OA18777A - Compositions comprising bacterial strains. - Google Patents

Compositions comprising bacterial strains. Download PDF

Info

Publication number
OA18777A
OA18777A OA1201700487 OA18777A OA 18777 A OA18777 A OA 18777A OA 1201700487 OA1201700487 OA 1201700487 OA 18777 A OA18777 A OA 18777A
Authority
OA
OAPI
Prior art keywords
4dbb
composition
asthma
compositions
disease
Prior art date
Application number
OA1201700487
Inventor
Angela Margaret PATTERSON
George Grant
Imke MULDER
Seanin MCCLUSKEY
Emma RAFTIS
Original Assignee
4D Pharma Research Limited
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 4D Pharma Research Limited filed Critical 4D Pharma Research Limited
Publication of OA18777A publication Critical patent/OA18777A/en

Links

Abstract

The invention provides compositions Comprising bacterial strains for treating and preventing inflammatory and autoimmune diseases.

Description

COMPOSITIONS COMPRISING BACTERIAL STRAINS
TECHNICAL FIELD
This invention is in the field of compositions comprising bacteriai strains îsolated from the mammalian digestive tract and the use of such compositions in the treatment of disease.
BACKGROUND TO THE INVENTION
The human intestine is thought to be stérile in utero, but it is exposed to a large variety of maternai and environmental microbes immediately after birth. Thereafter, a dynamic period of microbial colonization and succession occurs, which is influenced by factors such as delivery mode, environment, diet and host génotype, ail of which impact upon the composition of the gut microbiota, particularly during early life. Subsequently, the microbiota stabilizes and becomes adult-Iîke [l]. The human gut microbiota contains more than 500-1000 different phylotypes belonging essentially to two major bacteriai divisions, the Bacteroidetes and the Firmicutes [2]. The successful symbiotic relationships arising from bacteriai colonization of the human gut hâve yielded a wide variety of metabolic, structural, protective and other bénéficiai functions. The enhanced metabolic activities of the colonized gut ensure that otherwise indigestible dîetary components are degraded with release of by-products providing an important nutrient source for the host. Similarly, the immunological importance of the gut microbiota is well-recognized and is exemplîfied in germfree animais which hâve an impaired immune system that is functionally reconstituted following the introduction of commensal bacteria [3-5].
Dramatic changes in microbiota composition hâve been documented in gastrointestinal disorders such as inflammatory bowel disease (IBD). For example, the levels of Clostridium cluster XlVa bacteria are reduced in IBD patients whilst numbers of E. coli are increased, suggesting a shift in the balance of symbionts and pathobionts within the gut [6-9], Interestingly, this microbial dysbiosis is also associated with imbalances in T effector cell populations.
In récognition of the potential positive effect thaï certain bacteriai strains may hâve on the animal gut, various strains hâve been proposed for use in the treatment of various diseases (see, for example, [10-13]). Also, certain strains, including mostly Lactobacillus and Bifidobacterium strains. hâve been proposed for use in treating various inflammatory and autoimmune diseases that are not directly linked to the intestines (see [14] and [15] for reviews). However, the relationship between different diseases and different bacteriai strains, and the précisé effects of particular bacteriai strains on the gut and at a systemic levei and on any particular types of diseases, are poorly characterised.
There is a requirement in the art for new methods of treating inflammatory and autoimmune diseases. There is also a requirement for the potential effects of gut bacteria to be characterised so that new thérapies using gut bacteria can be developed.
SUMMARY OF THE INVENTION
The inventors hâve developed new thérapies for treating and preventing inflammatory and autoimmune diseases. In particular, the inventors hâve developed new thérapies for treating and preventing diseases and conditions mediated by IL-17 or the ThI7 pathway. In particular, the inventors hâve identified a new bacterial strain that is effective for reducing the Thl7 inflammatory response. As described in the examples, oral administration of compositions comprising the bacterium deposited under accession number NCIMB 42380 may reduce the severity of the inflammatory response, including the ThI7 inflammatory response, in mouse models of asthma, rheumatoid arthritis and multiple sclerosis. As also described in the examples, oral administration of compositions comprising the bacterium deposited under accession number NCIMB 42380 may reduce tumor size in mouse models of cancer that may be associated with the Thl7 inflammatory response.
Therefore, in a first embodiment, the invention provides a composition comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof, for use in a method of treating or preventing a disease or condition mediated by 1L-17 or the Thl7 pathway. The inventors hâve identified that treatment with such bacterial straîns can reduce levels of cytokines that are part of the Thl 7 pathway, including IL-17, can alleviate the Thl7 inflammatory response and can provide clinical benefits în mouse models of inflammatory and autoimmune diseases mediated by IL-17 and the Thl7 pathway.
In particular embodiments, the invention provides a composition comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof, for use in a method of treating or preventing a disease or condition selected from the group consisting of: multiple sclerosis; arthritis, such as rheumatoid arthritis, osteoarthritis, psoriatic arthritis, or juvénile idiopathic arthritis; neuromyelitis optica (Devîc's disease); ankylosing spondylitis; spondyloarthritis; psoriasis; systemic lupus erythematosus; inflammatory bowel disease, such as Crohn’s disease or ulcerative colitis; celîac disease; asthma, such as allergie asthma or neutrophilie asthma; chronic obstructive pulmonary disease (COPD); cancer, such as breast cancer, colon cancer, lung cancer or ovarian cancer; uveitis; scleritis; vasculitis; Behcet's disease; atherosclerosis; atopie dermatitis; emphysema; periodontitis; allergie rhinitis; and allograft rejection. The effect shown for the bacterium deposited under accession number NCIMB 42380 on the Thl7 inflammatory response may provide therapeutic benefits for diseases and conditions mediated by IL-17 and the Thl7 pathway, such as those listed above.
In preferred embodiments, the invention provides a composition comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof, for use in a method of treating or preventing asthma, such as neutrophilie asthma or allergie asthma. The inventors hâve identified that treatment with the bacterium deposited under accession number NCIMB 42380 can reduce recruitment of neutrophils and eosinophils into the lungs, which can help treat or prevent asthma.
Furthermore, the inventors hâve tested and demonstrated the efficacy of the bacterium deposited under accession number NCIMB 42380 in mouse models of asthma. In certain embodiments, the composition is for use in a method of treating or preventing neutrophilie asthma or éosinophilie asthma. The effect shown for the compositions of the invention on neutrophils and eosinophils mean that they may be particularly effective for treating or preventing neutrophilie asthma and éosinophilie asthma. Indeed, in certain embodiments, the composition is for use in a method of reducing a neutrophilie inflammatory response in the treatment or prévention of asthma, or the composition is for use in a method of reducing an éosinophilie inflammatory response in the treatment or prévention of asthma. In certain embodiments, the invention provides a composition comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof, for use in the treatment of asthma. In especially preferred embodiments, the invention provides a composition comprising the bacterium deposited under accession number NCIMB 42380, for use in the treatment of asthma, and in particular neutrophilie asthma. The bacterium deposited under accession number NCIMB 42380 is shown to hâve a particularly pronounced effect on neutrophils in asthma models and treatment with this bacterium may be particularly effective for treating neutrophilie asthma.
In further preferred embodiments, the invention provides a composition comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof, for use in a method of treating or preventing rheumatoid arthritis. The inventors hâve identified that treatment with the bacterium deposited under accession number NCIMB 42380 can provide clinical benefits in a mouse model of rheumatoid artliritis and can reduce joint swelling. In preferred embodiments, the invention provides a composition comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof for use in the treatment of rheumatoid arthritis. Compositions using the bacterium deposited under accession number NCIMB 42380 may be particularly effective for treating rheumatoid artliritis.
In further preferred embodiments, the invention provides a composition comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof, for use in a method of treating or preventing multiple sclerosis. The inventors hâve identified that treatment with the bacterium deposited under accession number NCIMB 42380 can reduce disease incidence and disease severity in a mouse model of multiple sclerosis. In preferred embodiments, the invention provides a composition comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof for use in the treatment of multiple sclerosis. Compositions using the bacterium deposited under accession number NCIMB 42380 may be particularly effective for treating multiple sclerosis.
In further preferred embodiments, the invention provides a composition comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof, for use in a method of treating or preventing cancer, such as breast, lung or liver cancer. The inventors hâve identified that treatment with the bacterium deposited under accession number NCIMB 42380 can reduce tumour growth in mouse models of breast, lung and liver cancer. In certain embodiments, the composition is for use in a method of reducing tumour size or preventing tumour growth in the treatment of cancer.
In further preferred embodiments, the invention provides a composition comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof, for use in a method of treating or preventing uveitis, such as posterior uveitis. Compositions comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof may be particularly effective for treating uveitis.
In certain embodiments, the compositions of the invention are for use in a method of reducing IL-17 production or reducing Thl7 cell différentiation in the treatment or prévention of a disease or condition mediated by IL-17 or the Thl7 pathway. In particular, the compositions of the invention may be used in reducing IL-17 production or reducing Thl7 cell différentiation in the treatment or prévention of asthma, rheumatoid arthritis or multiple sclerosis or of asthma, rheumatoîd arthritis, multiple sclerosis, cancer or uveitis. Preferably, the invention provides compositions comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof for use in reducing IL-17 production or reducing Thl7 cell différentiation in the treatment or prévention of asthma, rheumatoid arthritis or multiple sclerosis. The invention also provides compositions comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof, for use in reducing IL-17 production or reducing Thl 7 cell différentiation in the treatment or prévention of cancer.
In certain embodiments, the composition is for use in a patient with elevated IL-17 levels or Thl7 cells. The effect on the Thl7 inflammatory response shown for the bacterium deposited under accession number NCIMB 42380 may be particularly bénéficiai for such patients.
In preferred embodiments of the invention, the bacterial strain in the composition is the bacterium deposited under accession number NCIMB 42380. Biotype bacterial strains may also be used, such as bacterial strains that hâve a 16s rRNA sequence that is at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% identical to the I6s rRNA sequence of the bacterium deposited under accession number NCIMB 42380. Preferably, the bacterial strain has a 16s rRNA sequence that is at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% identical to SEQ ID NO:l. Preferably, the bacterial strain for use in the invention has the 16s rRNA sequence represented by SEQ ID NO:l.
In certain embodiments, the composition of the invention is for oral administration. Oral administration of the strains of the invention can be effective for treating IL-17- or Thl7 pathwaymediated diseases and conditions. Also, oral administration is convenient for patients and practitioners and allows delivery to and / or partial or total colonisation of the intestine.
In certain embodiments, the composition of the invention comprises one or more pharmaceutically acceptable excipients or carriers.
In certain embodiments, the composition of the invention comprises a bacterial strain that has been lyophilised. Lyophilisation is an effective and convenient technique for preparing stable compositions that allow delivery of bacteria.
In certain embodiments, the invention provides a food product comprising the composition as 5 described above.
In certain embodiments, the invention provides a vaccine composition comprising the composition as described above.
Additionally, the invention provides a method of treating or preventing a disease or condition medîated by IL-17 or the Thl7 pathway, comprising administering a composition comprising the 10 bacterium deposited under accession number NCIMB 42380 or a biotype thereof.
In developing the above invention, the inventors hâve identified and characterised a bacterial strain that is particuiarly useful for therapy. The bacterium deposited under accession number NCIMB 42380 is shown to be effective for treating the diseases described herein, such as arthritis, asthma and multiple sclerosis. Therefore, in another aspect, the invention provides a cell of the bacterium 15 deposited under accession number NCIMB 42380, or a dérivative thereof. The invention also provides compositions comprising such cells, or biologically pure cultures of such cells. The invention also provides a cell of the bacterium deposited under accession number NCIMB 42380, or a dérivative thereof, for use in therapy, in particular for the diseases described herein. The bacterium deposited under accession number NCIMB 42380 is also shown to be effective for treating cancer.
BR1EF DESCRIPTION OF DRAWINGS
Figure 1: Mouse model of house dust mite-induced asthma - Total BAL fluid cell counts.
Figure 2: Mouse model of house dust mite-induced asthma - Total eosinophil count in BALF.
Figure 3: Mouse model of house dust mite-induced asthma - Proportion of eosinophils in BALF.
Figure 4: Mouse model of house dust mite-induced asthma - Total macrophage count in BALF.
Figure 5: Mouse model of house dust mite-induced asthma - Proportion of macrophages in BALF.
Figure 6: Mouse model of house dust mite-induced asthma - Total neutrophil count in BALF.
Figure 7: Mouse model of house dust mite-induced asthma - Proportion of neutrophils in BALF.
Figure 8: Mouse model of house dust mite-induced asthma - Total lymphocyte count in BALF.
Figure 9: Mouse model of house dust mite-induced asthma - Proportion of lymphocytes in BALF.
Figure 10: Mouse model of severe neutrophilie asthma - Total BAL fluid cell counts.
Figure 11: Mouse model of severe neutrophilie asthma - Total eosinophil count in BALF.
Figure 12: Mouse model of severe neutrophilie asthma - Proportion of eosinophils in BALF.
Figure 13: Mouse model of severe neutrophilie asthma - Total macrophage count in BALF.
Figure 14: Mouse model of severe neutrophilie asthma - Proportion of macrophages in BALF.
Figure 15: Mouse model of severe neutrophilie asthma - Total neutrophil count in BALF.
Figure 16: Mouse model of severe neutrophilie asthma - Proportion of neutrophils in BALF.
Figure 17: Mouse model of severe neutrophilie asthma - Total lymphocyte count in BALF.
Figure 18: Mouse model of severe neutrophilie asthma - Proportion of lymphocytes in BALF.
Figure 19: Mouse model of rheumatoid arthritis - Bodyweights, days -14 to 0. Data are presented as Mean ± SEM percentages of the initial (Day -14) bodyweights. Statistical significance: ▲ p < 0.05 and ÀAÀA p< 0.0001 when compared to the vehicle-treated group.
Figure 20: Mouse model of rheumatoid arthritis - Bodyweights, days 0 to 42. Data are presented as Mean ± SEM percentages of the initial (Day 0) bodyweights. À p < 0.05, ♦ p < 0.05, ▲ À À p < 0.001, ···· p < 0.0001 when compared to the vehicle-treated group.
Figure 21: Mouse model of rheumatoid arthritis - Clinical Scores. Data are presented as Mean ± SEM. **** p < 0.0001 when compared to Day 21 in the vehicle-treated group. ♦,Op< 0.05 when compared to the vehicle-treated group on a given day.
Figure 22: Mouse model of rheumatoid arthritis - Splénocyte proliférative response to Collagen II. Media background subtracted [ClI-stimulated - media background] counts per minute based on 3HTdR incorporation. Ail data are presented as Mean ± SEM. ** p < 0.01 compared to Vehicle group.
Figure 23: Mouse model of rheumatoid arthritis - Levels of IFNy in tissue culture supematants. Lines represent group médian values.
Figure 24: Mouse model of rheumatoid arthritis - Levels of IL-17A in tissue culture supematants. Lines represent group médian values.
Figure 25: Mouse model of rheumatoid arthritis - Levels of IL-10 in tissue culture supematants. Lines represent group médian values.
Figure 26: Mouse model of rheumatoid arthritis - Levels of IL-6 in tissue culture supematants. Lines represent group médian values.
Figure 27: Histopathology scoring system.
Figure 28: Mouse model of house dust mite-induced asthma - Total IgE in Sérum
Figure 29: Mouse mode! of house dust mite-induced asthma - HDM spécifie IgGl in Sérum
Figure 30: Mouse model of house dust mite-induced asthma - Total IgE in BALF
Figure 31: Mouse model of house dust mite-induced asthma - HDM spécifie IgGl in BALF
Figure 32: Mouse model of house dust mite-induced asthma - Histological Analysis - Mean Peribronchiolar Infiltration Score
Figure 33: Mouse model of house dust mite-induced asthma - Histological Analysis - Mean Perîvascular Infiltration Score
Figure 34: Mouse model of house dust mite-induced asthma - Histological Analysis - Mean Inflammatory Score (Average of both Peribronchiolar and Perîvascular Infiltration Score)
Figure 35: Mouse model of house dust mite-induced asthma - Histological Analysis - Mucus Score
Figure 36: Mouse model of house dust mite-induced asthma - IL-9 level in lung tissue
Figure 37: Mouse model of house dust mite-induced asthma - IL-la level in lung tissue
Figure 38: Mouse model of house dust mite-induced asthma - IFNg level in lung tissue
Figure 39: Mouse model of house dust mite-induced asthma - IL-17A level in lung tissue
Figure 40: Mouse model of house dust mite-induced asthma - IL-4 level in lung tissue
Figure 41: Mouse model of house dust mite-induced asthma - IL-5 level in lung tissue
Figure 42: Mouse model ofhouse dust mite-induced asthma - IL-1 b level in lung tissue
Figure 43: Mouse model ofhouse dust mite-induced asthma - RANTES level in lung tissue
Figure 44: Mouse mode! ofhouse dust mite-induced asthma - ΜΙΡ-la level in lung tissue
Figure 45: Mouse model ofhouse dust mite-induced asthma - K.C level in lung tissue
Figure 46: Mouse model ofhouse dust mite-induced asthma - MIP-2 level in lung tissue
Figure 47: Mouse model of severe neutrophilie asthma - HDM spécifie IgGl in Sérum
Figure 48: Mouse model of severe neutrophilie asthma - HDM spécifie IgG2a in Sérum
Figure 49: Mouse model of severe neutrophilie asthma - HDM spécifie IgGl in BALF
Figure 50: Mouse model of severe neutrophilie asthma - HDM spécifie lgG2a in BALF
Figure 51: Mouse model of severe neutrophilie asthma - Histological Analysis - Mean Peribronchiolar Infiltration Score
Figure 52: Mouse model of severe neutrophilie asthma - Histological Analysis - Mean Perîvascular Infiltration Score s
Figure 53: Mouse model of severe neutrophilie asthma - Histological Analysis - Mean Inflammatory Score (Average of both Peribronchiolar and Perivascular Infiltration Score)
Figure 54: Mouse model of severe neutrophilie asthma - TNFa level in lung tissue
Figure 55: Mouse model of severe neutrophilie asthma - IL-la level in lung tissue
Figure 56: Mouse model of severe neutrophilie asthma - IFNg level in lung tissue
Figure 57: Mouse model of severe neutrophilie asthma - IL-17F level in lung tissue
Figure 58: Mouse model of severe neutrophilie asthma - IL-1 b level in lung tissue
Figure 59: Mouse model of severe neutrophilie asthma - RANTES level in lung tissue
Figure 60: Mouse mode! of severe neutrophilie asthma - MIP-2 level in lung tissue
Figure 61: Mouse model of severe neutrophilie asthma - K.C level in lung tissue
Figure 62: Mouse model of severe neutrophilie asthma - IL-17A level in lung tissue
Figure 63: Mouse model of severe neutrophilie asthma -ΜΙΡ-la level in lung tissue
Figure 64: Mouse model of severe neutrophilie asthma - IL-33 level in lung tissue
Figure 65: Mouse model of rheumatoid arthritis - Visual Template for Histopathology Scoring. Représentative images showing composite scores from mouse tarsal joints in a collagen-induced arthritis study.
Figure 66: Mouse model of rheumatoid arthritis - Histopathology: Inflammation Scores. Data are presented as Mean ± SEM. ** p < 0.01 when compared to the vehicle-treated group.
Figure 67: Mouse model of rheumatoid arthritis - Histopathology: Cartilage Scores. Data are presented as Mean ± SEM. *** p < 0.001 when compared to the vehicle-treated group.
Figure 68: Mouse model of rheumatoid arthritis - Histopathology: Bone Scores. Data are presented as Mean ± SEM. *** p < 0,001 when compared to the vehicle-treated group.
Figure 69: Mouse model of rheumatoid arthritis - Histopathology: Total Scores. Data are presented as Mean ± SEM. * p < 0.05, *** p < 0.001 when compared to the vehicle-treated group.
Figure 70; Mouse model of rheumatoid arthritis - Histopathology: Représentative Pictures. Animal ID (#n.n) and limb (R for right, L for left) are indicated between brackets. Top left image (vehicle): extensive joint and bone destruction with inflammation and fibrosis extending to the peri-articular soft tissues.
Figure 71: Mouse model of multiple sclerosis - clinical score.
Figure 72: Mouse model of multiple sclerosis - disease incidence.
ΙΟ or the Thl7 pathway. Bacterial strains that are biotypes of the bacterium deposited under accession number NCIMB 42380 are also expected to be effective for treating or preventing cancer. A biotype is a closely related strain that has the same or very similar physiological and biochemîcal characteristics.
In certain embodiments, the bacterial strain for use in the invention has a I6s rRNA sequence that is at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% identical to the I6s rRNA sequence of the bacterium deposited under accession number NCIMB 42380. Preferably, the bacterial strain for use in the invention has a 16s rRNA sequence that is at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% identical to SEQ ID NO:l. Preferably, the bacterial strain for use in the invention has the 16s rRNA sequence represented by SEQ ID NO: l.
Alternatively, strains that are biotypes of the bacterium deposited under accession number NCIMB 42380 and that are suitable for use in the invention may be identified by sequencing other nucléotide sequences for the bacterium deposited under accession number NCIMB 42380. For example substantially the whole genome may be sequenced and a biotype strain for use in the invention may hâve at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% sequence identity across at least 80% of its whole genome (e.g. across at least 85%, 90%, 95% or 99%, or across its whole genome). Other suitable sequences for use in identifying biotype strains may include hsp60 or répétitive sequences such as BOX, ERIC, (GTGh, or REP or [16]. Biotype strains may hâve sequences with at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% sequence identity to the conesponding sequence of the bacterium deposited under accession number NCIMB 42380.
In certain embodiments, the bacterial strain for use in the invention has a genome with sequence identity to SEQ ID NO:2 of WO2016203223. In preferred embodiments, the bacterial strain for use in the invention has a genome with at least 90% sequence identity (e.g. at least 92%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity) to SEQ ID NO:2 across at least 60% (e.g. at least 65%, 70%, 75%, 80%, 85%, 95%, 96%, 97%, 98%, 99% or 100%) of SEQ ID NO:2. For example, the bacterial strain for use in the invention may hâve a genome with at least 90% sequence identity to SEQ ID NO:2 across 70% of SEQ ID NO:2, or at least 90% sequence identity to SEQ ID NO:2 across 80% of SEQ ID NO:2, or at least 90% sequence identity to SEQ ID NO:2 across 90% of SEQ ID NO:2, or at least 90% sequence identity to SEQ ID NO:2 across 100% of SEQ ID NO:2, or at least 95% sequence identity to SEQ ID NO:2 across 70% of SEQ ID NO:2, or at least 95% sequence identity to SEQ ID NO:2 across 80% of SEQ ID NO:2, or at least 95% sequence identity to SEQ ID NO:2 across 90% of SEQ ID NO:2, or at least 95% sequence identity to SEQ ID NO:2 across 100% of SEQ ID NO:2, or at least 98% sequence identity to SEQ ID NO:2 across 70% of SEQ ID NO:2, or at least 98% sequence identity to SEQ ID NO:2 across 80% of SEQ ID NO:2, or at least 98% sequence identity to SEQ ID NO:2 across 90% of SEQ ID NO:2, or at least 98% sequence identity to SEQ ID NO:2 across 100% of SEQ ID NO:2.
H
Alternatively, strains that are biotypes of the bacterium deposîted under accession number NCIMB 42380 and that are suitable for use in the invention may be identified by using the accession number NCIMB 42380 deposit and restriction fragment analysis and/or PCR analysis, for example by using fluorescent amplified fragment length polymorphism (FAFLP) and répétitive DNA element (rep)PCR fingerprinting, or protein profiling, or partial 16S or 23s rDNA sequencing. In preferred embodiments, such techniques may be used to identify strains of the same species as the bacterium deposîted under accession number NCIMB 42380.
In certain embodiments, strains that are biotypes of the bacterium deposîted under accession number NCIMB 42380 and that are suitable for use in the invention are strains that provide the same pattern as the bacterium deposîted under accession number NCIMB 42380 when analysed by amplified ribosomal DNA restriction analysis (ARDRA), for example when using Sau3AI restriction enzyme (for exemplary methods and guidance see, for example,[17]). Alternatively, biotype strains are identified as strains that hâve the same carbohydrate fermentation pattems as the bacterium deposîted under accession number NCIMB 42380.
Bacterial strains that are biotypes of the bacterium deposîted under accession number NCIMB 42380 and that are useful în the compositions and methods of the invention may be identified using any appropriate method or strategy, including the assays described în the examples. For instance, biotypes for use in the invention may be identified by culturing in anaérobie YCFA and/or administering the bacteria to the type II collagen-înduced arthritis mouse model and then assessing cytokine levels. In particular, bacterial strains that hâve similar growth pattems, metabolic type and/or surface antigens to the bacterium deposîted under accession number NCIMB 42380 may be useful in the invention. A biotype strain will hâve comparable immune modulatory activity to the NCIMB 42380 strain. In particular, a biotype strain will elicit comparable effects on the asthma, arthritis, multiple sclerosis and cancer disease models and comparable effects on cytokine levels to the effects shown in the Examples, which may be identified by using the culturing and administration protocols described in the Examples.
A particularly preferred strain of the invention is the bacterium deposîted under accession number NCIMB 42380. This is the exemplary 751 strain tested in the examples and shown to be effective for treating disease. Therefore, the invention provides a cell, such as an isolated cell, of the bacterium deposîted under accession number NCIMB 42380, or a dérivative thereof. The invention also provides a composition comprising a cell of the bacterium deposîted under accession number NCIMB 42380, or a dérivative thereof. The invention also provides a biologically pure culture ofthe bacterium deposîted under accession number NCIMB 42380. The invention also provides a cell of the bacterium deposîted under accession number NCIMB 42380, or a dérivative thereof, for use in therapy, in particular for the diseases described herein.
A dérivative of the bacterium deposited under accession number NCIMB 42380 may be a daughter strain (progeny) or a strain cultured (subcloned) from the original. A dérivative of a strain of the invention may be modified, for example at the genetic level, without ablating the biological activity. In particular, a dérivative strain of the invention is therapeutically active. A dérivative strain will hâve comparable immune modulatory activity to the NCIMB 42380 strain. In particular, a dérivative strain will elicit comparable effects on the asthma, arthritis, multiple sclerosis and cancer disease models and same comparable effects on cytokine levels to the effects shown in the Examples, which may be identified by using the culturing and administration protocols described in the Examples. A dérivative of the NCIMB 42380 strain will generally be a biotype of the NCIMB 42380 strain.
References to cells of the bacterium deposited under accession number NCIMB 42380 encompass any cells that hâve the same safety and therapeutic efficacy characteristics as the strain deposited under accession number NCIMB 42380, and such cells are encompassed by the invention.
In preferred embodiments, the bacterial strains in the compositions of the invention are viable and capable of partially or totally colonising the intestine.
In certain embodiments, the bacterial strain for use in the invention has low adhérence to human intestinal épithélial cells, in particular Caco-2 cells. In a preferred embodiment, the bacterial strain for use in the invention has low adhérence to human intestinal épithélial cells, in particular Caco-2 cells, in YCFA compared to Bifidobacteria, in particular B. breve. In certain embodiments, the bacterial strain for use in the invention exhibits adhérence of less than l% of total culture, such as preferably less than 0.5% or less than 0.3%, when tested under the conditions described in Example 12.
In certain embodiments, the bacterial strain for use in the invention produces exopolysaccharides, for example wherein the exopolysaccharides are bound to the extracellular surface of the bacterial strain. In certain embodiments, the production of the bound exopolysaccharides increases the adhesion of the bacterial strain for use in the invention to mucus or to the surface of épithélial cells, for example human intestinal épithélial cells. In a preferred embodiment, the bacterial strain for use în the invention produces more bound surface exopolysaccharides compared to Bifidohacteria. in particular B. breve.
In a preferred embodiment, the bacterial strain for use in the invention both has low adhérence to human intestinal épithélial cells, in particular Caco-2 cells, in YCFA compared to Bifidohacteria, in particular B. breve (such as adhérence of less than l% of total culture, such as preferably less than 0.5% or less than 0.3%, when tested under the conditions described in Example 12), and produces more bound surface exopolysaccharides compared to Bifidohacteria, in particular B. breve.
In certain preferred embodiments, the bacterial strain for use in the invention is able to ferment the polysaccharide raffînose, for example when cultured in an appropriate suspension medium (such as API suspension medium) at 37°C for 4 hours.
In certain embodiments, the bacterial strain for use in the invention has reduced ability to ferment aglucosidase and/or β-glucosidase compared to Bifidobacteria, in particular B. breve, for example when cultured in an appropriate suspension medium (such as API suspension medium) at 37°C for 4 hours.
In certain embodiments, the bacterial strain for use in the invention comprises one or more of the genes iisted in Table l, such as 5, 10, 20, 50 or ail of the genes in Table I. In certain embodiments, the bacterial strain for use in the invention comprises one or more of the genes Iisted in Table l that are highlighted with single underlining, such as Transmembrane component BL0694 of energizing module of predicted ECF transporter and/or Duplicated ATPase component BL0693 of energizing module of predicted ECF transporter. In certain embodiments, the bacterial strain for use in the invention comprises one or more of the genes Iisted in Table l that are highlighted with double underlining and in bold, such as l, 2, 3, 4 or 5 genes selected from: maltodextrin glucosidase (EC 3.2.1.20), putative galactosidase, cellulose synthase (UDP-forming) (EC 2.4.1.12), chitinase (EC 3.2.1.14) and sensory box/GGDEF family protein. In certain embodiments, the bacterial strain for use in the invention comprises one or more of the genes Iisted in Table 1 that are highlighted with italics, such as 1,2, 3, 4, 5, 6, 7, 8 or 9 genes selected from: omega-3 polyunsaturated fatty acid synthase subunit PfaA, Type I polyketide synthase, putative glycosyl hydrolase of unknown fiinction (DUF1680), ATPase component BioM of energizing module of biotin ECF transporter, Cationtransporting ATPase E1-E2 family, Ribose ABC transport system permease protein RbsC (TC
3.A. 1.2.1), Ribose ABC transport system ATP-binding protein RbsA (TC 3.A. 1.2.1), 3’-to-5' oligoribonuclease (om), membrane protein related to Actinobacillus protein (1944168).
In preferred embodiments, the bacterial strain for use in the invention comprises one or more (such as 5, 10, 15, 20, 25, 30, 40, 45, 50 or ali) genes selected from: 2-succinyl-5-enolpyruvyl-6-hydroxy3- cyclohexene-l-carboxylic-acid synthase (EC 2.2.1.9); 3'-to-5‘ oligoribonuclease (om); Alphagalactosîdase (EC 3.2.1.22); ATPase component of general energizing module of ECF transporters; ATPase component STY3233 of energizing module of queuosine-regulated ECF transporter; ATPdependent DNA helicase recG (EC 3.6.1.-); Beta-glucosidase (EC 3.2.1.21); Cellulose synthase (UDP-forming) (EC 2.4.1.12); Chitinase (EC 3.2.1.14); COG1309: Transcriptional regulator; Dalanyl-D-alanine carboxypeptidase (EC 3.4.16.4); Duplicated ATPase component BL0693 of energizing module of predicted ECF transporter; Fructokinase (EC 2.7.1.4); Glucose/mannose:H+ symporter GlcP; Glycosyltransferase (EC 2.4.1.-); GMP synthase [glutamine-hydrolyzing] (EC 6.3.5.2); Hypothetical sugar kinase in cluster with indigoîdîne synthase indA , PfkB family of kinases; Inosîne-uridine preferring nucleoside hydrolase (EC 3.2.2.1); LSU ribosomal protein L31p @ LSU ribosomal protein L31p, zinc-independent; LSU ribosomal protein L33p @ LSU ribosomal protein L33p, zinc-independent; Maltodextrin glucosidase (EC 3.2.1.20); Membrane protein, related to Actinobacillus protein (1944168); Membrane-bound lytic murein transglycosylase D precursor (EC 3.2.1.-); Methyltransferase (EC 2.1.1.-); NADH-dependent butanol dehydrogenase A (EC 1.1.1,18777
I4 ); Phosphoglycolate phosphatase (EC 3.1.3.18); Phosphoribosylanthranilate isomerase (EC 5.3.1.24); Putative glycosyl hydrolase of unknown function (DUFI680); Rhamnose-containing polysacharide translocation permease; Ribokinase (EC 2.7.1.15); Ribose ABC transport system, ATP-binding protein RbsA (TC 3.A. 1.2.1); Ribose ABC transport system, ATP-binding protein RbsA (TC 5 3.A. 1.2.1); Ribose ABC transport system, hîgh affinity permease RbsD (TC 3.A. 1.2.1); Ribose ABC transport system, periplasmic ribose-binding protein RbsB (TC 3.A. 1.2.1); Ribose ABC transport system, permease protein RbsC (TC 3.A. 1.2.1); Ribose ABC transport System, permease protein RbsC (TC 3.A. 1.2.1); Sorbitol dehydrogenase (EC 1.1.1.14); SSU ribosomal protein S14p (S29e) @ SSU ribosomal protein S14p (S29e), zinc-independent; Substrate-specific component STY3230 of 10 queuosine-regulated ECF transporter; Sucrose-6-phosphate hydrolase (EC 3.2.1.B3); Teichoic acid export ATP-binding protein TagH (EC 3.6.3.40); Transmembrane component BL0694 of energizing module of predicted ECF transporter; Transmembrane component STY3231 of energizing module of queuosine-regulated ECF transporter; Two-component response regulator colocalized with HrtAB transporter; Type I restriction-modification system, DNA-methyltransferase subunit M (EC 15 2.1.1.72); Type I restriction-modification system, restriction subunit R (EC 3.1.21.3); Type I restriction-modification system, specificity subunit S (EC 3.1.21.3); Type 1 restriction-modification system, specificity subunit S (EC 3.1.21.3); Type 1 restriction-modification system, specificity subunit S (EC 3.1.21.3); Xylitol dehydrogenase (EC 1.1.1.9); and Xylose ABC transporter, periplasmic xylose-binding protein XylF. In preferred embodiments, the bacterial strain for use in the 20 invention comprises one or more (such as 5, 10, 15, 20, 25, 30, 35 or ail) genes that are listed in the preceding sentence and thaï are not highlighted in Table 1.
Therapeutic uses
As demonstrated in the examples, the bacterial compositions of the invention are effective for reducing the Th 17 inflammatory response. In particular, treatment with compositions of the invention 25 achieves a réduction in IL-I7A levels and other Thl7 pathway cytokines, and clinical improvements in animal models of conditions mediated by IL-17 and the Th 17 pathway. Therefore, the compositions of the invention may be useful for treating or preventing inflammatory and autoimmune diseases, and in particular diseases or conditions mediated by IL-17. In particular, the compositions of the invention may be useful for reducing or preventing élévation of the IL-17 30 inflammatory response.
Th 17 cells are a subset ofT helper cells that produce, for example, IL-17A, IL17-F, IL-21 and IL-22. Th 17 cell différentiation and IL-17 expression may be driven by IL-23. These cytokines and others form important parts of the Th 17 pathway, which is a well-established inflammatory sîgnalling pathway that contrîbutes to and underlies a number of inflammatory and autoimmune diseases (as 35 described in, for example, [18-23]). Diseases wherein the Thl7 pathway is activated are Thl7 pathway-mediated diseases. Th 17 pathway-mediated diseases can be ameliorated or alleviated by repressing the Th 17 pathway, which may be through a réduction in the différentiation of Th 17 cells or a réduction in their activity or a réduction in the level of Thl7 pathway cytokines. Diseases mediated by the Th 17 pathway may be characterised by increased levels of cytokines produced by Th 17 cells, such as IL-17A, IL-17F, IL-21, IL-22, IL-26, IL-9 (reviewed in [24]). Diseases mediated by the Th 17 pathway may be characterised by increased expression of Th-17-related genes, such as Stat3 or IL-23R. Diseases mediated by the Th 17 pathway may be associated with increased levels of Th 17 cells.
IL-17 is a pro-inflammatory cytokine that contributes to the pathogenesis of several inflammatory and autoimmune diseases and conditions. IL-17 as used herein may refer to any member ofthe IL-17 Family, including IL-17A, IL-17B, IL-17C, IL-17D, IL-17E, and IL-17F. IL-17-mediated diseases and conditions are characterised by high expression of IL-17 and/or the accumulation or presence of IL-17-positive cells in a tissue affected by the disease or condition. Similarly, IL-17-mediated diseases and conditions are diseases and conditions that are exacerbated by high IL-17 levels or an increase in IL-17 levels, and that are alleviated by low IL-17 levels or a réduction in IL-17 levels. The IL-17 inflammatory response may be local or systemic.
Examples of diseases and conditions that may be mediated by IL-17 or the Th 17 pathway include multiple sclerosis; arthritis, such as rheumatoid arthritis, osteoarthritis, psoriatic arthritis, or juvénile idiopathîc arthritis; neuromyelitis optica (Devic’s disease); ankylosing spondylitis; spondyloarthritis; psoriasis; systemic lupus erythematosus; inflammatory bowel disease, such as Crohn’s disease or ulcerative colitis; celiac disease; asthma, such as allergie asthma or neutrophilie asthma; chronic obstructive pulmonary disease (COPD); cancer, such as breast cancer, colon cancer, lung cancer or ovarian cancer; uveitis; scleritis; vasculitis; Behcet's disease; atherosclerosis; atopie dermatitis; emphysema; periodontitis; allergie rhinîtîs; and allograft rejection. In preferred embodiments, the compositions of the invention are used for treating or preventing one or more of these conditions or diseases. In further preferred embodiments, these conditions or diseases are mediated by IL-17 or the ThI 7 pathway.
In certain embodiments, the compositions of the invention are for use in a method of reducing IL-17 production or reducing Th 17 cell différentiation in the treatment or prévention of a disease or condition mediated by IL-17 or the Th 17 pathway. In certain embodiments, the compositions of the invention are for use in treating or preventing an inflammatory or autoimmune disease, wherein said treatment or prévention is achieved by reducing or preventing élévation ofthe Th 17 inflammatory response. In certain embodiments, the compositions of the invention are for use in treating a patient with an inflammatory or autoimmune disease, wherein the patient has elevated IL-17 levels or elevated Thl7 cells or is exhibiting a Thl7 inflammatory response. In certain embodiments, the patient may hâve been diagnosed with a chronic inflammatory or autoimmune disease or condition, or the composition of the invention may be for use in preventing an inflammatory or autoimmune disease or condition developing into a chronic inflammatory or autoimmune disease or condition. In certain embodiments, the disease or condition may not be responsive to treatment with TNF-a inhibitors. These uses of the invention may be applied to any of the spécifie disease or conditions listed in the preceding paragraph.
IL-17 and the Thl7 pathway are often associated with chrome inflammatory and autoimmune diseases, so the compositions of the invention may be particularly useful for treating or preventing chronic diseases or conditions as listed above. In certain embodiments, the compositions are for use in patients with chronic disease. In certain embodiments, the compositions are for use in preventing the development of chronic disease.
The compositions of the invention may be useful for treating diseases and conditions mediated by IL17 or the Thl7 pathway and for addressîng the Thl7 inflammatory response, so the compositions of the invention may be particularly useful for treating or preventing chronic disease, treating or preventing disease in patients that hâve not responded to other thérapies (such as treatment with TNF-α inhibitors), and/or treating or preventing the tissue damage and symptoms associated with IL17 and Th 17 cells. For example, IL-17 is known to activate matrix destruction in cartilage and bone tissue and IL-17 has an inhibitory effect on matrix production in chondrocytes and osteoblasts, so the compositions of the invention may be useful for treating or preventing bone érosion or cartilage damage.
In certain embodiments, treatment with compositions of the invention provides a réduction or prevents an élévation in IL-17 levels, in particular IL-17A levels. In certain embodiments, treatment with compositions of the invention provides a réduction or prevents an élévation in IFN-y, IL-Ιβ, RANTES, ΜΙΡ-Ια, IL-8 or IL-6 levels. Such réduction or prévention of elevated levels of these cytokines may be useful for treating or preventing inflammatory and autoimmune diseases and conditions, in particular those mediated by IL-17 or the Th 17 pathway.
In certain embodiments, treatment with the compositions of the invention provides a block of the attachment to or invasion of human cells, for example human épithélial cells by pathogenic cells, for example E. coli and/or 5. enteritidis.
In certain embodiments, treatment with the compositions of the invention reduces or prevents the binding of pathogenic cells, for example E. coli and/or 5. enteritidis, to the human épithélial cells, for example human intestinal épithélial cells.
In certain embodiments, the production and release of exopolysaccharides by the bacterial strains of the compositions of the invention may hâve protective effects against pathogenic species, for example E. coli and/or S. enteritidis. In certain embodiments, the production and release of exopolysaccharides by the bacterial strains of the compositions of the invention may médiate the effect of the bacteria on the IL-17 or Th 17 pathway and may influence the host immune response. In certain embodiments, the compositions of the invention are for use in producing exopolysaccharides in the treatment of inflammatory and autoimmune diseases, and in particular diseases or conditions mediated by IL-17.
In certain embodiments, the low adhérence to human intestinal épithélial cells, in particular Caco-2 cells, of the bacterial strains of the compositions of the invention may increase the bénéficiai effect of the compositions of the invention on the JL-17 or the Thl7 pathway and on diseases mediated by IL-17 or the Thl 7 pathway.
In certain embodiments, treatment with compositions of the invention provides increased fermentation of raffinose in the intestine. The examples demonstrate that the bacterial strains of the compositions of the invention ferment the polysaccharide raffinose, and raffinose fermentation may confer effects on the host such as increased caecal butyrate and increased gastrointestinal prolifération. In certain embodiments, the compositions of the invention are for use in increasing fermentation of raffinose in the intestine in the treatment of inflammatory and autoimmune diseases, and in particular diseases or conditions mediated by IL-17.
Asthma
In preferred embodiments, the compositions of the invention are for use in treating or preventing asthma. The examples demonstrate that the compositions of the invention achieve a réduction in the recruitment of neutrophils and/or eosinophils into the airways following sensitisation and challenge with house dust mite extract and so they may be useful in the treatment or prévention of asthma. Asthma is a chronic disease characterised by inflammation and restriction of the airways. The inflammation in asthma may be mediated by IL-17 and/or Thl7 cells, and so the compositions of the invention may be particularly effective for preventing or treating asthma. The inflammation in asthma may be mediated by eosinophils and/or neutrophils.
In certain embodiments, the asthma is éosinophilie or allergie asthma. Eosinophilie and allergie asthma are characterised by increased numbers of eosinophils in perîpheral blood and in airway sécrétions and is associated pathologically with thickening of the basement membrane zone and pharmacologically by corticosteroid responsiveness [25], Compositions that reduce or inhibit eosinophil recruitment or activation may be usefiil for treating or preventing éosinophilie and allergie asthma.
In additional embodiments, the compositions of the invention are for use in treating or preventing neutrophilie asthma (or non-eosinophilic asthma). High neutrophil numbers are associated with severe asthma that may be insensitive to corticosteroid treatment. Compositions that reduce or inhibit neutrophil recruitment or activation may be useful for treating or preventing neutrophilie asthma.
Eosinophilie and neutrophilie asthma are not mutually exclusive conditions and treatments that help address either the eosinophil and neutrophil responses may be useful for treating asthma in general.
Increased IL-17 levels and activation of the Thl7 pathway are associated with severe asthma, so the compositions of the invention may be usefiil for preventing the development of severe asthma or for treating severe asthma.
In certain embodiments, the compositions of the invention are for use in methods reducing an éosinophilie inflammatory response in the treatment or prévention of asthma, or for use in methods of reducing a neutrophilie inflammatory response in the treatment or prévention of asthma. As noted above, high levels of eosinophils in asthma is associated pathologically with thickening of the basement membrane zone, so reducing éosinophilie inflammatory response in the treatment or prévention of asthma may be able to specifically address this feature of the disease. Also, elevated neutrophils, either in combination with elevated eosinophils or in their absence, is associated with severe asthma and chronic airway narrowing. Therefore, reducing the neutrophilie inflammatory response may be particulariy usefiil for addressing severe asthma.
In certain embodiments, the compositions reduce peribronchiolar infiltration in allergie asthma, or are for use in reducing peribronchiolar infiltration in the treatment of allergie asthma. In certain embodiments, the compositions reduce peribronchiolar and/or perivascular infiltration in neutrophilie asthma, or are for use in reducing peribronchiolar and/or perivascular infiltration in the treatment of allergie neutrophilie asthma.
In certain embodiments, treatment with compositions of the invention provides a réduction or prevents an élévation in IL-Ιβ, IFNy, RANTES, ΜΙΡ-Ια or IL-8 levels.
In certain embodiments, the compositions of the invention are for use in a method of treating asthma that results in a réduction of the éosinophilie and/or neutrophilie inflammatory response. In certain embodiments, the patient to be treated has, or has previously been identified as having, elevated neutrophil or eosinophîl levels, for example as identified through blood sampling or sputum analysis.
The compositions of the invention may be useful for preventing the development of asthma in a newbom when administered to the new-bom, or to a prégnant woman. The compositions may be useful for preventing the development of asthma in children. The compositions of the invention may be useful for treating or preventing adult-onset asthma. The compositions of the invention may be usefiil for managing or alleviating asthma. The compositions of the invention may be particulariy useful for reducing symptoms associated with asthma that is aggravated by allergens, such as house dust mites.
Treatment or prévention of asthma may refer to, for example, an alleviation of the severity of symptoms or a réduction in the frequency of exacerbations or the range of triggers that are a problem for the patient.
In certain embodiments, treatment with compositions of the invention provides a réduction in concentrations of phenylalanine and/or histidine, for example in the intestines or in the plasma. The examples demonstrate that the bacterial strains of the compositions of the invention tested positive
I9 for fermentation of amino acids, including phenylalanine and histidine, and increased plasma concentrations of phenylalanine and histidine hâve been reported to be associated with adverse effects in asthma. In certain embodiments, the compositions of the invention are for use in reducing plasma concentrations of phenylalanine and/or histidine in the treatment of asthma, and in particular 5 in the treatment of histamine production or airway hyper-responsiveness associated with asthma.
In certain embodiments, treatment with compositions of the invention provides a réduction in concentrations of galactose and/or fructose, for example in the intestines. The examples demonstrate that the bacterial strains of the compositions of the invention ferment carbohydrate substrates including galactose and fructose, and galactose a-l,3- galactose derived from méat sources is a I0 known allergen and causative agent of anaphylaxis, and intake levels of dietary fructose are correlated with increased asthma severity. In certain embodiments, the compositions of the invention are for use in reducing concentrations of galactose and/or fructose in the treatment of asthma, and in particular in the treatment of severe asthma.
Arthritis
In preferred embodiments, the compositions of the invention are for use în treating or preventing rheumatoid arthritis (RA). The examples demonstrate that the compositions of the invention achieve a réduction in the clinical signs of RA in a mouse model, reduce cartilage and bone damage, and reduce the IL-17 inflammatory response, and so they may be useful în the treatment or prévention of RA. RA is a systemic inflammatory disorder that primarily affects joints. RA is associated with an 20 inflammatory response that results in swelling of joints, synovial hyperplasia, and destruction of cartilage and bone. IL-17 and Thl7 cells may hâve a key rôle in RA, for example because IL-17 inhibits matrix production in chondrocytes and osteoblasts and activâtes the production and function of matrix metalloproteinases and because RA disease activity is correlated to IL-17 levels and Th-l7 cell numbers [26,27], so the compositions of the invention may be particularly effective for preventing or treating RA.
In certain embodiments, the compositions of the invention are for use in lowering IL-17 levels or preventing élévation of IL-17 levels in the treatment or prévention of RA. In certain embodiments, treatment with compositions of the invention provides a réduction or prevents an élévation in IL-17 levels, in particular 1L-17A levels. In certain embodiments, treatment with compositions of the 30 invention provides a réduction or prevents an élévation in IFN-γ or IL-6 levels.
In certain embodiments, treatment with the compositions of the invention results in a réduction in the swelling of joints. In certain embodiments, the compositions of the invention are for use in patients with swollen joints or patients identified as at risk of having swollen joints. In certain embodiments, the compositions of the invention are for use in a method of reducing joint swelling in RA.
In certain embodiments, treatment with the compositions of the invention results in a réduction in cartilage damage or bone damage. In certain embodiments, the compositions of the invention are for use in reducing or preventing cartilage or bone damage in the treatment of RA. In certain embodiments, the compositions are for use in treating patient with severe RA that are at risk of cartilage or bone damage.
Increased IL-17 levels and Thl7 cell numbers are associated with cartilage and bone destruction in RA [26,27]. IL-17 is known to activate matrix destruction in cartilage and bone tissue and IL-17 has an inhibitory effect on matrix production in chondrocytes and osteoblasts. Therefore, in certain embodiments, the compositions of the invention are for use in preventing bone érosion or cartilage damage in the treatment of RA. In certain embodiments, the compositions are for use in treating patients that exhibit bone érosion or cartilage damage or patients identified as at risk of bone érosion or cartilage damage.
TNF-α is also associated with RA, but TNF-α is not învolved in the pathogenesis of the later stages of the disease. In contrast, IL-17 has a rôle throughout ail stages of chronic disease [28]. Therefore, in certain embodiments the compositions of the invention are for use in treating chronic RA or latestage RA, such as disease that includes joint destruction and loss of cartilage. In certain embodiments, the compositions of the invention are for treating patients that hâve previously received anti-TNF-α therapy. In certain embodiments, the patients to be treated do not respond or no longer respond to anti-TNF-α therapy.
The compositions of the invention may be usefiil for modulating a patient’s immune system, so in certain embodiments the compositions of the invention are for use in preventing RA in a patient that has been identified as at risk of RA, or that has been diagnosed with early-stage RA. The compositions of the invention may be usefiil for preventing the development of RA.
The compositions of the invention may be usefiil for managing or alleviating RA. The compositions of the invention may be particularly usefiil for reducing symptoms associated with joint swelling or bone destruction. Treatment or prévention of RA may refer to, for example, an alleviation of the severity of symptoms or a réduction in the frequency of exacerbations or the range of triggers that are a problem for the patient.
Multiple sclerosis
In preferred embodiments, the compositions of the invention are for use in treating or preventing multiple sclerosis. The examples demonstrate that the compositions of the invention achieve a réduction in the disease incidence and disease severity in a mouse model of multiple sclerosis (the EAE model), and so they may be usefiil in the treatment or prévention of multiple sclerosis. Multiple sclerosis is an inflammatory disorder associated with damage to the myelin sheaths of neurons, particularly in the brain and spinal column. Multiple sclerosis is a chronic disease, which is progressively incapacitating and which evolves in épisodes. IL-17 and Thl7 cells may hâve a key rôle in multiple sclerosis, for example because IL-17 levels may correlate with multiple sclerosis lésions, IL-17 can disrupt blood brain barrier endothélial cell tight junctions, and Thl7 cells can migrate into the central nervous system and cause neuronal loss [29,30], Therefore, the compositions of the invention may be particularly effective for preventing or treating multiple sclerosis.
In certain embodiments, treatment with the compositions of the invention results in a réduction in disease incidence or disease severity. In certain embodiments, the compositions of the invention are for use in reducing disease incidence or disease severity. In certain embodiments, treatment with the compositions of the invention prevents a décliné in motor function or results în improved motor function. In certain embodiments, the compositions of the invention are for use in preventing a décliné in motor function or for use in improving motor function. In certain embodiments, treatment with the compositions of the invention prevents the development of paralysis. In certain embodiments, the compositions of the invention are for use in preventing paralysis în the treatment of multiple sclerosis.
The compositions of the invention may be useful for modulating a patîent’s immune system, so in certain embodiments the compositions of the invention are for use in preventing multiple sclerosis in a patient that has been identified as at risk of multiple sclerosis, or that has been diagnosed with early-stage multiple sclerosis or “relapsing-remitting” multiple sclerosis. The compositions of the invention may be useful for preventing the development of sclerosis. Indeed, the examples show that administration of compositions of the invention prevented the development of disease in many mice.
The compositions of the invention may be useful for managing or alleviating multiple sclerosis. The compositions of the invention may be particularly useful for reducing symptoms associated with multiple sclerosis. Treatment or prévention of multiple sclerosis may refer to, for example, an alleviation of the severity of symptoms or a réduction in the frequency of exacerbations or the range of trîggers that are a problem for the patient.
Uveitis
In preferred embodiments, the compositions of the invention are for use in treating or preventing uveitis. The compositions of the invention may achieve a réduction in disease incidence and disease severity in an animal model of uveitis and so they may be useful in the treatment or prévention of uveitis. Uveitis is inflammation of the uvea and can resuit in retinal tissue destruction. It can présent in different anatomical forms (anterior, intermediate, posterior or diffuse) and resuit from different, but related, causes, including systemic autoimmune disorders. IL-17 and the Thl7 pathway are centrally involved in uveitis, so the compositions of the invention may be particularly effective for preventing or treating uveitis. References [31-38] describe elevated sérum levels of interleukin-17A in uveitis patients, spécifie association of IL17A genetic variants with panuveitis, the rôle of Thl7associated cytokines în the pathogenesis of experimental autoimmune uveitis, the imbalance between Thl7 Cells and regulatory T Cells during monophasic experimental autoimmune uveitis, the upregulation of IL-17A in patients with uveitis and active Adamantiades-Behçet and Vogt-Koyanagi18777
Harada (VKH) diseases, the treatment of non-infectious uveitis with secukinumab (anti-lL-l7A antibody), and Th 17 in uveitic eyes.
In certain embodiments, the uveitis is posterior uveitis. Posterior uveitis présents primarily with inflammation of the retina and choroid and the compositions of the invention may be effective for reducing retinal inflammation and damage.
In certain embodiments, treatment with the compositions of the invention results in a réduction in retinal damage. In certain embodiments, the compositions of the invention are for use in reducing or preventing retinal damage in the treatment of uveitis. In certain embodiments, the compositions are for use in treating patients with severe uveitis that are at risk of retinal damage. In certain embodiments, treatment with the compositions of the invention results in a réduction in optic dise inflammation. In certain embodiments, the compositions of the invention are for use in reducing or preventing optic dise inflammation. In certain embodiments, treatment with the compositions of the invention results in a réduction in retinal tissue infiltration by inflammatory cells. In certain embodiments, the compositions of the invention are for use in reducing retinal tissue infiltration by inflammatory cells. In certain embodiments, treatment with the compositions of the invention results in vision being maintained or improved. In certain embodiments. the compositions of the invention are for use in maintaining or improving vision.
In certain embodiments, the compositions are for use in treating or preventing uveitis associated with a non-infectious or autoimmune disease, such as Behçet disease, Crohn's disease, Fuchs hétérochromie iridocyclitis, granulomatosis with polyangiitis, HLA-B27 related uveitis, juvénile idiopathic arthritis, sarcoidosis, spondyloarthritis, sympathetic ophthalmia, tubulointerstitial nephritîs and uveitis syndrome or Vogt-Koyanagi-Harada syndrome. IL-17A has been shown to be involved in, for example, Behçet and Vogt-Koyanagi-Harada diseases.
Treatment or prévention of uveitis may refer to, for example, an alleviation of the severity of symptoms or a prévention of relapse.
Treating cancer
In preferred embodiments, the compositions of the invention are for use in treating or preventing cancer. The examples demonstrate that administration of the compositions of the invention can lead to a réduction in tumour growth in a number of tumour models.
In certain embodiments, treatment with the compositions of the invention results in a réduction in tumour size or a réduction in tumour growth. In certain embodiments, the compositions of the invention are for use in reducing tumour size or reducing tumour growth. The examples demonstrate that the compositions of the invention may be effective for reducing tumour size or growth. In certain embodiments, the compositions of the invention are for use in patients with solid tumours. In certain embodiments, the compositions of the invention are for use in reducing or preventing angiogenesis in the treatment of cancer. IL-17 and Thl7 cells hâve central rôles in angiogenesis. In certain embodiments, the compositions of the invention are for use in preventing metastasis.
In certain embodiments, the compositions of the invention are for use in treating or preventing breast cancer. The examples demonstrate that the compositions of the invention may be effective for treating breast cancer. In certain embodiments, the compositions of the invention are for use in reducing tumour size, reducing tumour growth, or reducing angiogenesis in the treatment of breast cancer. In preferred embodiments the cancer is mammary carcinoma. In preferred embodiments the cancer is stage IV breast cancer.
In certain embodiments, the compositions of the invention are for use in treating or preventing lung cancer. The examples demonstrate that the compositions of the invention may be effective for treating lung cancer. In certain embodiments, the compositions of the invention are for use in reducing tumour size, reducing tumour growth, or reducing angiogenesis in the treatment of lung cancer. In preferred embodiments the cancer is lung carcinoma.
In certain embodiments, the compositions of the invention are for use in treating or preventing liver cancer. The exampies demonstrate that the compositions of the invention may be effective for treating liver cancer. In certain embodiments, the compositions of the invention are for use in reducing tumour size, reducing tumour growth, or reducing angiogenesis in the treatment of liver cancer. In preferred embodiments the cancer is hepatoma (hepatocellular carcinoma).
In certain embodiments, the compositions of the invention are for use in treating or preventing carcinoma. The examples demonstrate that the compositions of the invention may be effective for treating numerous types of carcinoma. In certain embodiments, the compositions of the invention are for use in treating or preventing non-immunogenic cancer. The examples demonstrate that the compositions ofthe invention may be effective for treating non-immunogenic cancers.
IL-17 and the ThI7 pathway hâve central rôles in cancer development and progression and, although the rôles of IL-l 7 and Thl 7 cells in cancer are not fully understood, numerous pro-tumour effects of IL-17 and Thl7 cells are known. For example, Thl7 cells and IL-l 7 can promote angiogenesis, increase prolifération and survival of tumor cells and activate tumour-promoting transcription factors [39-41], Therefore, the compositions ofthe invention may be useful for treating or preventing cancer. Furthermore, the examples demonstrate that the compositions of the invention are effective for reducing tumour volume in breast, lung and liver cancer, and IL-l7 and Thl7 cells hâve important rôles in these spécifie types of cancer [42-44].
The therapeutic effects of the compositions of the invention on cancer may be mediated by a proinflammatory mechanism. Inflammation can hâve a cancer-suppressive effect [45] and proinflammatory cytokines such as TNFa are being învestigated as cancer thérapies [46], The compositions of the invention may be useful for treating cancer via a similar mechanism. For example, the compositions of the invention may elicit an IFNy-type response. IFNy is a potent macrophage-activating factor that can stimulate tumirocidal activity [47], and CXCL9 and CXCLIO, for example, also hâve anti-cancer efifects [48-50], Therefore, in certain embodiments, the compositions of the invention are for use in promoting inflammation in the treatment of cancer. In preferred embodiments, the compositions of the invention are for use in promoting Thl inflammation 5 in the treatment of cancer. Thl cells produce IFNy and hâve potent anti-cancer effects [45]. In certain embodiments, the compositions of the invention are for use in treating an early-stage cancer, such as a cancer that has not metastasized, or a stage 0 or stage l cancer. Promoting inflammation may be more effective against early-stage cancers [45]. In certain embodiments, the compositions of the invention are for use in promoting inflammation to enhance the effect of a second anti-cancer agent.
In further embodiments, the compositions of the invention are for use in treating or preventing acute lymphoblastic leukemia (ALL), acute myeloid leukemia, adrenocortical carcinoma, basal-cell carcinoma, bile duct cancer, bladder cancer, bone tumor, osteosarcoma/malignant fibrous histiocytoma. brainstem glioma, brain tumor, cerebellar astrocytoma, cérébral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, breast 15 cancer, bronchial adenomas/carcinoids, Burkitt's lymphoma, carcinoid tumor, cervical cancer, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative disorders, colon cancer, cutaneous T-cell lymphoma, endométrial cancer, ependymoma. esophageal cancer, Ewing's sarcoma, intraocular melanoma, retînoblastoma, gallbladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), germ cell tumor, glioma, 20 childhood visual pathway and hypothalamic, Hodgkin lymphoma, melanoma, islet cell carcinoma, Kaposi sarcoma, rénal cell cancer, laryngeal cancer, leukaemias, lymphomas, mesothelioma, neuroblastoma, non-Hodgkin lymphoma, oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, parathyroid cancer, pharyngeal cancer, pituitary adenoma, plasma cell neoplasia, prostate cancer, rénal cell carcinoma, retînoblastoma, sarcoma, testicular cancer, thyroid cancer, or 25 uterine cancer.
The compositions of the invention may be particularly effective when used in combination with further therapeutic agents. The immune-moduiatory effects of the compositions of the invention may be effective when combined with more direct anti-cancer agents. Therefore, in certain embodiments, the invention provides a composition comprising lhe bacterium deposited under accession number 30 NCIMB 42380 or a biotype thereof and an anticancer agent. In preferred embodiments the anticancer agent is an immune checkpoint inhibitor, a targeted antibody immunotherapy, a CAR-T cell therapy, an oncolytic virus, or a cytostatic drug. In preferred embodiments, the composition comprises an anti-cancer agent selected from the group consisting of: Yervoy (ipilimumab, BMS); Keytruda (pembrolizumab, Merck); Opdivo (nivolumab, BMS); MEDI4736 (AZ/Medlmmune); MPDL3280A 35 (Roche/Genentech); Tremelimumab (AZ/Medlmmune); CT-OII (pidilizumab, CureTech); BMS986015 (lirilumab, BMS); MEDI0680 (AZ/Medlmmune); MSB-0010718C (Merck); PF-05082566 (Pfizer); MEDI6469 (AZ/Medlmmune); BMS-986016 (BMS); BMS-663513 (urelumab, BMS);
IMP321 (Prima Biomed); LAG525 (Novartis); ARGX-HO (arGEN-X); PF-05082466 (Pfizer); CDX-H27 (varlilumab; CellDex Therapeutics); TRX-518 (GITR Inc.); MK-4166 (Merck); JTX2011 (Jounce Therapeutics); ARGX-H5 (arGEN-X); NLG-9189 (indoximod, NewLink Genetics); INCB024360 (Incyte); IPH2201 (Innate Immotherapeutics/AZ); NLG-919 (NewLink Genetics); antiVISTA (JnJ); Epacadostat (INCB24360, Incyte); F001287 (Flexus/BMS); CP 870893 (University of Pennsylvania); MGA271 (Macrogenix); Emactuzumab (Roche/Genentech); Galunisertib (Eli Lilly); Ulocuplumab (BMS); BKT140/BL8040 (Biokine Therapeutics); Bavituximab (Peregrine Pharmaceuticals); CC 90002 (Celgene); 852A (Pfizer); VTX-2337 (VentiRx Pharmaceuticals); IMO2055 (Hybridon, Idera Pharmaceuticals); LY2157299 (Eli Lilly); EW-7197 (Ewha Women's University, Korea); Vemurafenib (Plexxikon); Dabrafenib (Genentech/GSK); BMS-777607 (BMS); BLZ945 (Memorial Sloan-Kettering Cancer Centre); Unituxin (dinutuximab, United Therapeutics Corporation); Blincyto (blinatumomab, Amgen); Cyramza (ramucirumab, Eli Lilly); Gazyva (obinutuzumab, Roche/Biogen); Kadcyla (ado-trastuzumab emtansine, Roche/Genentech); Perjeta (pertuzumab, Roche/Genentech); Adcetris (brentuximab vedotin, Takeda/Millennium); Arzerra (ofatumumab, GSK); Vectibix (panitumumab, Amgen); Avastin (bevacizumab, Roche/Genentech); Erbitux (cetuximab, BMS/Merck); Bexxar (tositumomab-1131, GSK); Zevalin (ibritumomab tiuxetan, Biogen); Campath (alemtuzumab, Bayer); Mylotarg (gemtuzumab ozogamicin, Pfizer); Herceptin (trastuzumab, Roche/Genentech); Rituxan (rituximab, Genentech/Biogen); volociximab (Abbvie); Enavatuzumab (Abbvie); ABT-414 (Abbvie); Elotuzumab (Abbvie/BMS); ALX-014I (Ablynx); Ozaralizumab (Ablynx); Actimab-C (Actinium); Actimab-P (Actinium); Milatuzumab-dox (Actinium); Emab-SN-38 (Actinium); Naptumonmab estafenatox (Active Biotech); AFM 13 (Affimed); AFM11 (Affimed); AGS-16C3F (Agensys); AGS-16M8F (Agensys); AGS-22ME (Agensys); AGS-15ME (Agensys); GS-67E (Agensys); ALXN6000 (samalizumab, Alexion); ALT836 (Altor Bioscience); ALT-801 (Altor Bioscience); ALT-803 (Altor Bioscience); AMG780 (Amgen); AMG 228 (Amgen); AMG820 (Amgen); AMG172 (Amgen); AMG595 (Amgen); AMG110 (Amgen); AMG232 (adecatumumab, Amgen); AMG211 (Amgen/Medlmmune); BAY2010112 (Amgen/Bayer); Rilotumumab (Amgen); Denosumab (Amgen); AMP-514 (Amgen); MEDI575 (AZ/Medlmmune); MED13617 (AZ/Medlmmune); MEDI6383 (AZ/Medlmmune); MEDI551 (AZ/Medlmmune); Moxetumomab pasudotox (AZ/Medlmmune); MEDI565 (AZ/Medlmmune); MED10639 (AZ/Medlmmune); MEDI0680 (AZ/Medlmmune); MED1562 (AZ/Medlmmune); AV-380 (AVEO); AV203 (AVEO); AV299 (AVEO); BAY79-4620 (Bayer); Anetumab ravtansine (Bayer); vantictumab (Bayer); BAY94-9343 (Bayer); Sibrotuzumab (Boehringer Ingleheim); BI-836845 (Boehringer Ingleheim); B-701 (BioClin); B1IB015 (Biogen); Obinutuzumab (Biogen/Genentech); BI-505 (Bioinvent); BI-1206 (Bioinvent); TB-403 (Bioinvent); BT-062 (Biotest) BIL-OlOt (Biosceptre); MDX-1203 (BMS); MDX-1204 (BMS); Necitumumab (BMS); CAN-4 (Cantargia AB); CDX-011 (Celldex); CDX1401 (Celldex); CDX301 (Celldex); U31565 (Daiichi Sankyo); patritumab (Daiichi Sankyo); tigatuzumab (Daiichi Sankyo); nimotuzumab (Daiichi Sankyo); DS-8895 (Daiichi Sankyo); DS-8873 (Daiichi Sankyo); DS-5573 (Daiichi
Sankyo); MORab-004 (Eisai); MORab-009 (Eisai); MORab-003 (Eisai); MORab-066 (Eisai); LY3012207 (Eli Lilly); LY2875358 (Eli Lilly); LY2812176 (Eli Lilly); LY3012217(Eli Lilly); LY2495655 (Eli Lilly); LY3012212 (Eli Lilly); LY3012211 (Eli Lilly); LY3009806 (Eli Lilly); cixutumumab (Eli Lilly); Flanvotumab (Eli Lilly); IMC-TRl (Eli Lilly); Ramucirumab (Eli Lilly); Tabahimab (Eli Lilly); Zanolimumab (Emergent Biosolution); FG-3019 (FibroGen); FPA008 (Five Prime Therapeutics); FP-1039 (Five Prime Therapeutics); FPA144 (Five Prime Therapeutics); catumaxomab (Fresenius Biotech); IMAB362 (Ganymed); IMAB027 (Ganymed); HuMax-CD74 (Genmab); HuMax-TEADC (Genmab); GS-5745 (Gilead); GS-6624 (Gilead); OMP-21M18 (demcizumab, GSK); mapatumumab (GSK); IMGN289 (ImmunoGen); IMGN901 (ImmunoGen); IMGN853 (ImmunoGen); IMGN529 (ImmunoGen); IMMU-130 (Immunomedics); milatuzumabdox (Immunomedics); 1MMU-H5 (Immunomedics); IMMU-132 (Immunomedics); IMMU-106 (Immunomedics); IMMU-102 (Immunomedics); Epratuzumab (Immunomedics); Clivatuzumab (Immunomedics); 1PH41 (Innate Immunotherapeutics); Daratumumab (Janssen/Genmab); CNTO-95 (Intetumumab, Janssen); CNTO-328 (siltuximab, Janssen); KB004 (KaloBios); mogamulizumab (Kyowa Hakko Kirrin); KW-2871 (ecromeximab, Life Science); Sonepcizumab (Lpath); Margetuximab (Macrogenics); Enoblituzumab (Macrogenics); MGD006 (Macrogenics); MGF007 (Macrogenics); MK-0646 (dalotuzumab, Merck); MK-3475 (Merck); Sym004 (Symphogen/Merck Serono); DH7E6 (Merck Serono); MOR208 (Morphosys); MOR202 (Morphosys); Xmab5574 (Morphosys); BPC-IC (ensituximab, Précision Biologics); TAS266 (Novartis); LFA 102 (Novartis); BHQ880 (Novartis/Morphosys); QGE031 (Novartis); HCD122 (lucatumumab, Novartis); LJM716 (Novartis); AT355 (Novartis); OMP-21M18 (Demcizumab, OncoMed); OMP52M5I (Oncomed/GSK); OMP-59R5 (Oncomed/GSK); vantictumab (Oncomed/Bayer); CMC-544 (inotuzumab ozogamicin, Pfizer); PF-03446962 (Pfizer); PF-04856884 (Pfizer); PSMA-ADC (Progenics); REGN1400 (Regeneron); REGN910 (nesvacumab, Regeneron/Sanofï); REGN421 (enoticumab, Regeneron/Sanofï); RG7221, RG7356, RG7155, RG7444, RG7l 16, RG7458, RG7598, RG7599, RG7600, RG7636, RG7450, RG7593, RG7596, DCDS3410A, RG7414 (parsatuzumab), RG7160 (imgatuzumab), RG7159 (obintuzumab), RG7686, RG3638 (onartuzumab), RG7597 (Roche/Genentech); SAR307746 (Sanofi); SAR566658 (Sanofi); SAR650984 (Sanofi); SAR153192 (Sanofi); SAR3419 (Sanofi); SAR256212 (Sanofi), SGN-LIVIA (lintuzumab, Seattle Genetics); SGN-CD33A (Seattle Genetics); SGN-75 (vorsetuzumab mafodotin, Seattle Genetics); SGN-19A (Seattle Genetics) SGN-CD70A (Seattle Genetics); SEA-CD40 (Seattle Genetics); ibritumomab tiuxetan (Spectrum); MLN0264 (Takeda); ganitumab (Takeda/Amgen); CEP-37250 (Teva); TB-403 (Thrombogenic); VB4-845 (Viventia); Xmab25l2 (Xencor); Xmab5574 (Xencor); nimotuzumab (YM Biosciences); Carlumab (Janssen); NY-ESO TCR (Adaptimmune); MAGE-A-10 TCR (Adaptimmune); CTL0I9 (Novartis); JCAR015 (Juno Therapeutics); KTE-C19 CAR (Kite Pharma); UCARTI9 (Cellectis); BPX-401 (Bellicum Pharmaceuticals); BPX-601 (Bellicum Pharmaceuticals); ATTCK20 (Unum Therapeutics); CAR-NKG2D (Celyad); Onyx-0I5 (Onyx Pharmaceuticals); HlOl (Shanghai Sunwaybio); DNX-2401 (DNAtrix); VCN-01 (VCN Biosciences); Colo-Adl (PsiOxus
Therapeutics); ProstAtak (Advantagene); Oncos-102 (Oncos Therapeutîcs); CG0070 (Cold Genesys); Pexa-vac (JX-594, Jennerex Biotherapeutics); GL-ONCl (Genelux); T-VEC (Amgen); G207 (Medigene); HFIO (Takara Bio); SEPREHVIR (HSV1716, Virttu Biologics); OrienXOlO (OrienGene Biotechnology); Reolysin (Oncolytics Biotech); SVV-OOl (Neotropix); Cacatak (CVA21, Viralytics); Alimta (Eli Lilly), cisplatin, oxaliplatin, irinotecan, folinic acid, methotrexate, cyclophosphamide, 5-iluorouracil, Zykadia (Novartis), Tafinlar (GSK.), Xaikori (Pfizer), Iressa (AZ), Gilotrif (Boehringer Ingelheîm), Tarceva (Astellas Pharma), Halaven (Eisai Pharma), Veliparib (Abbvie), AZD9291 (AZ), Alectinib (Chugai), LDK378 (Novartis), Genetespib (Synta Pharma), Tergenpumatucel-L (NewLink Genetics), GVlOOl (Kael-GemVax), Tivantinib (ArQule); Cytoxan (BMS); Oncovin (Eli Lilly); Adriamycin (Pfizer); Gemzar (Eli Lilly); Xeloda (Roche); Ixempra (BMS); Abraxane (Celgene); Trelstar (Debiopharm); Taxotere (Sanofi); Nexavar (Bayer); IMMU-132 (Immunomedics); E7449 (Eisai); Thermodox (Celsion); Cometriq (Exellxis); Lonsurf (Taiho Pharmaceuticals); Camptosar (Pfizer); UFT (Taiho Pharmaceuticals); and TS-l (Taiho Pharmaceuticals).
Modes of administration
Preferably, the compositions of the invention are to be administered to the gastrointestinal tract in order to enable delivery to and / or partial or total colonisation of the intestine with the bacterial strain of the invention. Generally, the compositions of the invention are administered orally, but they may be administered rectally, intranasally, or via buccal or sublingual routes.
In certain embodiments, the compositions of the invention may be administered as a foam, as a spray or a gel.
In certain embodiments, the compositions of the invention may be administered as a suppository, such as a rectal suppository, for example in the form of a theobroma oïl (cocoa butter), synthetic hard fat (e.g. suppocire, witepsol), glycero-gelatin, polyethylene glycol, or soap glycerin composition.
In certain embodiments, the composition of the invention is administered to the gastrointestinal tract via a tube, such as a nasogastric tube, orogastric tube, gastric tube, jejunostomy tube (J tube), percutaneous endoscopie gastrostomy (PEG), or a port, such as a chest wall port that provides access to the stomach, jéjunum and other suitable access ports.
The compositions of the invention may be administered once, or they may be administered sequentially as part of a treatment regimen. In certain embodiments, the compositions of the invention are to be administered daily.
In certain embodiments of the invention, treatment according to the invention is accompanied by assessment of the patient’s gut microbiota. Treatment may be repeated if delivery of and / or partial or total colonisation with the strain of the invention is not achieved such that efficacy is not observed, or treatment may be ceased if delivery and / or partial or total colonisation is successful and efficacy is observed.
In certain embodiments, the composition of the invention may be administered to a prégnant animal, for example a mammal such as a human in order to prevent an inflammatory or autoimmune disease developing in her child in utero and / or after it is bom.
The compositions of the invention may be administered to a patient that has been diagnosed with a disease or condition mediated by IL-17 or the Thl7 pathway, or that has been identified as being at risk of a disease or condition mediated by IL-17 or the Thl7 pathway. The compositions may also be administered as a prophylactic measure to prevent the development of diseases or conditions mediated by IL-17 or the Thl 7 pathway in a healthy patient.
The compositions of the invention may be administered to a patient that has been identified as having an abnormal gut microbiota. For example, the patient may hâve reduced or absent colonisation by the bacterium deposited under accession number NCIMB 42380.
The compositions of the invention may be administered as a food product, such as a nutritional supplément.
Generally, the compositions of the invention are for the treatment of humans, although they may be used to treat animais including monogastric mammals such as poultry, pigs, cats, dogs, horses or rabbits. The compositions of the invention may be useful for enhancing the growth and performance of animais. If administered to animais, oral gavage may be used.
Compositions
Generally, the composition of the invention comprises bacteria. In preferred embodiments of the invention, the composition is formulated in freeze-dried form. For example, the composition of the invention may comprise granules or gelatin capsules, for example hard gelatin capsules, comprising a bacterial strain of the invention.
Preferably, the composition of the invention comprises lyophilised bacteria. Lyophilisation of bacteria is a well-established procedure and relevant guidance is available in, for example, references [51-53].
Alternatively, the composition ofthe invention may comprise a live, active bacterial culture.
In preferred embodiments, the composition of the invention is encapsulated to enable delivery of the bacterial strain to the intestine. Encapsulation protects the composition from dégradation until delivery at the target location through, for example, rupturîng with Chemical or physical stimuli such as pressure, enzymatic activity, or physical disintegration, which may be triggered by changes in pH. Any appropriate encapsulation method may be used. Exemplary encapsulation techniques include entrapment within a porous matrix, attachment or adsorption on solid carrier surfaces, self18777 aggregation by flocculation or with cross-linking agents, and mechanical containment behind a microporous membrane or a microcapsule. Guidance on encapsulation that may be useful for preparing compositions of the invention is available in, for example, référencés [54] and [55],
The composition may be administered orally and may be in the form of a tablet, capsule or powder. Encapsulated products are preferred because the bacterium deposîted under accession number NCIMB 42380 may be an anaerobe. Other ingrédients (such as vitamin C, for example), may be included as oxygen scavengers and prebiotic substrates to improve the delivery and / or partial or total colonisation and survival in vivo. Alternatively, the probiotic composition of the invention may be administered orally as a food or nutritional product, such as milk or whey based fermented dairy product, or as a pharmaceutical product.
The composition may be formulated as a probiotic.
A composition of the invention includes a therapeutically effective amount of a bacterial strain of the invention. A therapeutically effective amount of a bacterial strain is sufficient to exert a bénéficiai effect upon a patient. A therapeutically effective amount of a bacterial strain may be sufficient to resuit in delivery to and / or partial or total colonisation ofthe patient’s intestine.
A suitable daily dose of the bacteria, for exampie for an adult human, may be from about l x 103 to about l x l0H colony forming units (CFU); for example, from about l x 107 to about l x 1OI0 CFU; în another example from about l x 106 to about l x l0w CFU.
In certain embodiments, the composition contains the bacterial strain in an amount of from about l x I06to about l x 1ON CFU/g, respect to the weight ofthe composition; for example, from about I x !0s to about I x 1O10 CFU/g. The dose may be, for example, 1 g, 3g, 5g, and 10g.
Typically, a probiotic, such as the composition of the invention, is optionally combined with at least one suitable prebiotic compound. A prebiotic compound is usually a non-digestible carbohydrate such as an oligo- or polysaccharide, or a sugar alcohol, which is not degraded or absorbed in the upper digestive tract. Known prebiotics include commercial products such as inulin and transgalactooligosaccharides.
In certain embodiments, the probiotic composition of the present invention includes a prebiotic compound in an amount of from about 1 to about 30% by weight, respect to the total weight composition, (e.g. from 5 to 20% by weight). Carbohydrates may be selected from the group consisting of: fructo- oligosaccharides (or FOS), short-chain fructo-olîgosaccharides, inulin, isomaltoligosaccharides, pectins, xylo-oligosaccharides (or XOS), chitosan-oligosaccharides (or COS), betaglucans, arable gum modified and résistant starches, polydextrose, D-tagatose, acacia fibers, carob, oats, and citrus fibers. In one aspect, the prebiotics are the short-chain fructo-oligosaccharides (for simplicity shown herein below as FOSs-c.c); said FOSs-c.c. are not digestible carbohydrates, generally obtained by the conversion of the beet sugar and including a saccharose molécule to which three glucose molécules are bonded.
The compositions of the invention may comprise pharmaceutically acceptable excipients or carriers. Examples of such suitable excipients may be found in the reference [56]. Acceptable carriers or 5 diluents for therapeutic use are well known in the pharmaceutical art and are described, for example, in reference [57]. Examples of suitable carriers include lactose, starch, glucose, methyl cellulose, magnésium stéarate, mannitol, sorbitol and the like. Examples of suitable diluents include éthanol, glycerol and water. The choice of pharmaceutical carrier, excipient or diluent can be selected with regard to the intended route of administration and standard pharmaceutical practice. The I0 pharmaceutical compositions may comprise as, or in addition to, the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s). Examples of suitable binders include starch, gelatin, naturel sugars such as glucose, anhydrous lactose, freeflow lactose, beta-lactose, com sweeteners, naturel and synthetic gums, such as acacia, tragacanth or sodium alginate, carboxymethyl cellulose and polyethylene glycol. Examples of suitable lubricants 15 include sodium oleate, sodium stéarate, magnésium stéarate, sodium benzoate, sodium acetate, sodium chloride and the like. Preservatives, stabilizers, dyes and even flavouring agents may be provided in the pharmaceutical composition. Examples of preservatives include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid. Antioxidanls and suspending agents may be also used.
The compositions of the invention may be formulated as a food product. For example, a food product may provide nutritional benefit in addition to the therapeutic effect of the invention, such as in a nutritional supplément. Similarly, a food product may be formulated to enhance the taste of the composition of the invention or to make the composition more attractive to consume by being more similar to a common food item, rather than to a pharmaceutical composition. In certain embodiments, 25 the composition of the invention is formulated as a milk-based product. The term milk-based product means any liquid or semi-solid inilk- or whey- based product having a varying fat content. The milk-based product can be, e.g., cow's milk. goat's milk, sheep's milk, skimmed milk, whole milk, milk recotnbined from powdered milk and whey without any processing, or a processed product, such as yoghurt, curdled milk, curd, sour milk, sour whole milk, butter milk and other sour 30 milk products. Another important group includes milk beverages, such as whey beverages, fermented milks, condensed milks, infant or baby milks; flavoured milks, ice cream; milk-containing food such as sweets.
In certain embodiments, the compositions of the invention contain a single bacteriai strain or species and do not contain any other bacteriai strains or species. Such compositions may comprise only de 35 iiihümis or biologically irrelevant amounts of other bacteriai strains or species. Such compositions may be a culture that is substantially free from other species of organism.
The compositions for use in accordance with the invention may or may not require marketing approval.
In some cases, the lyophilised bacterial slrain is reconstituted prior to administration. In some cases, the reconstitution is by use of a diluent described herein.
The compositions of the invention can comprise pharmaceutically acceptable excipients, dîluents or carriers.
In certain embodiments, the invention provides a pharmaceutical composition comprising: a bacterial straîn of the invention; and a pharmaceutically acceptable excipient, carrier or diluent; wherein the bacterial strain is in an amount sufficient to treat a disorder when administered to a subject in need thereof; and wherein the disorder is selected from the group consisting of asthma, allergie asthma. neutrophilie asthma, osteoarthritis, psoriatic arthritis, juvénile idiopathic arthritis, neuromyelitis optica (Devîc's disease), ankylosing spondylilis, spondyloarthritis, systemic lupus erythematosus, celiac disease, chronic obstructive pulmonary disease (COPD), cancer, breast cancer, colon cancer, lung cancer, ovarian cancer, uveitis, scleritis, vasculîtis, Behcet's disease, atherosclerosis, atopie dermatitis, emphysema, periodontitis, allergie rhinitis, and allograft rejection.
In certain embodiments, the invention provides pharmaceutical composition comprising: a bacterial strain of the invention; and a pharmaceutically acceptable excipient, carrier or diluent; wherein the bacterial strain is in an amount sufficient to treat or prevent a disease or condition mediated by IL-l 7 or the Thl7 pathway. In preferred embodiments, said disease or condition is selected from the group consisting of rheumatoid arthritis, multiple sclerosis, psoriasis, inflammatory bowel disease, Crohn’s disease, ulcerative colitis, celiac disease, asthma, allergie asthma, neutrophilie asthma, osteoarthritis, psoriatic arthritis, juvénile idiopathic arthritis, neuromyelitis optica (Devic's disease), ankylosing spondylitis, spondyloarthritis, systemic lupus erythematosus, chronic obstructive pulmonary disease (COPD), cancer, breast cancer, colon cancer, lung cancer, ovarian cancer, uveitis, scleritis, vasculîtis, Behcet's disease, atherosclerosis, atopie dermatitis, emphysema, periodontitis, allergie rhinitis, and allograft rejection.
In certain embodiments, the invention provides the above pharmaceutical composition, wherein the amount ofthe bacterial strain is from about l χ 103 to about l x 10 colony forming units per gram with respect to a weîght of the composition.
In certain embodiments, the invention provides the above pharmaceutical composition, wherein the composition is administered at a dose of l g, 3 g, 5 g or 10 g.
In certain embodiments, the invention provides the above pharmaceutical composition, wherein the composition is administered by a method selected from the group consisting of oral, rectal, subeutaneous, nasal, buccal, and sublingual.
In certain embodiments, the invention provides the above pharmaceutical composition, comprising a carrier selected from the group consisting of lactose, starch, glucose, methyl cellulose, magnésium stéarate, mannitol and sorbitol.
In certain embodiments, the invention provides the above pharmaceutical composition, comprising a diluent selected from the group consisting of éthanol, glycerol and water.
In certain embodiments, the invention provides the above pharmaceutical composition, comprising an excipient selected from the group consisting of starch, gelatin, glucose, anhydrous lactose, freeflow lactose, beta-lactose, com sweetener, acacia, tragacanth, sodium alginate, carboxymethyl cellulose, polyethylene glycol, sodium oleate, sodium stéarate, magnésium stéarate, sodium benzoate, sodium acetate and sodium chloride.
In certain embodiments, the invention provides the above pharmaceutical composition, further comprising at least one of a preservative, an antioxidant and a stabilizer.
In certain embodiments, the invention provides the above pharmaceutical composition, comprising a preservative selected from the group consisting of sodium benzoate, sorbic acid and esters of phydroxybenzoic acid.
In certain embodiments, the invention provides the above pharmaceutical composition, wherein said bacterial strain is lyophilised.
In certain embodiments, the invention provides the above pharmaceutical composition, wherein when the composition is stored in a sealed container at about 4°C or about 25°C and the container is placed in an atmosphère having 50% relative humidity, at least 80% of the bacterial strain as measured in colony forming units, remains after a period of at least about: l month, 3 months, 6 months, l year, l .5 years, 2 years, 2.5 years or 3 years.
Culturing methods
The bacterial strains for use in the présent invention can be cultured using standard microbiology techniques as detailed in, for example, référencés [58-60].
The solid or liquid medium used for culture may be YCFA agar or YCFA medium. YCFA medium may include (per 100ml, approximate values): Casitone (l.O g), yeast extract (0.25 g), NaHCO3 (0.4 g), cysteine (0.1 g), K2HPO4 (0.045 g), KH2PO4 (0.045 g), NaCl (0.09 g), (NH4)2SO4 (0.09 g), MgSO4-7H2O (0.009 g), CaCl2 (0.009 g), resazurin (0.I mg), hemîn (l mg), biotin (l pg), cobalamin (I pg),/j-aminobenzoic acid (3 pg), folie acid (5 pg), and pyridoxamine ( I5 pg).
Bacterial strains for use in vaccine compositions
The inventors hâve identified that the bacterial strains of the invention are useful for treating or preventing diseases or conditions mediated by IL-17 or the Thl 7 pathway. This is likely to be a resuit of the effect that the bacterial strains of the invention hâve on the host immune System. Therefore, the compositions of the invention may also be useful for preventing diseases or conditions mediated by IL-17 or the Thl7 pathway, when administered as vaccine compositions. In certain such embodiments, the bacterial strains of the invention may be killed, inactivated or attenuated. In certain such embodiments, the compositions may comprise a vaccine adjuvant. In certain embodiments, the compositions are for administration via injection, such as via subcutaneous injection.
General
The practice of the présent invention will employ, unless otherwise indicated, conventional methods of chemistry, biochemistry, molecular biology, immunology and pharmacology, within the skill of the art. Such techniques are explained fully in the literature. See, e.g., references [6I] and [62-68], etc.
The terni “comprising encompasses “including as well as “consisting” e.g. a composition “comprising” X may consist exclusively of X or may include something additional e.g. X + Y,
The term “about” in relation to a numerical value x is optional and means, for example, x+10%.
The word “substantially” does not exclude completely” e.g. a composition which is “substantially free” from Y may be completely free from Y. Where necessary, the word “substantially” may be omitted from the définition of the invention.
References to a percentage sequence identity between two nucléotide sequences means that, when atigned, that percentage of nucléotides are the same in comparing the two sequences. This alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in section 7.7.18 of ref. [69], A preferred aligmnent is determined by the Smith-Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix of 62. The SmithWaterman homology search algorithm is disclosed in ref. [70],
Unless specifically stated, a process or method comprising numerous steps may comprise additional steps at the beginning or end of the method, or may comprise additional intervening steps. Also, steps may be combined, omitted or performed in an alternative order, if appropriate.
Various embodiments of the invention are described herein. It will be appreciated that the features specified in each embodiment may be combined with other specified features, to provide further embodiments. In particular, embodiments highlighted herein as being suitable, typical or preferred may be combined with each other (except when they are mutually exclusive).
MODES FOR CARRYING OUT THE INVENTION
Example 1 — Effîcacy of bacterial inocula in a mouse model of house dust mite-induced asthma
Summarv
Mice were administered with compositions comprising bacterial strains according to the invention 5 and were subsequently challenged with house dust mite (HDM) extract to elicit an allergie inflammatory response. The inflammatory response to HDM includes éosinophilie and neutrophilie components, is mediated by IL-17 and the Thl7 pathway, and is a model for asthma. The magnitude and characteristics of the inflammatory response exhibited by mice treated with compositions of the invention were compared to control groups. The compositions of the invention were found to 10 alleviate the inflammatory response, and to reduce recruitment of eosinophils and neutrophils, indicating that they may be useful for treating IL-17-mediated conditions such as eosinophilia, neutrophilia and asthma.
S train
751 : bacterium deposited under accession number NCIMB 42380
Study design
Groups:
1. Négative control group. Treatment with vehicle control (per oral).
4. Treatment with therapeutic bacteria inoculum strain 751 (per oral).
7. Positive control group. Treatment with Dexamethasone (i.p.).
8. Untreated Control Group.
Number of mice per group = 5
Day -14 to day 13: Daily administration of vehicle control per oral (Group l).
Day -14 to day 13: Daily administration of therapeutic bacteria inoculum per oral (Group 2-6).
Day 0, 2, 4, 7, 9, 11 Administration of 15ug HDM (house dust mite extract - Catalogue number: XPB70D3A25, Lot number: 231897, Greer Laboratories, Lenoir, NC, USA) in a volume of 30ul PBS per nasal (Group 1-8).
Day 0, 2, 4, 7, 9, 11 Administration of Dexamethasone (i.p., 3mg/kg, Sigma-Aldrich, Catalogue number DI 159) (Group 7).
Day 14 Sacrifice of ail animais for analysis.
Total number of mice = 40.
Endpoints and analysis
On day 14 animais were sacrificed by léthal intraperitoneal injection with pentabarbitol (Streuli Pharma AG, Uznach, Cat: 1170139A) immediately followed by a bronchoalveolar lavage (BAL).
Cells were isolated from the BAL (bronchoalveolar lavage) fluid and differential cell counts performed (200 cell counts/ samples).
Material and Methods
Mice. Female 7 week old BALB/c mice were purchased from Charles River Laboratories and randomly allocated to cages totally 5 mice per cage (Ventilated cages sourced from Indulab AG, Gams, Switzerland Cage type: “The SealsafeTM - IVC cage. Product number 1248L). Cages were labeled with study number, group number and experimental starting date. Mice were monitored weekly and acclimatized to facility for 7 days prior to initiation of study (Study Day -I4). Animais were 8 weeks old on Study Day -14, Potable water and food were available ad libitum. Cage enrichment was présent. Daily care of the animais was performed according to local authorization license number 2283.1 (issued and approved by: Service de la consommation et des affaires vétérinaires du Canton de Vaud). Potable water and food were available ad libitum and refreshed once daily. Cage enrichment was présent. Animal welfare régulations were observed as given by official authorities of Switzerland under ordînance 455.163 of the FVO (Fédéral Veterinary Office) on laboratory animal husbandry, production of genetically modified animais, and methods of animal expérimentation.
Culturing of bacteria inoculum. Within a stérile workstation, a cryo-vial of bacteria was thawed by wanning in gloved hand and -0.7 ml of contents injected into a Hungate tube (Cat Number, 1020471, Glasgerâtebau Ochs, Bovenden-Lenglem, Germany), containing 8 ml of anaérobie YCFA. Two tubes per strain were usually prepared. The Hungate tubes were then incubated (static) at 37°C for 16h (strain 751).
Culturing of vehicle control. A Hungate tube containing 8 ml of anaérobie YCFA was incubated (static) at 37°C for 16h.
Administration of bacteria inoculum or vehicle control. 400ul of cultured bacteria inoculum or vehicle control were administered per day per oral gavage.
Intranasal sensitization. Mice were anesthetized by i.p. injection with 9.75 mg xylasol and 48.75 mg ketasol per kg (Dr. E. Graeub AG, Bem, Switzerland) and administered with 15ug of HDM (Catalogue number: XPB70D3A25, Lot number: 231897, Greer Laboratories, Lenoir, NC, USA) in a volume of 30ul PBS per nasal.
Préparation and administration of positive control compound Dexamethasone. Dexamethasone 21-phosphate disodium sait (Sigma-Aldrich, Catalogue number Dl 159, Lot N° SLBD. 1030V) was solved in H2O and administered to the animais in a dose of 3mg/kg in a volume of 200ul per oral at days indicated in study protocol above.
Terminal procedure. On day 14 animais were sacrificed by léthal i.p. injection with pentabarbitol (Streuli Pharma AG, Uznach, Cat: 1170139A) immediately followed by bronchoalveolar lavage (BAL) in 500 ul of saline.
Measurement of cellular infiltrâtes into BAL. Cells were isolated from the BAL fluid and differential cell counts were performed based upon standard morphological and cytochemical criteria.
Graphs and statistical analysis. Ail graphs were generated with Graphpad Prism Version 6 and a one-way ANOVA was applied. Results from the statistical analysis were provided with the individual data tables. Error bars represent Standard Error ofthe Mean (SEM).
Results and analysis
The results of the experiments are shown in Figures l-9.
No morbidity or mortality was noted in the mice treated with the bacteria or the vehicle. The two Controls, vehicle treatment (négative control) and the dexamethasone treatment (positive control) behaved as expected, with impaired eosinophilia and neutrophilia noted following dexamethasone treatment.
The most important results of this experiment are displayed in Figures 6 and 7, which report on the total number and percentage of neutrophils detected in bronchiolar lavage following challenge with H DM. Strain 751 reduced total neutrophils and the proportion of neutrophils in BAL relative to the vehicle-only control.
Example 2 - Efficacy of bacterial inocula in a mouse niodel of severe neutrophilie asthma
Summarv
Mice were administered with compositions comprising bacterial strains according to the invention and were subsequently sensitised with subeutaneous administrations of house dust mite (HDM) extract and challenged with an intranasal administration of HDM in order to model the inflammatory response of severe neutrophilie asthma. The magnitude and characteristics of the inflammatory response exhibited by mice treated with compositions of the invention were compared to control groups. The compositions of the invention were found to alleviate the inflammatory response, and in particular to reduce recruitment of neutrophils, in a manner comparable to the positive control comprising administrations of anti-IL-17 antibodies. The data therefore indicate that the compositions of the invention may be useful for treating IL-17- and Thl7-mediated conditions such as neutrophilia and asthma.
Strain
751 : bacterium deposited under accession number NCIMB 42380
Studv design
Groups:
l. Négative control group. Treatment with vehicle control (per oral).
4. Treatment with therapeutic bacteria inoculum strain 751 (per oral).
7. Positive control group. Treatment anti-IL-17 (i.p.).
8. Untreated Control Group.
9: Healthy mîce (baseline).
Number of mice per group (Group l -8) = 5
Day -14 to day 17: Daily administration of vehicle control per oral (Group l).
Day -14 to day 17: Daily administration of therapeutic bacteria inocuhim per oral (Group 2-6).
Day 0: Sensitization with HDM in CFA (s.c.) (Group l-8).
Day 7: Sensitization with HDM in CFA (s.c.) (Group l-8).
Day 13, 15, 17: Administration of anti IL-17 neutralizing antibody per i.p. (Group 7).
Day 14, 15, 16, 17: Challenge with HDM in 30ul PBS per nasal (Group l-8).
Day 18: Sacrifice of ail animais for analysis.
Endpoints and analysis:
On day 14 animais were sacrificed by léthal intraperitoneal injection with pentabarbitol (Streuli Pharma AG, Uznach, Cat: I 170139A) immediately followed by a bronchoalveolar lavage (BAL). Cells were isolated from the BAL fluid and dififerential cell counts performed (200 cell counts/ samples).
Material and Methods.
Mice. Female 7 week old C57BL/6 mice were purchased from Charles River Laboratories and randomly allocated to cages totally 5 mice per cage (Ventilated cages sourced from Indulab AG, Gams, Switzerland Cage type: “The SealsafeTM - IVC cage. Product number 1248L). Cages were labelled with study number, group number and experimental starting date. Mice were monitored weekly and acclimatîzed to facility for 7 days prior to initiation of study (Study Day -14). Animais were 8 weeks old on Study Day -14. Potable water and food were available ad libitum. Cage enrichment was présent. Daily care of the animais was performed according to local authorization license number 2283.1 (issued and approved by: Service de la consommation et des affaires vétérinaires du Canton de Vaud). Potable water and food were available ad libitum and refreshed once daily. Cage enrichment was présent. Animal welfare régulations were observed as given by official authorities of Switzerland under ordinance 455.163 of the FVO (Fédéral Veterinary Office) on laboratory animal husbandry, production of genetically modified animais, and methods of animal expérimentation.
Culturing of bacteria inoculum. Within a stérile workstation, a cryo-vial of bacteria was thawed by warming in gloved hand and -0.7 ml of contents injected into a Hungate tube (Cat Number, 1020471, Glasgerâtebau Ochs, Bovenden-Lenglem, Germany), containing 8 ml of anaérobie YCFA. Two tubes per strain were usually prepared. The Hungate tubes were then incubated (static) at 37°C for 16h (strain 751).
Culturing of vehicle control· A Hungate tube containing 8 ml of anaérobie YCFA was incubated (static) at 37°C for 16h.
Administration of bacteria inoculum or vehicle control. 400ul of cultured bacteria inoculum or vehicle control were administered per day per oral gavage.
HDM sensitization. 50 pg of HDM (Catalogue number: XPB70D3A25, Lot number: 231897, Greer Laboratories, Lenoir, NC, USA) in PBS was emulsified in equal volume of complété Freund’s adjuvant (CFA Chondrex Inc. Washington, USA) and administered subcutaneously in a volume of 200 μ|, twice over two weeks on opposite flanks. A week after the second immunization, mice were anesthetized by i.p. injection with 9.75 mg xylasol and 48.75 mg ketasol per kg (Dr. E. Graeub AG, Bem, Switzerland) and then given intranasal challenges of 15 pg of HDM in a volume of 30ul PBS on 4 consecutive days. Analysis was performed one day after the final challenge.
Préparation and administration of positive control compound anti mouse IL-17 antibody. AntiIL-17 neutralizing antibody was sourced from Bio X Cell and was stored at 4qC (Clone 17F3, Cat. Number BE0173, Bio X Cell) and administered per i.p. at a dose of 12.5 mg/kg at days indicated in study protocol above.
Terminal procedure. On day 18 animais were sacrificed by léthal i.p. injection with pentabarbitol (Streuli Pharma AG, Uznach, Cat: Il70139A) immediately followed by bronchoalveolar lavage (BAL) in 500 ul of saline,
Measurement of cellular infiltrâtes into BAL. Cells were isolated from the BAL fluid and differential cell counts were performed based upon standard morphological and cytochemical criteria.
Graphs and statistical analysis. Ail graphs were generated with Graphpad Prism Version 6 and a one-way ANOVA was applied. Results from the statistical analysis are provided with the individual data tables. Error bars represent Standard Error of the Mean (SEM).
Results and analysis
The results of the experiment are shown in Figures 10-18.
No morbidity or mortality was noted in the mice treated with the bacteria or the vehicle. As shown in Figures 15 and 16, strain 751 was highly efficacious in alleviating the magnitude of the neutrophilie inflammatory response. Indeed, the treatment with strain 751 showed comparable results to treatment with anti-IL-17 antibodies. In addition, strain 751 reduced eosînophil numbers relative to the Controls, as shown in Figures 11 and 12.
Example 3 - Efftcacy of bacterial inocula to treat arthritis in a type II collagen-induced arthritis mouse model
Materials and methods
Strains
751 : bacterium deposited under accession number NCIMB 42380
Bacterial cultures
Bacterial cultures were grown up for administration in an anaérobie workstation (Don Whitley Scientific).
Bacterial strain #751 was grown using glycerol stocks. The glycerol stocks were stored at -80°C. Three times per week, glycerol stocks were thawed at room température and streaked on YCFA plates. A new glycerol aliquot was used on each occasion. Bacteria were allowed to grow on a given plate for up to 72 hours.
Solutions to be administered to the animais were prepared twice daily with an eight hour interval for moming (AM) and aftemoon (PM) treatments. A bacterial colony was picked from the streaked plate and transferred into a tube containing YCFA media. Bacterial strain #751 was allowed to grow for 16 hours before AM administrations. Bacteria were sub-cultured at l% into YCFA media for PM administrations. OD values were recorded for each strain after moming and afternoon treatment préparations.
Type II collagen-induced arthritis mouse model
Adult male DBA/l mice were randomly allocated to experimental groups and allowed to acclimatise for two weeks. On Day 0, animais were administered by subeutaneous injection with 100 microliters of an émulsion containing 100 micrograms of type II collagen (Cil) in incomplete’s Freund’s adjuvant supplemented with 4 mg/ml Mycobacterium tuberculosis H37Ra. On Day 21, animais were administered by subeutaneous injection with a booster émulsion containing 100 pg of type II collagen in incomplète Freund’s adjuvant.
Treatments were given according to the administration schedule below. From Day -14 until the end of the experiment on Day 45, animais were weighed three times per week. From Day 21 until the end of the experiment, animais were scored three times per week for clinical signs of arthritis to include swelling of the hind- and front paws, radio-carpal (wrist) joints and tibio-tarsal (ankle) joints.
On Day 45 mice were culled and terminal blood samples were taken for cytokine analysis.
On Day -14, Day 0 and Day 45, faecal samples were collected for microbiological analysis, immediately snap-frozen and stored at -80°C.
The collagen-induced arthritis (CIA) mouse model is a well-established mouse model for rheumatoid arthritis [7I], Immunisation with Cil causes a pathogenesis that includes several important pathological features of rheumatoid arthritis, including synovial hyperplasia, mononuclear cell infiltration and cartilage dégradation. Significantly, the development of CIA is mediated by Thl7 cells through sécrétion of IL-17A [72], The immune response underlying the arthritis model is enhanced by the use of Freund’s adjuvant supplemented with Mycobacterium tuberculosis.
On Day 21, spleens were collected from three satellite animais in each group. Cells were cultured for 72 hours in the presence or absence of type II collagen. Cytokines, including TNF-α, lL-6, IFN-γ, IL4, IL-IO and IL-l7, were quantified in the culture supematants and in terminal sérum by Luminex. Cell prolifération was quantified using a tritiated thymidine incorporation method.
Treatment Groups and Dosages
Ail Groups were n=l 5 (n=l2 for the main study group and n=3 for satellite groups)
The vehicle used for the biotherapeutics was Yeast extract-Casitone-Fatty Acids (YCFA) medium.
Group Dose A dministration Disease Induction
Route Regimen
I Vehicle 5 ml/kg PO BID: Day -14-End Day 0: Collagen/CFA, once, SC Day 21: Collagen/IFA, once, SC
2 Biotherapeutic #751 5 ml/kg PO BID: Day -14-End
PO: oral gavage, SC: subcutaneous injection, BID: twice a day, CFA: complété Freund’s adjuvant.
Bodyweights
From Day -14 until the end of the experiment, animais were weighed three times per week. Data were graphed (Mean ± SEM).
Non-specifïc clinical observations
From Day -14 until the end ofthe experiment, animais were checked daily for non-specific clinical signs to include abnormal posture (hunched), abnormal coat condition (piloerection) and abnormal activity levels (reduced or increased activity),
Clinical Observations
From Day 21 until the end of the experiment on Day 45, animais were scored three times per week for clinical signs of arthritis to include swelling of the hind- and front paws, radio-carpal (wrist) joints and tibîo-tarsal (ankle) joints. Each limb was scored using the followîng scale: (0) normal, ( l ) slight swelling, (2) mild swelling, (3) moderate swelling and (4) severe swelling. A clinical score was calculated by adding each limb score. The maximum possible clinical score for an animal was ( 16). Animais with a score equal to ( 12) on two consecutive occasions and animais with a score greater than (12)on any one occasion were culled. Data were graphed (Mean ± SEM).
Cell prolifération analysis
On Day 21, three satellite animais per group were culled and spleens were dissected out. Spleen cells were cultured for 72 hours in presence or absence of type II Collagen. After 72 hours, cells were pulsed ovemight in the presence of tritiated thymidine. Cell prolifération was quantified by measuring thymidine incorporation. Data were graphed (Mean ± SEM). Supematants were taken and tested for the presence of key cytokines.
Cytokine analysis
Terminal supematants from the spleen cell cultures were tested in order to quantitate TNF-α. IL-6, IFN-γ, IL-4, IL-10 and IL-17 by Luminex. Data were graphed (Mean ± SEM).
Microbiological analysis
On Day -14, Day 0 and Day 45, faecal sampies were collected from each animal, immediately snapfrozen, and stored at -80°C. Caeca (including content) were immediately snap-frozen and stored at -80°C. A bacterial identification test was performed daily by plating the bacteria.
Histopathology
At the end of the experiment. hind paws were stored in tissue fixative. Sampies were transferred into décalcification solution. Tissue sampies were processed, sectioned and stained with Haematoxylin & Eosin. Sections were scored by a qualified histopathologist, blind to the experimental design, for signs of arthritis to include inflammation, articular cartilage damage and damage to the underlying metaphyseal bone. A detailed scoring system was used (see below). Data were graphed (Mean ± SEM). Raw and analysed data were provided as well as représentative pictures.
Table 1: Histopathology Scoring System
Grade Description
Inflammation
Normal joint
Mild synovial hyperplasîa with inflammation dominated by neutrophils. Low numbers of neutrophils and macrophages in joint space.
Synovial hyperplasîa with moderate to marked inflammation involving both neutrophils and macrophages. Neutrophils and macrophages in joint space; may be some necrotic tissue débris.
Synovial hyperplasîa with marked inflammation involving both neutrophils and macrophages. Loss of synoviocyte lining. Inflammation may extend from synovium to surrounding tissue including muscle. Numerous neutrophils and macrophages in joint space, together with significant necrotic
tissue débris.
Articular cartilage damage
0 l 2 3 Normal joint Articular cartilage shows only mild degenerative change. Early pannus formation may be present peripherally. Articular cartilage shows moderate degenerative change and focal loss. Pannus formation is present focally, Significant disruption and loss of articular cartilage with extensive pannus formation.
Damage to the underlying metaphyseal bone
0 1 2 3 Normal joint No change to underlying metaphyseal bone. May be focal necrosis or fibrosis of metaphyseal bone. Disruption or collapse of metaphyseal bone. Extensive inflammation, necrosis or fibrosis extending to medullary space ofthe metaphysis.
Residts and analysis
Survival and Νοη-specifïc Ciinical Observations
Some animais were culled prior to the scheduled end of the study due to the severîty of the ciinical signs of arthritis or due to the severîty of the non-specifïc ciinical observations.
Three animais were culled or found dead or culled during the pre-treatment period (Day -14 to Day 0): one animal în Group l (vehicle-treated, animal arrived from supplier with broken leg and was culled) and two animais in Group 2 (biotherapeutic #75l-treated, possible lung dosing on first pretreatment day, and post-dose ciinical signs on second pre-treatment day).
Eight animais were culled due to the severîty of the ciinical signs of arthritis: five animais in Group l 10 (vehicle-treated) and three animais in Group 2 (biotherapeutic #75l-treated).
Four animais were culled due to the severîty of the non-specîfic ciinical signs including abnormal posture (hunched), abnormal coat condition (piloerection), abnormal activity levels (reduced activity): three animais in Group l (vehicle-treated) and one animal in Group 2 (biotherapeutic #751treated).
Bodyweights
Bodyweîght data recorded from Day -14 untîl Day 0 and expressed as a percentage of the initial (Day -14) bodyweights were analysed by two-way AND VA followed by Dunnett's post-test for multiple comparisons with Day -14 then for multiple comparison with the vehicle-treated group. The data are presented in Figure 19. Data from animais culled prior to the scheduled end of the experiment were excluded from the analyses.
When compared to Day -14, twîce daily administrations by oral gavage induced a significant bodyweight loss in the vehicle-treated group on Day -9 and Day -7.
The bodyweights measured between Day -14 and Day -l in the biotherapeutic-treated groups did not differ from the bodyweights measured in the vehicle-treated group on any given day.
Bodyweight data recorded from Day 0 until Day 28 and expressed as a percentage of the initial (Day 0) bodyweights were analysed by two-way ANOVA followed by Dunnett’s post-test for multiple comparisons with Day 0 in the Vehicle group then for multiple comparison with the vehicle-treated group. The data are presented in Figure 20. Data from animais culled prior to the scheduled end of the experiment and from Satellite animais were excluded from the analyses. Day 28, Day 35 and Day 42 data were further analysed by one-way ANOVA followed by Dunnett’s post-test for multiple comparisons to the vehicle-treated group.
The onset of clînical signs of arthritis was associated with a significant bodyweight loss on Day 26 and Day 28 (p < 0.0001 ) when compared to Day 0 in the vehicle-treated group.
There was no significant différence between experimental groups on Day 35 or Day 42.
Clinical Observations
Clînical score data were analysed by two-way ANOVA followed by Dunnett’s post-test for multiple comparisons between days in the vehicle-treated group then for multiple comparisons between experimental groups and the vehicle-treated group each day. The data are presented in Figure 2l. Data recorded from animais culled prior to the end of the experiment were excluded from the analysis. When animais were culled due to the severity of the clinical signs of arthritis, the last recorded score was reported for the following days and used in the statistical analyses.
A significant increase of the clinical scores was observed in the vehicle-treated group from Day 28 until Day 45 (p < 0.0001 ) when compared to Day 21.
Biotherapeutic #751 induced a réduction of the clînical scores when compared to the vehicle-treated group from Day 31 until Day 45, although the différence was non-significant.
Cell prolifération analysis
To validate the assay, splénocytes were cultured in the presence of soluble anti-CD3 and anti-CD28 (anti-CD3/CD28) as positive control stimuli to confirm the proliférative potential of the cells.
Strong proliferative responses to anti-CD3/CD28 were seen in ail experimental groups, showing cells were healthy, viable and able to respond to activation signais.
To test the proliférative response in presence of Collagen II (Cil), splénocytes were cultured in the presence of Cil at 50 pg/ml. Splénocyte proliférative response to Cil were analysed by two-way ANOVA followed by Sydak’s post-test for multiple comparisons between unstimulated and CIIstimulated splénocytes and one-way ANOVA followed by Dunnett’s post-test for comparison of CIIstimulated response in different experimental groups with the vehicle-treated group. The data are presented in Figure 22.
Cil induced a highly significant increase of 3H-thymidine incorporation (cpm) when compared to the unstimulated splénocytes in the vehicle-treated group (p < 0.0001 ).
The groups treated with biotherapeutic #75ldemonstrated significantly lower levels of CH-induced splénocyte prolifération than the vehicle-treated group.
Cytokine levels in tissue culture supernatants
Levels of each cytokine were measured in tissue culture supernatants derived from anti-CD3/CD28 stimulated cultures by luminex analysis. These showed robust responses for ail cytokines measured (mean levels in vehicle group were as follows: IL-4 = 6,406 pg/ml; IL-6 = 306 pg/ml; IL-10 = 10,987 pg/ml; IL-17A = 11,447 pg/ml; IFN-γ = 15.581 pg/ml; TNF-α = 76 pg/ml).
The following sections summarise the data obtained from the Collagen Il-stimulated cultures. Where applicable, statistical analyses of the différences between cytokine levels in supernatants of unstimulated and ClI-stimulated splénocytes were conducted using two-way ANOVA followed by Sidak’s post-test for multiple comparisons, while one-way ANOVA followed by Dunnett’s post-test was used for comparison of ClI-stimulated response in biotherapeutic-treated groups with the vehicle-treated group. There was no significant différence in cytokine levels between the groups in both cases. This is likely due to the small sample size used (n=3).
In order to more accurately present the distribution of the data for the cytokines with substantîal spread of the data, these are presented as scatter plots.
The group means of 1L-4 in tissue culture supernatants after stimulation with CH were <5pg/ml. These are not considered biologically significant and not included here. The group means of TNF-a in tissue culture supernatants after stimulation with collagen were below limit of quantitation.
Supernatant levels of IFN-γ (Figure 23)
Along with IL-17, IFN-γ is the major cytokine driving disease in the CIA model. The scatter plot in Figure 23 demonstrates IFN-γ levels after Cil stimulation, with group médian being higher for the Vehicle-treated group compared to the biotherapeutic. The outlier resuit from the same group 2 subject is responsible for the higher médian in this group for IFN-γ and IL-10.
Supernatant levels of IL-17A (Figure 24)
Levels of IL-17A were 50pg/ml in ClI-stimulated cultures for the Vehicle-treated group. The levels of this cytokine appeared to be lower in the biotherapeutic group compared to the Vehicle-treated.
Supernatant levels of IL-10 (Figure 25)
Levels of IL-10 in Vehicle-treated group were 13 pg/ml and 2.1 pg/ml for Cll-stimulated, and media control cultures, respectively. Higher levels of IL-10 (which is an anti-inflammatory cytokine) for the vehicle-treated group may be expected because inflammation and pro-inflammatory cytokine induction could be accompanied by an anti-inflammatory feedback mechanism.
Supernatant levels of IL-6 (Figure 26)
Inflammatory cytokines such as IL-6 and TNF-α are not typically produced at high levels in anti-CII cultures. However, their levels may be altered as a resuit of immune modulation. Levels of IL-6 in Cll-stimulated cultures were tnodest, reaching lOpg/ml. Although higher than in media control cultures, these différences were too small to provide rationale for performing statistical analyses.
Microbiological analysis
Bacterial growth was confirmed by measuring the optical density at 600 nm using a spectrophotometer. Bacterial identity was confirmed by comparing streaked plate pictures to reference pictures.
Following the improved bacterial préparation method. consistently high doses of bacterial straîn were administered from Day -2 and Day -3 as indicated by the high OD values measured.
Faecal samples were collected and snap-frozen on Day -14, Day 0 and at termination.
Histopathology
The histopathology results are shown in Figures 66-70. As expected for this model, intra-individual and inter-individual variabîlity was observed in tenus of the presence/absence of arthritis or the severity of change présent.
The nature of the pathology was as expected for this model, with extensive mixed chronic-active inflammation of the synovium and bursa extending to involve the peri-articular soft tissues (muscle, adipose tissue, dermal collagen). In the most severely affected joints there was articular cartilage degeneration and loss with intra-articular débris and inflammation and disruption of the joint and bone structure by fibrosis and inflammation.
The incidence of histopathological changes was: vehicle - 80% (16/20); Biotherapeutic #751 - 45% (9/20). Treatment with Biotherapeutic #751 reduced the incidence of histopathological scores in mouse hind limbs when compared to the vehicle-treated group (see Figures 66-69). Histopathology scores were analysed by one-way ANOVA for non-parametric data (Kruskal-Wallis test) followed by
Dunn’s post-test for multiple comparisons to the vehicle-treated group. Biotherapeutic #751 induced a significant réduction of the joint inflammation scores observed in histopathology when compared to the vehicle-treated group (p < 0.01). Biotherapeutic #751 induced a significant réduction ofthe cartilage damage scores observed in histopathology when compared to the vehicle-treated group (p < 0.001). Biotherapeutic #751 induced a significant réduction of the bone damage scores observed in histopathology when compared to the vehicle-treated group (p < 0.001). Biotherapeutic #751 induced a significant réduction of the total histopathology scores when compared to the vehicle-treated group (p < 0.01).
Summary
Increased clinical scores were observed from Day 28 after the first administration of type II collagen, as expected in this model of arthritis in DBA/1 mice. Biotherapeutic #751 was shown to be effective at treating arthritis in this model and Biotherapeutic #751 was effective for reducing the severity of the clinical scores. Biotherapeutic #751 was also effective for reducing pathological disease in the joints, as demonstrated in the histopathological analysis.
Proliférative recall responses to Collagen II were seen in splénocyte cultures from ail experimental groups. The collagen-specific response was significantly reduced following treatment with biotherapeutic #751 (Group 2).
Most of the T cell cytokines tested showed détectable increases between Collagen II-stimulated and media Controls in the Vehicle-treated group. These increases were not as obvious in the biotherapeutic-treated group. This broadly supports the proliférative recall responses to Collagen II described above.
There was evidence of suppression of the Thl/Thl7 axis, which is the pathogenic response in this model and în human RA. Corrélation of reduced levels of cytokines with reduced prolifération is suggestive of immune modulation. There was no evidence that this modulation resulted either from enhanced levels of Th2 associated IL-4 or with increases in the immune modulating cytokine, IL-10.
Example 4 - Further analysis of the effect of bacterial inocula in the mouse model of house dust mite-induced asthma
The mîce tested in Example 1 were subjected to further analyses to further characterise the effect of the compositions ofthe invention on the allergie asthma inflammatory response.
Materials and methods
Blood withdrawal and sérum préparation on day 14. Blood samples of animais were collected via cardiac puncture. Sérum was isolated from the blood sample by centrifugation for 5 min at 14000g and stored at -20 °C.
Organ removal on day 14. Collection of the left lung lobe in fonnalin for follow-on histological analysis. Collection of the right lung lobes (ail remaining lobes) and removal of sérum for snap freezing and follow-on analysis. Remaining BAL fluid was snap frozen for follow-on analysis.
Measurement of antibody levels in sérum and BAL fluid
Total IgE and house-dust-mite (HDM) spécifie IgGl antibody production were measured in the BAL and sérum by EL1SA assay.
Isolation of lung and histological analysis
Left lung lobes were fixed in formalin followed by embedment in paraffin, sectioning, and staining with hematoxylin and eosin and PAS. Subséquent histological scoring was performed biinded as followed: Five random fields of view per sample were scored for inflammation (peribronchial infiltration and perivascular infiltration) and mucus production. Inflammatory infiltration was scored with the following grading system:
- normal
- mild inflammatory infiltrâtes
- moderate inflammatory infiltrâtes
- marked inflammatory infiltrâtes
- severe inflammatory infiltrâtes
- very severe inflammatory infiltrâtes
In each field of view, airways were measured in size and mucus cell numbers were quantified/ um.
Measurement of inflammatory mediators in lung tissue
Right lung lobes (ail remaining lobes) isolated for quantification of inflammatory mediators were snap frozen for subséquent measurement of CCL11, IFN-gamma, IL-1 alpha, IL-1 beta, IL-4, IL-5, IL-9, IL-17A, CXCL1, CCL3, CXCL2 and CCL5 by commercially available multiplex assay (Merck-Millipore). Analysis was performed according to the manufacturées instructions.
Results and analysis
The results of the experiments are shown in Figures 28-46.
In support of the findings described in Example 1, analysis of the cellular infiltrâtes in the lung tissue of mice treated with strain 751 showed a notable and statistically significant réduction in mean inflammation score (see Figures 32 and 34).
Antibody levels in the BAL fluid and sérum were analysed (see Figures 28-31). No clear effect of the bacterial treatment on sérum antibody levels was observed. This may reflect a failure in the experiment, because the spread of data and the error bars for each treatment are large, and the positive and négative Controls do not appear to hâve behaved as would be expected. Also, the baseline sérum antibody levels could hâve masked any changes.
Similarly, no clear effect of the bacterial treatment on cytokine levels in lung tissue was observed (see Figures 36-46). Again, this may reflect a failure in the experiment, because the spread of data and the error bars for each treatment are large, and the positive and négative Controls do not appear to hâve behaved as would be expected. It is also possible that the mechanism of action involved influences earlier cytokine responses that were no longer détectable on day 4 post the final HDM airway challenge. Some care should be taken when interpreting the cytokine data in the current study, due to the variability in the levels detected. This variability could în part be explained by the fact that the lung tissue was separated for the different analyses, and thus one lung lobe might not hâve been fully représentative or comparable to the same lobe in other mice due to patchy distribution of the inflammation.
Example 5 - Further analysis of the effect of bacterial inocula in the mouse model of severe neutrophilie asthma
The mice tested in Example 2 were subjected to further analyses to further characterise the effect of the compositions of the invention on the neutrophilie response associated with severe asthma.
Materials and methods
Organ removal on day 18. Collection of the left lung lobe in formalin for follow-on histological analysis. Collection of the right lung lobes (ail remaining lobes) and removal of sérum for snap freezing and follow-on analysis. Remaining BAL fluid was snap frozen for follow-on analysis.
Measurement of inflammatory mediators in lung tissue (follow-on analysis). Right lung lobes (ail remaining lobes) isolated for quantification of inflammatory mediators were snap frozen for subséquent measurement of IFN-gamma, 1L-I alpha, 1L-1 beta, CXCL1, CCL3, CXCL2, CCL5, IL17A, TNF-alpha, IL-17F, IL-23 and IL-33 by commercially available multiplex assay (MerckMîllipore). Analysis was performed according to the manufacturer’s instructions.
Measurement of antibody levels in sérum and BAL fluid (follow-on analysis). House-dust-mite (HDM) spécifie IgGl and IgG2a antibody production were measured in the BAL and sérum by ELISA assay.
Isolation of lung and histological analysis (followr-on analysis). Left lung lobes were fixed in formalin followed by embedment in paraffin, sectioning, and staining with hematoxylin and eosin and PAS. Subséquent histological scoring was performed blinded as followed: Five random fields of view per sample were scored for inflammation (peribronchial infiltration and perivascular infiltration) and mucus production. Inflammatory infiltration was scored with the following grading system:
- normal
- mild inflammatory infiltrâtes
- moderate inflammatory infiltrâtes
- marked inflammatory infiltrâtes
- severe inflammatory infiltrâtes
- very severe inflammatory infiltrâtes
Results and analysis
The results of the experiments are shown in Figures 47-64.
Further analysis of antibody levels revealed that the efficacy of bacterial strain 751 was also reflected in reduced HDM-specific IgGl levels in the BAL fluid and sérum (see Figures 47 and 49). Firm conclusions regarding an effect on IgG2a levels cannot be drawn. Overall, the data from the antibody analysis is suggestive of a réduction related to an overall reduced inflammatory response, as opposed to a sélective effect on antibody isotype switchîng.
Histologîcal analysis supported the differential cell counts from the BAL fluid, showing a reduced cellular infiltrate in mice treated with Strain 751 (see Figures 51-53).
In relation to cytokine levels, as for Example 4, the spread of data and the error bars for each treatment are large, and the positive and négative Controls do not appear to hâve behaved as necessarily would be expected. It is also possible that the mechanism of action involves influencing earlier cytokine responses that were no longer détectable on day 4 post the final HDM airway challenge. Some care should be taken when interpreting the cytokine data in the current study, due to the variability in the levels detected. This variability could in part be explained by the fact that the lung tissue was separated for the different analyses, and thus one lung lobe might not hâve been fully représentative or comparable to the same lobe in other mice due to patchy distribution of the inflammation. Despite this variability, a clear anti-inflammatory effect on cytokine levels for strain 751 was shown, and the positive control anti-IL-17 Ab generally behaved as expected.
With the above caveats, the data in Figures 56, 58, 59, 61 and 63 suggest that treatment with the bacterial strains ofthe invention, and in particular strain 751 may achieve a réduction in the levels of IL-lb, IFNg, RANTES, ΜΙΡ-la and KC (the mouse orthologue of human IL-8), which may be indicative of a mechanism of action related to influences on chemokine release (and thus recruîtment of cells) by stromal or înnate immune cells. These cytokines are part of the Thl7 pathway. Taking this dataset together, a clear conclusion can be drawn that Strain 751 was highly effective at protecting mice against inflammation in this mouse model of severe neutrophilie asthma.
Example 6 - Efficacy of bacterial inocula in a mouse model of multiple sclerosis
Summarv
Mice were administered with compositions comprising bacterial strains according to the invention and the mice were subsequently immunised with myelin oligodendrocyte glycoprotein to induce experimental autoimmune encephalomyelitis (EAE). EAE is the most commonly used experimental model for human multiple sclerosis. The compositions of the invention were found to hâve a striking effect on disease incidence and disease severity.
Strain
751 : bacterium deposited under accession number NCIMB 42380
Studv design
Groups:
l. Négative control group. Treatment with vehicle control (per oral).
4. Treatment with therapeutic bacteria inoculum strain 751 (pérorai).
9. Positive control group. Treatment with Dexamethasone (i.p.).
10. Untreated Control Group.
Number of mice per group = 10
Days -14 to day 27: Daily administration of vehicle control per oral (Group l ).
Days -14 to day 27: Daily administration of therapeutic bacteria inoculum per oral (Group 4).
Days 0-28: administration of Dexamethasone (i.p.) three times a week (Group 9)
Day 0: MOG35-55 (myelin oligodendrocyte glycoprotein - 2mg/ml) and CFA (2mg/ml MTB) were mixed l:l resulting in Img/ml solutions. ΙΟΟμΙ of the peptide-CFA mixture was injected subcutaneously into each hind leg. Administration of peilussis toxin intraperitoneally (300ng).
Day I: Administration ofpertussis toxîn intraperitoneally (300ng).
Days 7-onwards: Measurement of disease incidence and weight three times a week.
Endpoints and analysis
Mice were analysed for disease incidence and disease severity three times a week. Scorîng was performed blind. Disease severity was assessed using a clinical score ranging from 0 to 5, with 5 indicating a dead mouse (see clinical scoring system below).
Monitoring
On the indicated days mice were weighed and observed for disease activity score and disease incidence.
Disease activity score observations:
- No obvious changes in motor function compared to non-immunized mice.
0.5 - Tip of tail is limp.
l.O- Limp tail.
1.5 - Limp tail and hind leg inhibition.
2.0 - Limp tail and weakness of hind legs.
OR - There are obvious signs of head tilting when the walk is observed. The balance is poor.
2.5 - Limp tail and dragging of hind legs.
OR - There is a strong head tilt that causes the mouse to occasionally fail over.
3.0 - Limp tail and complété paralysis of hind legs.
3.5 - Limp tail and complété paralysis of hind legs.
In addition to: Mouse is moving around the cage, but when placed on its side, is unable to right itself.
Hind legs are together on one side of body.
4.0 - Limp tail, complété hind leg and partial front leg paralysis.
Mouse is minimally moving around the cage but appears alert and feeding
4.5 - Complété hind and partial front leg paralysis, no movement around the cage.
Mouse is îmmediately euthanized and removed from cage.
5.0 Mouse is euthanized due to severe paralysis.
When an animal has equal or greater disease activity score of l, it is considered to hâve a positive disease incidence score.
Results
The results of the study are shown in Figures 71 and 72.
Disease induction in the négative control groups was successful with high scores shown by the vehicle control and the untreated control. The effect of treatment with strain 751 was striking and the mice treated with strain 751 exhibited notably reduced disease incidence and disease severity. Indeed, the réduction in disease incidence and disease severity was comparable to the positive control group. These data indicate the strain 751 may be useful for treating or preventing multiple sclerosis.
Example 7 — Effîcacy of bacterial inocula in mouse models of cancer
Summarv
This study tested the effîcacy of compositions comprising bacterial strains according to the invention in four tumor models.
Materials
Test substance - Bacterial strain &MRX004 (strain 751).
Reference substance - Anti-CTLA-4 antibody (clone: 9H10, catalog: BE0131, isotype: Syrian Hamster IgGl, Bioxcell).
Test and reference substances vehicles - Bacterial culture medium (Yeast extract, Casitone, Fatty Acid medium (YCFA)). Each day of injection to mice, antibody was diluted with PBS (ref: BE145I6F, Lonza, France).
Treatment doses - Bacteria: 2xl08 in 200 μι. The a-CTLA-4 was injected at 10 mg/kg/inj. AntiCTLA-4 was administered at a dose volume of 10 mL/kg/adm (i.e. for one mouse weighing 20 g, 200 μι. of test substance will be administered) according to the most recent body weight of mice.
Routes of administration - Bacterial inoculum was administered by oral gavage (per os, PO) via a cannula. Cannulas were decontaminated every day. Anti-CTLA-4 was injected into the peritoneal cavity ofmice (Intraperitoneally, IP).
Culture conditions of bacterial strain - The culture conditions for the bacterial strain were as follows:
• Pipette I0 mL of YCFA (from the prepared 10 mL E&O lab bottles) into Hungate tubes • Seal the tubes and flush with CO; using a syringe input and exhaust system • Autoclave the Hungate tubes • When cooled, inoculate the Hungate tubes with l mL of the glycerol stocks • Place the tubes în a static 37°C incubator for about 16 hours.
• The following day, take l mL of this subculture and inoculate 10 mL of YCFA (pre-warmed flushed Hungate tubes again, ail in duplîcate) • Place them in a static 37°C incubator for 5 to 6h
Cancer cell line and culture conditions The cell lines that were used are detailed in the table below:
Cell line Type Mouse strain Origin
EMT-6 Breast carcinoma BALB/c ATCC
LL/2 (LLCl) Lung carcinoma C57BL/6 ATCC CRL1642
Hepa l -6 Hepatocellular carcinoma C57BL/6 1PSEN INNOVATION
The EMT-6 cell line was established from a transplantable murine mammary carcinoma that arose in a BALB/cCRGL mouse after implantation of a hyperplastic mammary alveolar nodule [73].
The LL/2 (LLCl) cell line was established from the lung of a C57BL mouse bearing a tumor resulting from an implantation of primary Lewis lung carcinoma [74],
The Hepa l-6 cell line is a dérivative of the BW7756 mouse hepatoma that arose in a C57/L mouse [75].
Cell culture conditions - Ail cell lines were grown as monolayer at 37°C in a humidified atmosphère (5% CCF, 95% air), The culture medium and supplément are indicated in the table below;
Cell line Culture medium Supplément
EMT6 RPMI 1640 containing 2mM L-glutamine (ref: BE12-702F, Lonza) 10% fêta! bovine sérum (ref: #3302, Lonza)
LL/2 (LLCl) RPMI 1640 containing 2mM L-glutamine (ref: BE12-702F, Lonza) 10% fêtai bovine sérum (ref: #3302, Lonza)
Hepal-6 DMEM (ref: 11960-044, Gibco) 10% fêtai bovine sérum (ref: #3302, Lonza) 2mM L-Glutamine penicillin-streptomycin (Sigma G-6784)
For experimental use, adhèrent tumor cells were detached from the culture flask by a 5 minute treatment with trypsin-versene (ref: BE17-I61E, Lonza), in Hanks' medium without calcium or magnésium (ref; BEI0-543F, Lonza) and neutralized by addition of complété culture medium. The cells were counted in a hemocytometer and their viability will be assessed by 0.25% trypan blue exclusion assay.
Ose of animais Healthy female Balb/C (BALB/cByJ) mice, of matching weight and âge, were obtained from CHARLES RIVER (L'Arbresles) for the EMT6 model experiments.
Healthy female C57BL/6 (C57BL16J) mice, of matching weight and âge, were obtained from CHARLES RIVER (L'Arbresles) for the LL/2(LLCl) and the Hepal-6 model experiments.
Animais were maintained in SPF health status according to the FELASA guidelines, and animal housing and experimental procedures according to the French and European Régulations and NRC Guide for the Care and Use of Laboratory Animais were followed [76,77]. Animais were maintained in housing rooms under controlled environmental conditions: Température: 22 ± 2°C, Humidity 55 ± 10%, Photoperiod ( I2h Iight/I2h dark), HEPA filtered air, 15 air exchanges per hour with no recirculation. Animal enclosures were provided with stérile and adéquate space with bedding material, food and water, environmental and social enrichment (group housing) as described: 900 cm2 cages (ref: green, Tecniplast) in ventilated racks, Epicéa bedding (SAFE),l0 kGy Irradiated diet (A04-10, SAFE), Complété food for immuno-competent rodents - R/M-H Extrudate, water from water bottles.
Experimental design and treatments
Antitumor activity, EMT6 niodel
Treatment schedule - The start of first dosing was considered as DO. On DO, non-engrafted mice were randomized according to their individual body weight into groups of 9/8 using Vivo manager® software (Biosystemes, Coutemon, France). On DO, the mice received vehicle (culture medium) or bacterial strain. On Dl4, ail mice were engrafted with EMT-6 tumor cells as described below. On D24, mice from the positive control group received anti-CTLA-4 antibody treatments.
The treatment schedule is summarized in the table below:
Group No. Animais Treatment Dose Route Treatment Schedule
l 8 Untreated - -
2 8 Vehicle (media) - PO QlDx42
3 9 Bacterial strain #l (MRX004) 2x108 bacteria PO QIDx42
4 8 Anti-CTLA4 10 mg/kg IP TWx2
The monitoring of animais was performed as described below.
Induction of EMT6 tumors in animais - On Dl4, tumors were induced by subcutaneous injection of IxlO6 EMT-6 cells in 200 pL RPMl 1640 into the right flank of mice.
Euthanasia - Each mouse was euthanized when it reached a humane endpoint as described below, or after a maximum of 6 weeks post start of dosing.
Antitumor activity-, LL/2 (LLC1) model
Treatment schedule - The start of first dosing was considered as DO. On DO, non-engrafted mice were randomized according to their individual body weight into 7 groups of 9/8 using Vivo manager® software (Biosystemes, Coutemon, France). On DO, the mice will received vehicle (culture medium) or bacterial strain. On Dl4, ail mice were engrafted with LL/2 tumor cells as described below. On D27, mice from the positive control group received anti-CTLA-4 antibody treatments.
The treatment schedule is summarized in the table below:
Group No. Animais Treatment Dose Route Treatment Schedule
l 8 Untreated - - -
2 9 Vehicle (media) - PO QlDx42
3 9 Bacterial strain #l (MRX004) 2xlOs bacteria PO QlDx42
4 8 Anti-CTLA4 10 mg/kg IP TWx2
The monitoring of animais was performed as described below.
Induction of LL/2 (LLCl) tumors in animais - On Dl 4, tumors were induced by subcutaneous injection ofIxlO6 LL/2 (LLCl) cells in 200 pL R.PMI 1640 into the right flank of mice.
Euthanasia - Each mouse was euthanized when it reached a humane endpoint as described below, or after a maximum of 6 weeks post start of dosing.
Antitumor activity, Hepal-6 model
Treatment schedule - The start of first dosing was considered as DO. On DO, non-engrafted mice were randomized according to their individual body weight into 7 groups of 9 using Vivo manager® 10 software (Biosystemes, Coutemon, France). On DO, the mice received vehicle (culture medium) or bacterial strain. On Dl4, ail mice were engrafted with Hepa l-6 tumor cells as described below. On Dl6, mice from the positive control group received anti-CTLA-4 antibody treatments.
The treatment schedule is summarized in the table below:
Group No. Animais Treatment Dose Route Treatment Schedule
l 9 Lfntreated - - -
2 9 Vehicle (media) - PO QlDx42
4 9 Bacterial strain #2 (MRX004) 2xl O8 bacteria PO QlDx42
7 9 Anti-CTLA4 10 mg/kg IP TWx2
The monitoring of animais was performed as described below.
Orthotopic induction of Hepa l-6 tumor cells in animais by intrasplenîc injection - On Dl4, one million (IxlO6) Hepa l-6 tumor cells in 50 pL RPMI 1640 medium were transplanted via intrasplenic injection into mice. Briefly, a small left subcostal flank incision was made and the spleen was exteriorized. The spleen was exposed on a stérile gauze pad, and injected under visual control with the cell suspension with a 27-gauge needle. After the cell inoculation, the spleen was excised.
Euthanasia - Each mouse was euthanized when it reached a humane endpoint as described in section below, or after a maximum of 6 weeks post start of dosing.
Evaluation of tumor burden at euthanasia - At the time of termination, livers were collected and weighed.
Animal monitoring
Clinical monitoring - The length and width of the tumor was measured twice a week with callipers and the volume of the tumor was estimated by this formula [78]:
_ . width2 x lensth
Tumor volume =----------—
Humane endpoints [79]: Signs of pain, suffering or distress: pain posture, pain face mask, behaviour; Tumor exceeding 10% of normal body weight, but non-exceeding 2000 mm3; Tumors interfering with ambulation or nutrition; Ulcerated tumor or tissue érosion; 20% body weight loss remaining for 3 consecutive days; Poor body condition, émaciation, cachexia, déhydration; Prolonged absence of voluntary responses to external stimuli; Rapid laboured breathing, anaemia, significant bleeding; Neurologie signs: circling, convulsion, paralysis; Sustained decrease in body température; Abdominal distension.
Anaesthesia - Isoflurane gas anesthésia were used for ail procedures: surgery or tumor inoculation, i.v. injections, blood collection. Ketamine and Xylazine anesthésia were used for stereotaxia surgîcal procedure.
Analgesîa - Carprofen or multimodal carprofen/buprenorphine analgesia protocol were adapted to the severity of surgical procedure. Non-phaimacological care was provided for ail painful procedures. Additionally, pharmacological care not interfering with studîes (topic treatment) were provided at the recommendation of the attending veterinarian.
Euthanasia - Euthanasia of animais was performed by gas anesthésia over-dosage (Isoflurane) followed by cervical dislocation or exsanguination.
Results
Antitumor activity, EMT6 model
The results are shown in Figure 73. Treatment with the bacterial strain of the invention led to a clear réduction in tumour volume relative to both the négative Controls. The positive control also led to a réduction in tumour volume, as would be expected.
Antitumor activity, LL/2 (LLC1) mode!
The results are shown in Figure 74. The négative and positive Controls do not appear as would be expected, because tumour volume was greater in the mice treated with the positive control than in the négative control groups. Nevertheless, tumour volume in the mice treated with the bacterial straîn of the invention was comparable to the positive control group, which is consistent with a useful therapeutic effect.
Antitumor activity, Hepal-6 mode!
The results are shown in Figure 75. The untreated négative control does not appear as would be expected. because liver weight was lower in this group than the other groups. However, the vehicle négative control and the positive control groups both appear as would be expected, because mice treated with vehicle alone had larger livers than mice treated with anti-CTLA4 antibodies, reflecting a greater tumour burden in the vehicle négative control group. Treatment with the bacterial strain of the invention led to a clear réduction in liver weight (and therefore tumour burden) relative to the mice in the vehicle négative control group.
These data indicate that strain 751 ! MRX004 may be useful for treating or preventing cancer, and in particular for reducing tumour volume in breast, lung and liver cancers.
Exemple 8 — Attachaient to human cells in YCFA medium
Sumniary
The level of binding of strain 751 and a number of Bifidobacterium hreve strains to human cells was determined at 3 distinct time points in YCFA medium. The bacteria attached to the human cells were resuspended in medium and the optîcal density of the medium was then analysed - the liigher the optical density, the higher the number of bacterial cells and thus, the higher the level of binding of the bacterial cells to human cells. The 751 strain was found to display reduced attachment to human cells compared to the Bifidobacterium hreve reference strains.
Results and analysis
The results of the experiment are shown in Figure 76.
As shown in Figure 76, the Bifidobacterium hreve strains show a high level of attachment to human cells at ail time points. On the other hand, the 751 strain has a drastically reduced level of attachment to human cells. Therefore, the low adhérence to human cells of strain 751 may increase the bénéficiai effect of the compositions of the invention on the IL-17 or the Thl7 pathway and on dîseases mediated by IL-l 7 or the Thl 7 pathway.
Example 9 — Assay detecting the production of exopolysaccharides
Summarv
The level of exopolysaccharide (EPS) production by the bacterial strain of the invention (751) and a number of Bifidobacterium brève strains was analysed at 37°C for 48 hours and at 30°C for 72 hours. EPSs are polysaccharides produced by certain bacteria which bind to the outside surface of the bacterial cell. The level of EPSs on the surface of bacteria can be determined using a Congo Red assay which binds to the polysaccharides. A higher intensity of Congo Red absorbance indicates a higher concentration of EPSs on the surface of the bacteria. The bacterial strain of the invention was found to produce and bind more EPSs than the Bifidobacterium breve strains.
Résulte and analysis
The results of this experiment are shown in Figure 77.
As shown in Figure 77, the bacterial strain of the invention showed a greater Congo Red absorbance than the Bifidobacterium breve strains at both températures and time-points. Therefore, the strain of the invention displays greater EPS production and a greater level of extracellular bound EPSs. As the EPSs enable bacteria to bind to mucus and épithélial cells, the bacterial strain of the invention may be useful for competing with pathogenic cells for binding sites on épithélial cells and within mucus membranes. Thus, the bacterial strain of the invention may be useful for modulating the microbiome and treating a number of diseases associated with the microbiome.
Example 10 — Stability testing
A composition described herein containing at least one bacterial strain described herein is stored in a sealed container at 25 C or 4 C and the container is placed in an atmosphère having 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90% or 95% relative humidity. After 1 month, 2 months, 3 months, 6 months, 1 year, 1.5 years, 2 years, 2.5 years or 3 years, at least 50%, 60%, 70%, 80% or 90% of the bacterial strain shall remain as measured in colony forming units determined by standard protocols.
Example 11 — MRX004 bound and released exopolysaccharide production assay
For EPS extraction, MRX004 was cultured in 10 ml YCFA until it reached late exponential phase, when bacterial cells and supematants were separated by centrifugation. Cells were washed once with PBS to remove any remaining culture medium. Secreted or released EPS (EPS-R) was precipitated from culture supematants by treatment with ice-cold 100 % éthanol (ovemight at 4°C with gentle agitation). To extract capsular or bound EPS (EPS-B), cells were incubated with 0.05 M EDTA (ovemight at 4 °C with gentle agitation), and supematants from this treatment were collected and subsequently treated with 100 % ice-cold éthanol (ovemight at 4 °C with gentle agitation) to precipitate EPS-B. Precipitated EPS-B and EPS-R were pelleted by centrifugation and were allowed to dry briefly in a laminar hood before they were resuspended in enough stérile Ultrapure water to achieve a uniform solution. To further purify samples, they were dialysed against stérile Ultrapure water at a ratio of 1:100 for 48 hours with 3 buffer changes. EPS-B and EPS-R were quantified using the phenol-sulphuric acid method using glucose as standard. Using this assay, MRX004 was found to produce a greater amount of EPS-R (115 pg) than EPS-B ( 17 pg) (Figure 78).
Example 12 - Attachaient assay of MRX004 to Caco-2 cells
Attachment of MRX004 to host cells was analysed using an in vitro co-culture assay with Caco-2 intestinal épithélial cells. Caco-2 cells were seeded at a density of 1 x 10 Bacteria were cultured in 10 ml YCFA until they reached late exponential phase, when they were pelleted, washed twice with PBS, and resuspended in antibiotic-free cell culture medium. Bacterial density was adjusted to obtain an approximate multiplicity of infection (MOI) of 10:1 (which was confirmed by plating on YCFA agar using the WAS? standard protocol), and MRX004 was co-incubated with Caco-2 cells in anaérobie conditions at 37°C for 2 hours. Medium was subsequently removed, and unbound bacteria were removed by washing Caco-2 cells three times with PBS. Bacteria-bound Caco-2 cells were lysed and removed from the vessel using treatment with 0.1 % Triton X-I00, and 50 μΐ volumes of diluted lysate were plated in YCFA agar using the WASP. Attachment was calculated by counting the numbers of bacteria recovered from lysate and expressing this as a percentage of total bacteria. MRX004 was found to display low-level adhérence (0.3 % of total culture) to Caco-2 cells (Figure 79).
Example 13 - Characterisation of enzymatic activity
The Analytical Profile Index (API®) test system consists of strips that contain miniaturised biochemical tests that assay for enzymatic activity in bacterial species. MRX004 (strain 751, the bacterium deposited under accession number NCIMB 42380) was characterised using two API test Systems: Rapid ID 32A - This system is designed specîfically for anaérobie species and encompasses tests for carbohydrate, amino acid and nitrate metabolism as well as alkaline phosphatase activity; and API® 50 CH - This system tests for the fermentation of 49 carbohydrate sources, and can be utilised in conjunction with API® CHL Medium for analysis of anaérobie species.
Rapid ID 32A testing was carried out on bacterial colonies as per manufacturer’s instructions. Briefly, bacteria were cultured on YCFA agar for 24 hours at 37 °C in an anaérobie workstation. Colonies were removed from plates using a stérile 5 μΐ inoculating loop and resuspended in a 2 ml ampoule of API® Suspension Medium until a density roughly équivalent to that of McFarland standard No. 4 was achieved. Fifty-five microlitres of bacterial suspension was added to each cupule on a Rapid ID 32A strip, and the urease test was overlayed with two drops of minerai oil. Strips were covered with a plastic lid and incubated aerobically at 37 °C for 4 hours, following which the bottom row of cupules were developed using the following reagents: NIT: 1 drop each of NIT1 and NIT2; IND: 1 drop of James reagent; ail remaining cupules: 1 drop of FastBlue reagent. Strips were incubated at room température for 5 minutes, following which the colour of each cupule was recorded and assigned a value of négative, intermedîate positive or positive.
The results of the Rapid ID 32A analysis are shown in Figure 80. MRX004 tested positive for fermentation of several carbohydrate sources, namely α-galactosidase and β-galactosidase, aglucosidase and β-glucosîdase, α-arabtnose, mannose and raffinose, as well as the amino acids arginine, proline, phenylalanine, leucine, tyrosine, glycine and histidine. Interestingly, roles hâve 5 been reported for some of these amino acids in asthma. For instance, increased plasma concentrations of phenylalanine and histidine hâve been reported to be associated with adverse effects in asthma, including increased inflammation, histamine production and airway hyper-responsiveness. In addition, arginine metabolism is implicated in asthma pathogenesis, as increased levels of the arginine métabolite L-ornithine hâve been reported in paediatric patients, and administration of 10 arginine attenuated inflammation in an in vivo asthma model. Based on these reports, it is possible that amino acid metabolism by MRX004 may be involved in the anti-asthma effects of this strain.
Comparative Rapid ID 32A analysis was carried out between MRX004 and four B. breve type strains, which are annotated in Figure 80B as Bif Ref l (DSM 20091), Bif Ref 2 (DSM 20213), Bif Ref 6 (JCM 7017) and Bif Ref 7 (UCC2003). This analysis demonstrated that MRX004 was the only 15 strain tested to ferment the polysaccharide raffinose, which may be significant, because raffinose is involved in the production of bacterial components such as exopolysaccharides, and raffinose fermentation can also reportedly confer effects on the host such as increased caecal butyrate, increased gastrointestinal prolifération and weight loss.
API® 50 CH testîng was carried out to further examine carbohydrate metabolism in MRX004. As 20 per manufacturées instructions, bacteria were cultured in 10 ml YCFA broth for 16-18 hours at 37°C in an anaérobie workstatîon. This culture was diluted in 10 ml API® CHL Medium so as to achieve a density roughly équivalent to McFarland standard No. 2, and HO pl of this mixture was used to inoculate each cupule on a set of API® 50 CH test strips. Test strips were incubated în a humidified incubation box at 37 °C in an anaérobie workstatîon for 48 hours, following which the colour of each 25 cupule was recorded and assigned a value of négative, intermediate positive, positive or doubtful.
Using API® 50, MRX004 tested positive for utilisation of the following carbohydrate sources: amidon (starch), amygdalin, arbutin, cellobiose, esculin, galactose, gentiobiose, glucose, glycogen, fructose, fucose, lactose, makose, mannose, mannitol, melibiose, melezitose, methyl a-Dglucopyranoside, N-acetylglucosamine, ribose, saccharose (sucrose), salicin, sorbitol, trehalose, 30 turanose and xylitol. These results correlated with those obtained for Rapid ID 32A testing in that MRX004 demonstrated fermentation of galactose, glucose, mannose and raffinose in both test Systems. Interestingly, some MRX004 carbohydrate substrates, namely galactose and fructose, may be implicated in the mechanisin of action of this strain, based on their reported effects in the literature. Galactose a-l,3- galactose derived from méat sources is a known allergen and causative agent of anaphylaxis, and intake levels of dietary fructose are correlated with increased asthma severity. Taken together both sets of API® data for MRX004 suggest that the metabolism of this strain may play a rôle in its anti-asthma effects.
Exampie 14 — Genome analysis
A comparison ofthe genome content of strain MRX004 and the reference strains of B. breve, 1, 2, 6 and 7 was carried out using blastn as part of the BLAST+ 2.3.0 suite of programs. A maximum Evalue cut-off score of 10E-5 was employed throughout the analysis.
333 genes were identified (Table 1) that are present in the genome of strain MRX004 but are absent from the B. breve reference strains 1 (DSM 20091), 2 (DSM 20213), 6 (JCM 7017) and 7 (UCC2003). Many of the genes iisted in Table 1 are frequently observed as being hypervariable among B. breve strains [80], As expected, the régions of variability include genes that code for proteins involved in carbohydrate metabolism and transport, phage-associated genes, mobile éléments, as well as 173 genes predicted to encode proteins or genes of unknown fonction.
Genes that are present in MRX004 but absent from B. breve reference strains 1, 2, 6 and 7 are Iisted in Table 1. Genes that are not highlighted are absent in more than one of the four reference strains. The large number of genes that are present in MRX004 but are not present in numerous B. breve reference strains suggests that MRX004 is distinct from and/or distinguishable from these known B. breve strains. Genes highlighted with single underlining are present in MRX004 but absent in B. breve reference strain 1. Genes highlighted with double underlining and in bold are present in MRX004 but absent in B. breve reference strain 2. Genes highlighted with italics are present in MRX004 but absent in B. breve reference strain 6. A maximum E-value cut-off score of 10E-5 was employed for the blastn analysis.
Table 1
4DBb 0021c Multiple sugar ABC transporter, substrate-binding protein
4DBb 0023 Probable Lad-type transcrîptional regulator
4DBb 0024 Sucrose-6-phosphate hydrolase (EC 3.2.1 .B3)
4P B b 0026c Maltodextrin elucosidase ÎEC 3.2.1.20)____________________
4DBb 0036c Hypothetical protein
4DBb 0038c MSM (multiple sugar metabolism) operon regulatory protein
4DBb 0119c Hypothetical protein
4DBb 0120c Hypothetical protein
4DBb 0187 Hypothetical protein
4DBb 0188 Hypothetical protein
4DBb 0203c Cell division protein FtsL
4DBb 0204c Hypothetical protein
4DBb 0205c Hypothetical protein
4DBb 0206c Transcriptional regulator, HxlR family
4DBb 0207 Rrf2-linked NADH-flavin reductase
4DBb 0208 ATP-dependent DNA helicase RecG-related protein
4DBb 0209c Hypothetical protein
4DBb 0210 Putative transporter
4DBb 0211 Omega-3 polyunsaturatedfatty acid synthase subuni f, PfaA
4DBb_0212 Type I polyketide synthase
4DBb 0213c Hypothetical protein
4DBb 0214c Hypothetical protein
4DBb 0215 Hypothetical protein
4DBb 0216c Conserved hypothetical protein
4DBb 0218c Hypothetical protein
4DBb 0219c DNA-cytosine methyltransferase
4DBb 0220c Hypothetical protein
4DBb 0221c Hypothetical protein
4DBb 0222c Hypothetical protein
4DBb 0223c Integrase
4DBb 0256 Hypothetical protein
4DBb 0257c Lacl-type transcriptional regulator
4DBb 0258 Putative glycosyl hydrolase of unknown function (DUF1680)
4DBb 0284 Transcriptional regulator, AraC family
4DBb 0285 N-Acetyl-D-glucosamine ABC transport system, sugarbinding protein
4DBb 0286 Sugar ABC transporter permease
4DBb 0287 N-Acetyl-D-glucosamine ABC transport system, permease protein 2
4DBb 0288 Alpha-galactosidase (EC 3.2.1.22)
4DBb 0329c A TPase component BioM of energizing module of biotin ECF transporter
4DBb 0330 Major facilitator superfamily MFS 1
4DBb 0368 GMP synthase [glutamine-hydrolyzing] (EC 6.3.5.2)
4DBb 0369c Mu-like prophage protein gp29
4DBb 0410 Putative galactosidase
4DBb 0419 Hypothetical protein
4DBb 0421 Glycosyltransferase SypP
4DBb 0422 Capsular polysaccharide biosynthesis protein
4DBb 0423 Hypothetical protein
4DBb 0424 Glycosyltransferase
4DBb 0425 Membrane protein involved in the export of O-antigen, teichoic acid lipoteichoic acids
4DBb 0426 Glycosyltransferase (EC 2.4.1.-)
4DBb 0427 2-succinyl-5-enoIpyruvyl-6-hydroxy-3- cyclohexene-1 carboxylic-acid synthase (EC 2.2.1.9)
4DBb 0428 Hypothetical protein
4DBb 0429 Mobile element protein
4DBb 0430 Hypothetical protein
4DBb 043le Hypothetical protein
4DBb 0432c Mobile element protein
4DBb 0433c Hypothetical protein
4DBb 0434c Hypothetical protein
4DBb 0435 Hypothetical protein
4DBb 0436 Hypothetical protein
4DBb 0437c Hypothetical protein
4DBb 0438c Mobile element protein
4DBb 0439c Hypothetical protein
4DBb 0440c Mobile element protein
4DBb 0518c PIN domain protein
4DBb 0519c Hypothetical protein
4DBb 0555c Hypothetical protein
4DBb 0556c Hypothetical protein
4DBb 0557c LSU ribosomal protein L31p @ LSU ribosomal protein L3Ip, zinc-independent
4DBb 0558c SSU ribosomal protein S14p (S29e) @ SSU ribosomal protein S14p (S29e), zinc-independent
4DBb 0559c LSU ribosomal protein L33p @ LSU ribosomal protein L33p, zinc-independent
4DBb 0560c Hypothetical protein
4DBb 0561 Hypothetical protein
4DBb 0613c Cellulose svnthase (UDP-formins) IEC 2.4.1.12)
4DBb 0614 Chitinase ŒC 3.2.1.14)
4DBb 0615 Sensory box/GGDEF familv nrotein
4DBb 0660 Mobile element protein
4DBb 0662 Mobile element protein
4DBb 0663c Neuraminidase NanP
4DBb 0664 Hypothetical protein
4DBb 0665 Hypothetical protein
4DBb_0666 Mobile element protein
4DBb 0667 Mobile element protein
4DBb 0668 Mobile element protein
4DBb 0718 Predicted biotin regulatory protein BioR (GntR family)
4DBb 0719 Hypothetical protein
4DBb 0720 Hypothetical protein
4DBb 0778 Hypothetical protein
4DBb 0789c Mobile element protein
4DBb 0790c Mobile element protein
4DBb 0837c Possible conserved intégral membrane protein.
4DBb 0840 Macroltde-efflux protein
4DBb 0866 Cation-transportingATPase. E1-E2family
4DBb 0867 Hypothetical protein
4DBb 0872 Hypothetical protein
4DBb 0879c Transcriptional regulator, Cro/CI family
4DBb 0880c Intégral membrane protein
I 4DBb 0946 MFS general substrate transporter
4DBb 0947c Putative membrane protein
4DBb 0948c Mobile element protein
4DBb 0952c Hypothetical protein
4DBb 0953 Hypothetical protein
4DBb 0954c Hypothetical protein
4DBb 0955c Hypothetical protein
4DBb 0956c Predicted permeases
4DBb 0957 Narrowly conserved hypothetical protein
4DBb 0958c Conserved hypothetical protein
4DBb 0986ç Hypothetical protein
4DBb 0987ç Duplicated ATPase component BL0693 of enersizing module
of predicted ECF transporter
4DBb 0988ç Transmembrane component BL0694 of energizing module of
predicted ECF transporter
4DBb 1009c Phage holin
4DBb 1010c Membrane-bound lytic murein transglycosylase D precursor (EC 3.2.1.-)
4DBb lOllc Hypothetical protein
4DBb_10l2 Hypothetical protein
4DBb 1013c Hypothetical protein
4DBb 1014c Hypothetical protein
4DBb ΙΟΙ5c Hypothetical protein
4DBb 1016c Hypothetical protein
4DBb 1017c Phage tail protein
4DBb 1018c Phage tail length tape-measure protein
4DBb 1019c Hypothetical protein
4DBb 1020c Hypothetical protein
4DBb 1021c Hypothetical protein
4DBb 1022c Hypothetical protein
4DBb 1023c hypothetical protein
4DBb 1024c Hypothetical protein
4DBb 1025c Hypothetical protein
4DBb 1026c Hypothetical protein
4DBb 1027c Hypothetical protein
4DBb 1028c Hypothetical protein
4DBb 1029c Hypothetical protein
4DBb 1030c Phage protein
4DBb 1031c Phage terminase, large subunit # Pham2
4DBb 1032c Phage terminase, large subunit
4DBb 1033c hypothetical protein
4DBb 1034 hypothetical protein
4DBb 1035 hypothetical protein
4DBb 1036c FIG00424913: hypothetical protein
4DBb 1037c hypothetical protein
4DBb 1038c hypothetical protein___________
4DBb_lO39c hypothetical protein
4DBb 1040c hypothetical protein
4DBb l04lc hypothetical protein
4DBb 1042c hypothetical protein
4DBb 1043c hypothetical protein
4DBb 1044c hypothetical protein
4DBb 1045c hypothetical protein
4DBb 1046c hypothetical protein
4DBb 1047c Chromosome (plasmid) partitioning protein ParB
4DBb 1048c hypothetical protein
4DBb 1049c hypothetical protein
4DBb 1050c Single-stranded DNA-binding protein
4DBb_1051c hypothetical protein
4DBb 1052c hypothetical protein
4DBb 1053c hypothetical protein
4DBb 1054c hypothetical protein
4DBb 1055c hypothetical protein
4DBb 1056 hypothetical protein
4DBb 1057c hypothetical protein
4DBb 1058c hypothetical protein
4DBb 1059 hypothetical protein
4DBb 1060 hypothetical protein
4DBb 1061 hypothetical protein
4DBb 1062c hypothetical protein
4DBb 1063c hypothetical protein
4DBb 1064 putative phage integrase
4DBb 1113 Permeases of the major facilitator superfamily
4DBb 1142 hypothetical protein
4DBb 1143c hypothetical protein
4DBb 1172 Integrase
4DBb 1173c hypothetical protein
4DBb 1174c Narrowly conserved hypothetical protein
4DBb 1175 hypothetical protein
4DBb 1176c hypothetical protein
4DBb 1177 ABC transporter, ATP-binding protein
4DBb_1178 hypothetical protein
4DBb 1179 hypothetical protein
4DBb 1180 hypothetical protein
4DBb 1181 hypothetical protein
4DBb 1182 two-component system sensor kinase
4DBb 1183 hypothetical protein
4DBb 1203c regulatory protein, Lacl
4DBb 1204c FIG01131316: hypothetical protein
4DBb 1205c transport system permease
4DBb 1206c Predicted rhamnose oligosaccharide ABC transport system,
permease component 2
4DBb 1207c extracellular solute-binding protein, family 1
4DBb 1212c hypothetical protein
4DBb 1213c hypothetical protein
4DBb 1214 Mobile element protein
4DBb 1215 Mobile element protein
4DBb 1219 Hypothetical protein
4DBb 1220c Hypothetical protein,
4DBb 1221c Protein of unknown function DUF262 family
4DBb 1222c Hypothetical protein
4DBb 1223 Hypothetical protein
4DBb_1224 Mobile element protein
4DBb_ 1234c ABC-type sugar transport System, periplasmic component
4DBb 1235 Hypothetical protein
4DBb 1328c Putative phage integrase
4DBb 1329 Hypothetical protein
4DBb 1330 Hypothetical protein
4DBb 1331c Hypothetical protein
4DBb 1332 Hypothetical protein
4DBb 1333 Hypothetical protein
4DBb 1334 Hypothetical protein
4DBb 1335c Hypothetical protein
4DBb 1336c Négative regulator of beta-lactamase expression
4DBb 1337c Hypothetical protein
4DBb 1338c Hypothetical protein
4DBb 1339c Hypothetical protein
4DBb 1340c Hypothetical protein
4DBb 1341c Hypothetical protein
4DBb 1342c Hypothetical protein
4DBb. ,1343c Hypothetical protein
4DBb_I344 Hypothetical protein
4DBb 1345c Phage tail fiber protein
4DBb 1346c Hypothetical protein
4DBb 1347c Phage minor tail protein
4DBb 1348c Hypothetical protein
4DBb 1349c Hypothetical protein
4DBb ,1350c Phage protein
4DBb 1351c Phage protein
4DBb 1352c Phage protein
4DBb 1353c Phage protein
4DBb 1354c Hypothetical protein
4DBb 1355c Hypothetical protein
4DBb 1356c Phage major capsid protein #Fam0025 # Phaml64
4DBb 1357c Putative phage prohead protease
4DBb 1358c Phage portai protein
4DBb 1359c f gp2, terminase
4DBb 1360c Hypothetical protein
4DBb 1361c Hypothetical protein
4DBb 1362c Hypothetical protein
4DBb 1363c Hypothetical protein
4DBb 1364c Hypothetical protein
4DBb_1365c Hypothetical protein
4DBb 1366c Hypothetical protein
4DBb 1367c Hypothetical protein
4DBb 1368c Hypothetical protein
4DBb 1369c Hypothetical protein
4DBb 1370c Hypothetical protein
4DBb 1371c Hypothetical protein
4DBb 1372c Single-stranded DNA-binding protein
4DBb 1373c Hypothetical protein
4DBb 1374c Recombinational DNA repair protein RecT (prophage associated)
4DBb 1375c phage-related protein
4DBb 1376c Hypothetical protein
4DBb 1377c Hypothetical protein
4DBb 1378c Hypothetical protein
4DBb 1379c Hypothetical protein
4DBb 1380c Methyltransferase (EC 2.1.1.-)
4DBb 1381c Hypothetical protein
4DBb 1382c Hypothetical protein
4DBb 1383c Hypothetical protein
4DBb 1384 Hypothetical protein
4DBb 1385 Hypothetical protein
4DBb 1386 Hypothetical protein
4DBb 1387 Hypothetical protein
4DBb 1388 Hypothetical protein
4DBb 1456 Sucrose permease, major facilitator superfamily
4DBb_1486c Esterase/lipase
4DBb 1487c Glucose/mannose:H+ symporter GIcP
4DBb 1488c Two-component response regulator yc’iW
4DBh 1533c Hypothetical protein
4DBb 1534c Hypothetical protein
4DBb 1535c Type I restriction-modification system, restriction subunit R (EC 3.1.21.3)
4DBb 1536c ATP-dependent DNA helîcase recG (EC 3.6.1.-)
4DBb 1537c Type I restriction-modification system, specificity subunit S (EC 3.1.21.3)
4DBb_1538c Type I restriction-modification system, DNAmethyltransferase subunit M (EC 2.1.1.72)
4DBb 1539c Hypothetical protein
4DBb 1540 Type I restriction-modification system, specificity subunit S
4DBb l587
4DBb 1588
4DBb 1620c
Transcriptional regulator, AbrB family
Hypothetical protein __3'-to-5' oligoribonuclease (orn)
Transcriptional activatorMltR
Xylitol dehydrogenase (EC 1,1.1.g)
RibitoLOCylitoI/Arabitoi transporter, MFS superfamily Glyoxalase family protein
4DBb 1769c
4DBb 1770c
4DBb 1771c
Γ4ΡΒό_1773~ i
[jpBb 1774c~l RjbitoFXvlitol/Arabitol transporter, mFs superfamjl?
|4DBb 1775c ί Sorbitol dehydrogenase (EC 1 1 1 14)__________ ' WBb l926c I SX*
4DBb 1928c 4DBb 1929c ; 4DBb 1930c ! 4DBb 1934c ; 4DBb 1935 ί 4DBb~ 1936c
4DBb 1937c 4DBb 1965
4DBb 2010c _Glycosyl transferase, group 2 family protein__
- Teichoïc acid expon ATP-binding protein TagH (EC 3,6.3.40) | JUiamnose-containing polysacharide translocation permease Ί i Hypothetical protein
Possible glycosyltransfcrase
Cell wall surface anchor family protein
J3-alanyl-D-alanine carboxypeptidase (EC 3,4.16.4) Hypothetical protein
4DBb 2011
4DBb 2012
Mobile element protein
Transcriptional regulator, Lad family
Xylose ABC transporter, periplasmic xylose-binding protein transP^ystcm, ATP-bmdm^e^ï^Â W’
4DBb 2013
4DBb 2014 R^ose ABC transport System, pennease protein RbsC (TC
4DBb 2015
Hypothetical protein
4DBb 2016 Mobile element protein
4DBb 2028c Beta-giucosidase (EC 3.2.1.21 )
Sequences
SEQ ID N0:l (consensus 16S rRNA sequence for strain 751)
GGGACAGGCTCAGGATGAACGCCGGCGGCGTGCTTAACACATGCAAGTCGAACGGGATCCATCGGGCTTTGCCT
GGTGGTGAGAGTGGCGAACGGGTGAGTAATGCGTGACCGACCTGCCCCATGCACCGGAATAGCTCCTGGAAACG
GGTGGTAATGCCGGATGCTCCATCACACCGCATGGTGTGTTGGGAAAGCCTTTGCGGCATGGGATGGGGTCGCG
TCCTATCAGCTTGATGGCGGGGTAACGGCCCACCATGGCTTCGACGGGTAGCCGGCCTGAGAGGGCGACCGGCC
ACATTGGGACTGAGATACGGCCCAGACTCCTACGGGAGGCAGCAGTGGGGAATATTGCACAATGGGCGCAAGCC
TGATGCAGCGACGCCGCGTGAGGGATGGAGGCCTTCGGGTTGTAAACCTCTTTTGTTAGGGAGCAAGGCACTTT
GTGTTGAGTGTACCTTTCGAATAAGCACCGGCTAACTACGTGCCAGCAGCCGCGGTAATACGTAGGGTGCAAGC
GTTATCCGGAATTATTGGGCGTAAAGGGCTCGTAGGCGGÏTCGTCGCGTCCGGTGTGAAAGTCCATCGCTTAAC
GGTGGATCCGCGCCGGGTACGGGCGGGCTTGAGTGCGGTAGGGGAGACTGGAATTCCCGGTGTAACGGTGGAAT
GTGTAGATATCGGGAAGAACACCAATGGCGAAGGCAGGTGTGTGGGCCGTTACTGACGCTGAGGAGCGAAAGCG
TGGGGAGCGAACAGGA~TAGATACCG”GGTAGTCCACGCGGTAAACGG~GGATGCTGGATGTGGGGCCCGTTCC
ACGGGT7CCGTGTCGGAGCTAACGGGTTAAGCATCCCGCC7GGGGAGTACGGCCGCAAGGCTAAAACTCAAAGA
AA7TGACGGGGGCCCGCACAAGCGGCGGAGCATGCGGATÏAATTCGATGCAACGCGAAGAACCTTACCTGGGCT
TGACA7G77CCCGACGATCCCAGAGATGGGGTTTCCCTTCGGGGCGGGTTCACAGGTGGTGCATGGTCGTCGTC
AGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAACGAGCGCAACCCTCGCCCCGTGTTGCCAGCGGATTGTG
CCGGGAACTCACGGGGGACCGCCGGGGTTAACTCGGAGGAAGGTGGGGATGACGTCAGATCATCATGCCCCTTA
CGTCCAGGGCTTCACGCATGCTACAATGGCCGGTACAACGGGATGCGACAGCGCGAGCTGGAGCGGATCCCTGA
AAACCGGTCTCAGTTCGGATCGCAGTCTGCAACTCGACTGCGTGAAGGCGGAGTCGCTAGTAATCGCGAATCAG
CAACGTCGCGGTGAATGCGTTCCCGGGCCTTGTACACACCGCCCGTCAAGTCATGAAAGTGGGCAGCACCCGAA GCGGGTGGCCTAACCCCTGCGGGAGGGAGCCKC
SEQ ID N0:2 (strain 751 genome sequence) - see WQ2016203223.
REFERENCES
[l ] Spor et al. (2011) Nat Rev Microbiol. 9(4):279-90.
[2] Eckburg et al. (2005) Science. I0;308(5728): 1635-8.
[3] Macpherson et al. (2001 ) Microbes Infect. 3( 12): 1021-35
[4] Macpherson et al. (2002) Cell Mol Life Sci. 59(12):2088-96.
[5] Mazmanian et al. (2005) Cell 15; 122(1 ):107-18.
[6] Frank et al. (2007) PNAS 104(34):13780-5.
[7] Scanlan et al. (2006) J Clin Microbiol. 44(11 ):3980-8.
[8] K.ang et al. (2010) Inflamm Bowel Dis. 16(12):2034-42.
[9] Machiels et al. (2013) Gut. 63(8):1275-83.
[10] WO 2013 050792
[11] WO 03,046580
[12] WO 2013 008039
[13] WO 2014 167338
[14] Goldm and Gorbach (2008) Clin Infect Dis. 46 Suppl 2:S96-IOO.
[15] Azad et al. (2013) BMJ 347:(6471.
[16] Masco et al. (2003) Systematic and Applied Microbiology. 26:557-563.
[ 17] Srûtkovâ et al. (201 ! ) J Microbiol Methods, 87( 1 ): 10-6.
[18] \'eet al. (2015) PLoS One. 10( l):e0117704.
[19] Fabro et al. (2015) Immunobiology. 220(1):124-35.
[20] Yin et al. (2014) Immunogenetics. 66(3):215-8.
[21] Cheluvappa et al. (2014) Clin Exp Immunol. 175(2):316-22.
[22] Schieck et al. (2014) J Allergy Clin Immunol. 133(3):888-91.
[23] Balato et al. (2014) J Eur Acad Dermatol Penereol. 28(8):1016-24.
[24] Monteleone et al. (2011) BMC Medicine. 2011,9:122.
[25] Fahy (2009) Proc Am Thorac Soc 6.256-259
[26] Miossec and Kolls (2012) Nat Rev Drug Discov. 11 ( 10):763-76.
[27] Yang et al. (2014) Prends Pharmacol Sci. 35(10):493-500.
[28] Koenders et al. (2006) J. Immunol. 176:6262-6269.
[29] Amedei et al. (20]2) Int J Moi Sci. 13(10):13438-60.
[30] Shabgah et al. (2014) Postepy. Dermatol. Alergol. 31(4):256-61.
[31] Zhang (2015) Inflammation. Aug 23.
[32] Sun et al. (2015) Cytokine. 74(1):76-80.
[33] Mucientes et al. (2015) BrJOphthalmol. 99(4):566-70.
[34] Jawad et al. (2013) Ocul Immunol Inflamm. 21(6):434-9.
[35] Maya et al. (2014) J. Ophlhalmology. 310329
[36] Chi et al. (2007) J. Allergy and Clinical Immunology. 119(5):1218-1224.
[37] Chi et al. (2008) Investigative Ophthalmology &Pisual Science. 49(7): 3058-3064
[38] Luger and Caspi (2008) Semin. Immunopathol. 30(2): 134-143.
[39] Numasaki et al. (2003) Blood. 101:2620-2627.
[40] Zhang et al. (2008) Biochem. Biophys. Res. Commun. 374: 533-537.
[4 J ] Karin (2006) Nature. 44 t : 431 -436.
[42] Faghih et al. (2013). Iranian Journal of Immunology. 10(4): 193-204.
[43] Numasaki et al. (2005) J. Immunol. 175: 6177-6189
[44] Hammerich and Tacke (2014) Clin Exp Gastroenterol. 7:297-306.
[45] Haabeth et al. (2012) Oncolmmunology 1(1):1146-1152.
[46] Lejeune et al. (2006) Cancer Immun. 6:6
[47] Pace et al. (1983) PNAS. 80:8782-6.
[48] Sgadari et al. (1996) PNAS. 93:13791-6.
[49] Arenberg et al. ( 1996) J. Exp. Med. 184:981-92.
[50] Sgadari étal. (1997)W>4 89:2635-43.
[51 ] Miyamoto-Shinohara et al. (2008) J. Gen. Appl. Microbiol., 54, 9-24.
[52] Cryopreservation and Freeze-Drying Protocols, ed. by Day and McLelIan, Humana Press.
[53] Leslie et al. (1995) Appl. Environ. Microbiol. 61, 3592-3597.
[54] Milropoulou et al. (20I3) JNutr Metab. (2013) 71686L
[55] Kailasapathy et al. (2002) Curr Issues Intest Microbiol. 3(2):39-48.
[56] Handbook of Pharmaceutical Excipients, 2nd Edition, (1994), Edited by A Wade and P J Weller
[57] Remington's Pharmaceutical Sciences, Mack Publishing Ço. (A. R. Gennaro edit. 1985)
[58] Handbook of Microbiological Media, Fourth Edition (2010) Ronald Atlas, CRC Press.
[59] Maintaining Cultures for Biotechnology and Industry (1996) Jenrtie C. Hunter-Cevera, Academie Press
[60] Strobel (2009) Methods Mol Biol. 581:247-61.
[61 ] Gennaro (2000) Remington: The Science and Practice of Pharmacy 20th édition, ISBN: 0683306472.
[62] Molecular Biology Techniques: .-In Intensive Laboratory Course, (Ream et al., eds., 1998, Academie Press).
[63] Methods In Encymology (S. Colowick and N. Kaplan, eds.. Academie Press, Inc.)
[64] Handbook of Experimental bnmunology. Vols. I-IV (D.M. Weir and C.C. Blackwell. eds. 1986, Blackwell Scientifie Publications)
[65] Sambrook et al (2001 ) Molecular Cloning .4 Laboratory Manual. 3rd édition (Cold Spring Harbor Laboratory Press).
[66] Handbook of Surface and Colloïdal Cheniistry (Birdi. K.S. ed., CRC Press. 1997)
[67] Ausubel et al. (eds) (2002) Short protocols in molecular biology, 5th édition (Current Protocols).
[68] PCR (Introduction to Biotechniques Sériés). 2nd ed. (Newton & Graham eds., 1997, Springer Verlag)
[69] Current Protocols in Molecular Biology (F.M. Ausubel et al., eds., 1987) Supplément 30
[70] Smith & Waterman ( 1981 ) Adv. Appl. Math. 2: 482-489.
[71] Brandein/. (20Q1) Nature Protocols. 2(5):1269-1275
[72] Jiao et al. (2014) Immunopathology and Infections Diseases. 184(4):1085-93.
[73] Rockwell et al., (1972) J Nat! Cancer Inst. 49:735-49.
[74] Bertram and Janik (1980) Cancer Lett. 11:63-73.
[75] Darlington (1987) Meth Enzymol. 151:19-38.
[76] Principe d’éthique de l'expérimentation animale. Directive n°2010/63 CEE 22nd September 2010, Décrêt n°2013-I18 Ist February 2013.
[77] Guide for the Care and Use of Laboratory Animais: Eighth Edition. The National Academies Press; 2011
[78] Simpson-Herren and Lloyd (1970) Cancer Chemother Rep. 54:143-74.
[79] Workman et al. (2010) Br. J. Cancer. 102:1555-77.
£80] Bottacini et a/(2014) BMC Genomics. 15:170,001: 10.1186/1471-2164-15-170, PMID: 24581150

Claims (12)

1. A composition comprising the bacterium deposited under accession number NCIMB 42380 or a biotype thereof, for use in a method of treating or preventing an inflammatory or autoimmune disease, or cancer.
2. The composition of claim l, wherein the composition is for use in a method of treating or preventing a disease or condition selected from the group consisting of asthma, including allergie asthma or neutrophilie asthma; arthritis, including rheumatoid arthritis, osteoarthritis, psoriatic arthritis, or juvénile idiopathic arthritis; multiple sclerosis; neuromyelitis optica (Devic's disease); ankylosing spondylitis; spondyloarthritis; psoriasis; systemic lupus erythematosus; inflammatory bowel disease, including Crohn’s disease or ulcerative colitis; celiac disease; chronic obstructive pulmonary disease (COPD); cancer, including breast cancer, colon cancer, lung cancer or ovarian cancer; uveitis; scleritis; vasculitis; Behcet’s disease; atherosclerosîs; atopie dermatitis; emphysema; periodontitis; allergie rhinitis; and allograft rejection.
3. The composition of claim 2, wherein the composition is for use in in a method of treating or preventing asthma, including neutrophilie asthma or allergie asthma.
4. The composition of claim 3, wherein the composition is for use in a method of reducing neutrophilia or eosinophilia in the treatment of asthma.
5. The composition of claim 2, wherein the composition is for use in in a method of treating or preventing rheumatoid arthritis.
6. The composition of claim 5, wherein the composition is for use in a method of reducing joint swelling in rheumatoid arthritis.
7. The composition of claim 2, wherein the composition is for use in in a method of treating or preventing multiple sclerosis.
8. The composition of claim 7, wherein the composition is for use in a method of reducing disease incidence or disease severity.
9. The composition of claim 2, wherein the composition is for use in a method of treating or preventing cancer, including lung cancer, breast cancer or liver cancer.
10. The composition of claim 9, wherein the composition is for use in a method of reducing tumour size, reducing tumour growth, preventing metastasis or preventing angiogenesis.
11. The composition of claim 2, wherein the composition is for use in a method of treating or preventing uveitis.
12. The composition of claim 11, wherein the composition is for use in a method of reducing or preventing retinal damage in uveitis.
OA1201700487 2015-06-15 2016-06-15 Compositions comprising bacterial strains. OA18777A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1510467.2 2015-06-15
GB1520501.6 2015-11-20

Publications (1)

Publication Number Publication Date
OA18777A true OA18777A (en) 2019-06-28

Family

ID=

Similar Documents

Publication Publication Date Title
US11331352B2 (en) Compositions comprising bacterial strains
US10780134B2 (en) Compositions comprising bacterial strains
US20180344780A1 (en) Compositions comprising bacterial strains
OA18777A (en) Compositions comprising bacterial strains.