. Mucoadhesive microorganism
WO2023277778A1 (en) * 2021-06-29 2023-01-05 Milmed Unico Ab Yeast for the treatment of inflammation
US11623952B2 (en) 2019-06-21 2023-04-11 Sorriso Pharmaceuticals, Inc. IL-23 and TNF-alpha binding bi-specific heavy chain polypeptides
US11667719B2 (en) 2019-06-21 2023-06-06 Sorriso Pharmaceuticals, Inc. VHH immunoglobulin chain variable domain that binds to IL-7R and methods of use thereof for treating autoimmune and/or inflammatory diseases
US11684677B2 (en) 2016-09-30 2023-06-27 Sorriso Pharmaceuticals, Inc. Compositions

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2015370982B2 (en) 2014-12-23 2019-03-21 Ilya Pharma Ab Methods for wound healing
WO2020114616A1 (en) * 2018-12-07 2020-06-11 Tillotts Pharma Ag Topical treatment of immune checkpoint inhibitor induced diarrhoea, colitis or enterocolitis using antibodies and fragments thereof
CN110893194B (en) * 2019-11-20 2023-03-14 内蒙古伊利实业集团股份有限公司 New application of bifidobacterium lactis BL-99 in inhibiting intestinal inflammation
JP2023139384A (en) * 2022-03-22 2023-10-04 株式会社HanaVax Human norovirus gii.2-specific antibody

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3631229A1 (en) 1986-09-13 1988-03-24 Basf Ag MONOCLONAL ANTIBODIES AGAINST HUMAN TUMORNESCROSE FACTOR (TNF) AND THEIR USE
US5919452A (en) * 1991-03-18 1999-07-06 New York University Methods of treating TNFα-mediated disease using chimeric anti-TNF antibodies
US6838254B1 (en) 1993-04-29 2005-01-04 Conopco, Inc. Production of antibodies or (functionalized) fragments thereof derived from heavy chain immunoglobulins of camelidae
GB9521568D0 (en) * 1995-10-20 1995-12-20 Lynxvale Ltd Delivery of biologically active polypeptides
EP1123314B1 (en) * 1998-10-20 2004-02-18 Vlaams Interuniversitair Instituut voor Biotechnologie vzw. Use of a cytokine-producing lactococcus strain to treat colitis
FR2810337B1 (en) * 2000-06-20 2002-09-13 Univ Clermont Auvergne MICROORGANISMS ACTIVE IN THE DIGESTIVE ENVIRONMENT
MXPA05004955A (en) * 2002-11-08 2005-11-17 Ablynx Nv Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor.
US20060034845A1 (en) * 2002-11-08 2006-02-16 Karen Silence Single domain antibodies directed against tumor necrosis factor alpha and uses therefor
EP1481681A1 (en) 2003-05-30 2004-12-01 Claudio De Simone Lactic acid bacteria combination and compositions thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1948206A2 *

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3255149A2 (en) 2006-05-02 2017-12-13 Intrexon Actobiotics NV Microbial intestinal delivery of obesity related peptides
WO2008071751A1 (en) * 2006-12-14 2008-06-19 Actogenix N.V. Delivery of binding molecules to induce immunomodulation
US9079955B2 (en) 2007-10-02 2015-07-14 Avaxia Biologics, Incorporated Compositions comprising TNF-specific antibodies for oral delivery and methods of use thereof to treat inflammatory bowel disease
EP2725035A1 (en) * 2007-10-02 2014-04-30 Avaxia Biologics, Inc. Antibody therapy for use in the digestive tract
US9346891B2 (en) 2008-07-02 2016-05-24 Argen-X.N.V. Antigen binding polypeptides
US9428580B2 (en) 2008-07-02 2016-08-30 Argen-X B.V. Antigen binding polypeptides
US8444976B2 (en) 2008-07-02 2013-05-21 Argen-X B.V. Antigen binding polypeptides
US8524231B2 (en) 2008-07-02 2013-09-03 Argen-X B.V. Antigen binding polypeptides
US9315576B2 (en) 2008-07-02 2016-04-19 Argen-X N.V. Antigen binding polypeptides
US9221918B2 (en) 2008-07-02 2015-12-29 Argen-X B.V. Antigen binding polypeptides
CN104740631B (en) * 2008-10-29 2019-04-16 阿布林克斯公司 The preparation of single domain antigen-binding molecule
CN104740631A (en) * 2008-10-29 2015-07-01 阿布林克斯公司 Formulations Of Single Domain Antigen Binding Molecules
US8754198B2 (en) 2010-01-07 2014-06-17 University Of Ljubljana Modified food grade microorganism for treatment of inflammatory bowel disease
WO2011083080A1 (en) 2010-01-07 2011-07-14 University Of Ljubljana Modified food grade microorganism for treatment of inflammatory bowel disease
EP2343319A1 (en) 2010-01-07 2011-07-13 University of Ljubljana Genetically modified food grade microorganism for treatment of inflammatory bowel disease
US9458467B2 (en) 2011-09-23 2016-10-04 Intrexon Actobiotics Nv Modified gram positive bacteria and uses thereof
US9347036B2 (en) 2011-09-23 2016-05-24 Intrexon Actobiotics Nv Modified gram positive bacteria and uses thereof
US10793825B2 (en) 2011-09-23 2020-10-06 Intrexon Actobiotics Nv Modified gram positive bacteria and uses thereof
WO2013041673A1 (en) 2011-09-23 2013-03-28 Actogenix Nv Modified gram positive bacteria and uses thereof
US9982228B2 (en) 2011-09-23 2018-05-29 Intrexon Actobiotics Nv Modified gram positive bacteria and uses thereof
US10030234B2 (en) 2011-09-23 2018-07-24 Intrexon Actobiotics Nv Modified gram positive bacteria and uses thereof
WO2013041672A1 (en) 2011-09-23 2013-03-28 Actogenix Nv Modified gram positive bacteria and uses thereof
US10519418B2 (en) 2011-09-23 2019-12-31 Intrexon Actobiotics Nv Modified gram positive bacteria and uses thereof
US10633438B2 (en) 2015-03-31 2020-04-28 Vhsquared Limited Polypeptides
US10544211B2 (en) 2015-11-12 2020-01-28 Ablynx N.V. TNF binders
EP3266798A2 (en) 2015-11-12 2018-01-10 Ablynx NV Improved tnf binders
US9745372B2 (en) 2015-11-12 2017-08-29 Ablynx N.V. TNF binders
US10808014B2 (en) 2016-09-13 2020-10-20 Intrexon Actobiotics N.V. Mucoadhesive microorganism
US11384123B2 (en) 2016-09-13 2022-07-12 Intrexon Actobiotics N.V. Mucoadhesive microorganism
US11684677B2 (en) 2016-09-30 2023-06-27 Sorriso Pharmaceuticals, Inc. Compositions
US11623952B2 (en) 2019-06-21 2023-04-11 Sorriso Pharmaceuticals, Inc. IL-23 and TNF-alpha binding bi-specific heavy chain polypeptides
US11667719B2 (en) 2019-06-21 2023-06-06 Sorriso Pharmaceuticals, Inc. VHH immunoglobulin chain variable domain that binds to IL-7R and methods of use thereof for treating autoimmune and/or inflammatory diseases
WO2023277778A1 (en) * 2021-06-29 2023-01-05 Milmed Unico Ab Yeast for the treatment of inflammation

Also Published As

Publication number Publication date
US9017662B2 (en) 2015-04-28
EP1948206A2 (en) 2008-07-30
US20080274084A1 (en) 2008-11-06
WO2007025977A8 (en) 2008-12-11
EP3181141A1 (en) 2017-06-21
ES2626610T3 (en) 2017-07-25
JP5049973B2 (en) 2012-10-17
EP1948206B1 (en) 2017-03-01
CA2925307C (en) 2019-12-03
WO2007025977A3 (en) 2007-06-07
JP2009506095A (en) 2009-02-12
CA2619748C (en) 2016-06-14
AU2006286563A1 (en) 2007-03-08
CA2925307A1 (en) 2007-03-08
CA2619748A1 (en) 2007-03-08
AU2006286563B2 (en) 2012-02-23

Similar Documents

Publication Publication Date Title
EP1948206B1 (en) Anti-tnf alpha producing lactic acid bacteria for the treatment of inflammatory bowel disease
US20100303777A1 (en) Delivery of binding molecules to induce immunomodulation
AU770726B2 (en) Use of a cytokine-producing lactococcus strain to treat colitis
US10238697B2 (en) Lactococcus lactis producing TSLP or IL-25 and their uses as probiotics and therapeutics
EP1931762B1 (en) Use of recombinant yeast strain producing an anti -inflammatory compound in the manufacture of a medicament to treat colitis
JP6843997B2 (en) Gastrointestinal disease therapeutic drug transfer microorganisms expressing and secreting P8 protein and pharmaceutical compositions for the prevention or treatment of gastrointestinal disease containing the same.
Steidler Microbiological and immunological strategies for treatment of inflammatory bowel disease
MX2007004374A (en) Chimeric protein.
Zahirović et al. Targeting IL-6 by engineered Lactococcus lactis via surface-displayed affibody
CN115806615A (en) Antibody and mesenchymal stem cell exosome preparation method and application of antibody and mesenchymal stem cell exosome in combined treatment of diseases
JP2024521419A (en) Genetically modified Saccharomyces yeast strains as prophylactic and therapeutic agents
JP6802924B2 (en) Pegiocockspentosaseus strain for drug transmission for treating gastrointestinal diseases expressing and secreting cystatin and a pharmacological composition for preventing or treating gastrointestinal diseases containing the same.
WO2000006764A1 (en) Gastrointestinal bacterial antibody factories
KR20200110342A (en) Combination therapy to treat or prevent cancer
KR20200110341A (en) Combination therapy to treat or prevent cancer
WO2023126446A1 (en) Treatment of therapy-induced enteropathy
KR20160000949A (en) RANKL producing recombinant lactic acid bacteria and use thereof
Zhang et al. An M cell-targeting recombinant L. lactis vaccine against four H. pylori adhesins
Joosten et al. BLOCKADE OF ENDOGENOUS INTERLEUKIN-12 RESULTS IN SUPPRESSION OF MURINE STREPTOCOCCAL CELL WALL (SCW) ARTHRITIS BY ENHANCEMENT OF IL-10 AND IL-1Ra.

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2619748

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2008528509

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 12065335

Country of ref document: US

REEP Request for entry into the european phase

Ref document number: 2006778386

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2006286563

Country of ref document: AU

Ref document number: 2006778386

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2006286563

Country of ref document: AU

Date of ref document: 20060830

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2006286563

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2006778386

Country of ref document: EP