COMPOSITIONS COMPRISING BACTERIAL STRAINS

The invention provides compositions comprising bacterial strains for treating and preventing central nervous system disorders and conditions.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE

This application is a continuation of U.S. application Ser. No. 16/692,667, filed Nov. 22, 2019, which is a continuation of International Application No. PCT/GB2018/051389, filed May 22, 2018, which claims the benefit of Great Britain Application No. 1708176.1, filed May 22, 2017, Great Britain Application No. 1714309.0, filed Sep. 6, 2017, Great Britain Application No. 1714298.5, filed Sep. 6, 2017, Great Britain Application No. 1714305.8, filed Sep. 6, 2017, Great Britain Application No. 1716493.0, filed Oct. 9, 2017, and Great Britain Application No. 1718551.3, filed Nov. 9, 2017, all of which are hereby incorporated by reference in their entirety.

SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in ANSI format and is hereby incorporated by reference in its entirety. Said ANSI copy, created on Nov. 17, 2021, is named 56708_723_302_SL and is 4,325,277 bytes in size.

TECHNICAL FIELD

This invention is in the field of compositions comprising bacterial strains isolated 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 sterile in utero, but it is exposed to a large variety of maternal 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 genotype, all of which impact upon the composition of the gut microbiota, particularly during early life. Subsequently, the microbiota stabilizes and becomes adult-like [1]. The human gut microbiota contains more than 500-1000 different phylotypes belonging essentially to two major bacterial divisions, the Bacteroidetes and the Firmicutes [2]. The successful symbiotic relationships arising from bacterial colonization of the human gut have yielded a wide variety of metabolic, structural, protective and other beneficial functions. The enhanced metabolic activities of the colonized gut ensure that otherwise indigestible dietary 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 exemplified in germfree animals which have an impaired immune system that is functionally reconstituted following the introduction of commensal bacteria [3-5].

The discovery of the size and complexity of the human microbiome has resulted in an on-going evaluation of many concepts of health and disease. Certainly, dramatic changes in microbiota composition have been documented in gastrointestinal disorders such as inflammatory bowel disease (IBD)[6-9]. More recently, there is increased interest in the art regarding alternations in the gut microbiome that may play a pathophysiological role in human brain diseases [10]. Preclinical and clinical evidence are strongly suggesting a link between brain development and microbiota [11].

A growing body of preclinical literature has demonstrated bidirectional signalling between the brain and the gut microbiome, involving multiple neurocrine and endocrine signalling systems. Indeed, increased levels of Clostridium species in the microbiome have been linked to brain disorders [12], and an imbalance of the Bacteroidetes and Firmicutes phyla has also been implicated in brain development disorders [13]. Suggestions that altered levels of gut commensals, including those of Bifidobacterium, Lactobacillus, Sutterella, Prevotella and Ruminococcus genera and of the Alcaligenaceae family are involved in immune-mediated central nervous system (CNS) disorders, are questioned by studies suggesting a lack of alteration in the microbiota between patients and healthy subjects [14]. This indicates that, at present, the practical effect of the link between the microbiome and human brain diseases is poorly characterised. Accordingly, more direct analytical studies are required to identify the therapeutic impact of altering the microbiome on CNS disorders.

In recognition of the potential positive effect that certain bacterial strains may have on the animal gut, various strains have been proposed for use in the treatment of various diseases (see, for example, [14-17]). Also, certain strains, including mostly Lactobacillus and Bifidobacterium strains, have been proposed for use in treating various inflammatory and autoimmune diseases that are not directly linked to the intestines (see [18] and [19] for reviews). In addition, a range of probiotics have been investigated in animal models to determine a role of the gut microbiome in modulating emotional behaviour, and Bifidobacterium and Lactobacillus are the main genera showing beneficial effects, reducing anxiety and repetitive behaviours, and increasing social interaction [20-22]. However, the relationship between different diseases and different bacterial strains, and the precise effects of particular bacterial strains on the gut and at a systemic level and on any particular types of diseases, are poorly characterised, particularly for central nervous system diseases.

There is a growing body of evidence to suggest that the microbiota-gut-brain axis is affected in autism spectrum disorders (ASD) and other neurodevelopmental and neuropsychiatric disorders. Animal models have provided considerable insight into how the microbiota may be involved in ASD. Furthermore, preclinical studies have demonstrated that targeting the gut microbiota through administration of beneficial live biotherapeutics display efficacy in improving autistic-related behaviour in animal models, including the maternal immune activation (MIA) mouse model and the black and tan, brachyuric (BTBR) mouse. The BTBR mouse is a genetically modified, inbred mouse strain that displays a number of behaviours associated with ASD such as impaired sociability, repetitive behaviour and increased anxiety. Moreover, these mice also exhibit gastrointestinal dysfunctions along with alterations to the composition of the gut microbiota. Consequently, it represents an appropriate animal model for investigating the role of the microbiota-gut-brain axis in ASD.

Accordingly, there is a requirement in the art for new methods of treating central nervous system disorders. There is also a requirement for the potential effects of gut bacteria to be characterised so that new therapies using gut bacteria can be developed.

SUMMARY OF THE INVENTION

The inventors have developed new therapies for treating and preventing central nervous system disorders. In particular, the inventors have developed new therapies for treating and preventing central nervous system disorders and conditions mediated by the microbiota-gut-brain axis. In particular, the inventors have identified that bacterial strains of the genus Blautia can be effective for treating and preventing diseases and conditions mediated by the microbiota-gut-brain axis. As described in the examples, oral administration of compositions comprising a Blautia strain may reduce symptoms associated with dysfunction of the microbiota-gut-brain axis in a mouse model of autism spectrum disorders. In addition, as described in the examples, oral administration of compositions comprising a Blautia strain may modulate the levels of signalling molecules associated with the function of the microbiota-gut-brain axis, and neurodevelopmental and neuropsychiatric disorders.

Therefore, in a first embodiment, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating or preventing a central nervous system disorder or condition. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia stercoris for use in a method of treating or preventing a central nervous system disorder or condition. Compositions using Blautia stercoris may be particularly effective for treating a central nervous system disorder or condition. In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia wexlerae for use in a method of treating or preventing a central nervous system disorder or condition. In particular embodiments, the central nervous system disorder or condition is mediated by the microbiota-gut-brain axis. In further embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating or preventing a neurodevelopmental disorder or a neuropsychiatric condition. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia stercoris for use in a method of treating or preventing a neurodevelopmental disorder or neuropsychiatric condition. Compositions using Blautia stercoris may be particularly effective for treating a neurodevelopmental disorder or neuropsychiatric condition. In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia wexlerae for use in a method of treating or preventing a neurodevelopmental disorder or neuropsychiatric condition. The inventors have identified that treatment with bacterial strains from this genus can provide clinical benefits in mouse models of central nervous system disorders, in particular those mediated by the microbiota-gut-brain axis. The inventors have identified that treatment with bacterial strains from this genus may modulate signalling in the central, autonomic and enteric nervous systems; may modulate the activity of the hypothalamus-pituitary-adrenal (HPA) axis pathway; may modulate neuroendocrine and/or neuroimmune pathways; and/or may modulate the levels of commensal metabolites, inflammatory markers and/or gastrointestinal permeability of a subject. Compositions using Blautia stercoris may be particularly effective at modulating signalling in the central, autonomic and enteric nervous systems; modulating the activity of the hypothalamus-pituitary-adrenal (HPA) axis pathway; modulating neuroendocrine and/or neuroimmune pathways; and/or modulating the levels of commensal metabolites, inflammatory markers and/or gastrointestinal permeability of a subject. In certain embodiments compositions using Blautia wexlerae may also be effective.

In particular embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating or preventing a disease or condition selected from the group consisting of: autism spectrum disorders (ASDs); child developmental disorder; obsessive compulsive disorder (OCD); major depressive disorder; depression; seasonal affective disorder; anxiety disorders; chronic fatigue syndrome (myalgic encephalomyelitis); stress disorder; post-traumatic stress disorder; schizophrenia spectrum disorders; schizophrenia; bipolar disorder; psychosis; mood disorder; dementia; Alzheimer's; Parkinson's disease; and/or chronic pain. In further embodiments, the compositions of the invention may be useful for treating or preventing multiple sclerosis; motor neuron disease; Huntington's disease; Guillain-Barre syndrome and/or meningitis. The effect shown for the bacterial strains from the genus Blautia on the microbiota-gut-brain axis and on diseases mediated by the microbiota-gut-brain axis may provide therapeutic benefits for other diseases and conditions mediated by the microbiota-gut-brain axis, such as those listed above. In other embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating comorbidities associated with diseases and conditions mediated by the microbiota-gut-brain axis, such as those listed above. In particularly preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating gastrointestinal comorbidities associated with diseases and conditions mediated by the microbiota-gut-brain axis, such as those listed above. The mouse model experiments used in this application for the assessment of the symptoms of autism spectrum disorders are known in the art to be applicable for the assessment of the symptoms other central nervous system disorders including those listed above [23-25].

In particularly preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating or preventing autism spectrum disorders, such as autism. The inventors have identified that treatment with Blautia strains can reduce symptom severity in a mouse model of autism spectrum disorders and can prevent or reduce stereotyped, repetitive, compulsive and anxious behaviour. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in the treatment of autism spectrum disorders. Compositions using Blautia may be particularly effective for treating autism spectrum disorders. In preferred embodiments, the invention provides a composition for use in reducing stereotyped, repetitive, compulsive or anxious behaviour, in particular in the treatment of autism spectrum disorders. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in the treatment of the behavioural symptoms of autism spectrum disorders. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia for use in the treatment of the gastrointestinal symptoms of autism spectrum disorders. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in the treatment of the behavioural and gastrointestinal symptoms of autism spectrum disorders. Treatment with Blautia strains may modulate signalling in the central, autonomic and enteric nervous systems; may modulate the activity of the HPA axis pathway; may modulate neuroendocrine and/or neuroimmune pathways; and/or may modulate the levels of commensal metabolites, inflammatory markers and/or gastrointestinal permeability of a subject, all of which are implicated in the neuropathology of autism spectrum disorders. In certain embodiments, treatment with Blautia strains may modulate the levels of oxytocin and/or vasopressin hormones. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia stercoris for use in a method of treating or preventing autism spectrum disorders. Compositions using Blautia stercoris may be particularly effective for treating autism spectrum disorders. In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia wexlerae for use in a method of treating or preventing autism spectrum disorders.

In further preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating or preventing obsessive compulsive disorder (OCD). In preferred embodiments, the invention provides a composition for use in reducing stereotyped, repetitive, compulsive or anxious behaviour in the treatment of OCD. Treatment with Blautia strains may modulate signalling in the central, autonomic and enteric nervous systems; may modulate the activity of the HPA axis pathway; may modulate neuroendocrine and/or neuroimmune pathways; and/or may modulate the levels of commensal metabolites and/or gastrointestinal permeability of a subject, all of which are implicated in the neuropathology of OCD. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia stercoris for use in a method of treating or preventing OCD. Compositions using Blautia stercoris may be particularly effective for treating OCD. In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia wexlerae for use in a method of treating or preventing OCD.

In further preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating or preventing major depressive disorder (MDD). Treatment with Blautia strains may provide clinical benefits in a mouse model of depression. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in the treatment of depression. Compositions using Blautia strains may be particularly effective for treating depression. In preferred embodiments, the invention provides a composition for use in reducing stereotyped, repetitive, compulsive or anxious behaviour in the treatment of depression. Treatment with Blautia strains may modulate signalling in the central, autonomic and enteric nervous systems; may modulate the activity of the HPA axis pathway; may modulate neuroendocrine and/or neuroimmune pathways; and may modulate the levels of commensal metabolites, inflammatory markers and/or gastrointestinal permeability of a subject, all of which are implicated in the neuropathology of MDD. In certain embodiments, treatment with Blautia strains may modulate the levels of oxytocin and/or vasopressin hormones. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia stercoris for use in a method of treating or preventing MDD. Compositions using Blautia stercoris may be particularly effective for treating MDD. In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia wexlerae for use in a method of treating or preventing MDD.

In further preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating or preventing anxiety disorders. Treatment with Blautia strains reduces disease incidence and disease severity in a mouse model of anxiety in the examples of this application. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in the treatment of anxiety disorder. Compositions using Blautia strains may be particularly effective for treating anxiety disorder. In preferred embodiments, the invention provides a composition for use in reducing stereotyped, repetitive, compulsive or anxious behaviour in the treatment of anxiety. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia stercoris for use in a method of treating or preventing anxiety disorders. Compositions using Blautia stercoris may be particularly effective for treating anxiety disorders. In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia wexlerae for use in a method of treating or preventing anxiety disorders.

In further preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating or preventing stress disorders, such as post-traumatic stress disorder. Compositions comprising a bacterial strain of the genus Blautia may reduce stress in mouse models of stress disorders. Treatment with Blautia strains may modulate signalling in the central, autonomic and enteric nervous systems; may modulate the activity of the HPA axis pathway; may modulate neuroendocrine and/or neuroimmune pathways; and may modulate the levels of commensal metabolites, inflammatory markers and/or gastrointestinal permeability of a subject, all of which are implicated in the neuropathology of stress disorder. In certain embodiments, treatment with Blautia strains may modulate the levels of oxytocin and/or vasopressin hormones. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia stercoris for use in a method of treating or preventing stress disorders. Compositions using Blautia stercoris may be particularly effective for treating stress disorders. In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia wexlerae for use in a method of treating or preventing stress disorders.

In further preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating or preventing schizophrenia spectrum and psychotic disorders, such as schizophrenia. Compositions comprising a bacterial strain of the genus Blautia may improve positive and negative symptoms in mouse models of schizophrenia spectrum and psychotic disorders. Treatment with Blautia strains may modulate signalling in the central, autonomic and enteric nervous systems; may modulate the activity of the HPA axis pathway; may modulate neuroendocrine and/or neuroimmune pathways; and may modulate the levels of commensal metabolites and/or gastrointestinal permeability of a subject, all of which are implicated in the neuropathology of schizophrenia spectrum and psychotic disorders. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia stercoris for use in a method of treating or preventing schizophrenia spectrum and psychotic disorders. Compositions using Blautia stercoris may be particularly effective for treating schizophrenia spectrum and psychotic disorders. In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia wexlerae for use in a method of treating or preventing schizophrenia spectrum and psychotic disorders.

In further preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating or preventing bipolar disorder. Compositions comprising a bacterial strain of the genus Blautia may reduce occasions of mania and/or depression in mouse models of bipolar disorder. Treatment with Blautia strains may modulate signalling in the central, autonomic and enteric nervous systems; may modulate the activity of the HPA axis pathway; may modulate neuroendocrine and/or neuroimmune pathways; and may modulate the levels of commensal metabolites, inflammatory markers and/or gastrointestinal permeability of a subject, all of which are implicated in the neuropathology of bipolar disorder. In certain embodiments, treatment with Blautia strains may modulate the levels of oxytocin and/or vasopressin hormones. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia stercoris for use in a method of treating or preventing bipolar disorder. Compositions using Blautia stercoris may be particularly effective for treating bipolar disorder. In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia wexlerae for use in a method of treating or preventing bipolar disorder.

In further preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating or preventing neurocognitive disorders, such as Alzheimer's disease. Compositions comprising a bacterial strain of the species genus Blautia may improve cognitive and behavioural functioning in mouse models of neurocognitive disorders. Treatment with Blautia strains may modulate signalling in the central, autonomic and enteric nervous systems; may modulate the activity of the HPA axis pathway; may modulate neuroendocrine and/or neuroimmune pathways; and may modulate the levels of commensal metabolites and/or gastrointestinal permeability of a subject, all of which are implicated in the neuropathology of neurocognitive disorders. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia stercoris for use in a method of treating or preventing neurocognitive disorders. Compositions using Blautia stercoris may be particularly effective for treating neurocognitive disorders. In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia wexlerae for use in a method of treating or preventing neurocognitive disorders.

In further preferred embodiments, the invention provides a composition comprising a bacterial strain of the genus Blautia, for use in a method of treating or preventing Parkinson's disease. Compositions comprising a bacterial strain of the genus Blautia may improve motor and cognitive functions in mouse models of Parkinson's disease. Treatment with Blautia strains may modulate signalling in the central, autonomic and enteric nervous systems; may modulate the activity of the HPA axis pathway; may modulate neuroendocrine and/or neuroimmune pathways; and may modulate the levels of commensal metabolites, inflammatory markers and/or gastrointestinal permeability of a subject, all of which are implicated in the neuropathology of Parkinson's disease. In certain embodiments, treatment with Blautia strains may modulate the levels of oxytocin and/or vasopressin hormones. In preferred embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia stercoris for use in a method of treating or preventing Parkinson's disease. Compositions using Blautia stercoris may be particularly effective for treating Parkinson's disease. In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia wexlerae for use in a method of treating or preventing Parkinson's disease.

In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia hydrogenotrophica for use in a method of treating or preventing a central nervous system disorder or condition. Compositions using Blautia hydrogenotrophica may be particularly effective for treating a central nervous system disorder or condition.

In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia hydrogenotrophica for use in a method of treating or preventing a neurodevelopmental disorder or neuropsychiatric condition. Compositions using Blautia hydrogenotrophica may be particularly effective for treating a neurodevelopmental disorder or neuropsychiatric condition.

Compositions using Blautia hydrogenotrophica may be particularly effective at modulating signalling in the central, autonomic and enteric nervous systems; modulating the activity of the hypothalamus-pituitary-adrenal (HPA) axis pathway; modulating neuroendocrine and/or neuroimmune pathways; and/or modulating the levels of commensal metabolites, inflammatory markers and/or gastrointestinal permeability of a subject. In a particularly preferred embodiment, Blautia hydrogenotrophica modulates the levels of butyrate. In certain embodiments, the modulation of the levels of butyrate treats or prevents a central nervous system disorder or condition.

In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia hydrogenotrophica for use in a method of treating or preventing autism spectrum disorders. Compositions using Blautia hydrogenotrophica may be particularly effective for treating autism spectrum disorders.

In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia hydrogenotrophica for use in a method of treating or preventing OCD. Compositions using Blautia hydrogenotrophica may be particularly effective for treating OCD.

In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia hydrogenotrophica for use in a method of treating or preventing MDD. Compositions using Blautia hydrogenotrophica may be particularly effective for treating MDD.

In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia hydrogenotrophica for use in a method of treating or preventing anxiety disorders. Compositions using Blautia hydrogenotrophica may be particularly effective for treating anxiety disorders.

In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia hydrogenotrophica for use in a method of treating or preventing stress disorders. Compositions using Blautia hydrogenotrophica may be particularly effective for treating stress disorders.

In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia hydrogenotrophica for use in a method of treating or preventing schizophrenia spectrum and psychotic disorders. Compositions using Blautia hydrogenotrophica may be particularly effective for treating schizophrenia spectrum and psychotic disorders.

In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia hydrogenotrophica for use in a method of treating or preventing bipolar disorder. Compositions using Blautia hydrogenotrophica may be particularly effective for treating bipolar disorder.

In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia hydrogenotrophica for use in a method of treating or preventing neurocognitive disorders. Compositions using Blautia hydrogenotrophica may be particularly effective for treating neurocognitive disorders.

In certain embodiments, the invention provides a composition comprising a bacterial strain of the species Blautia hydrogenotrophica for use in a method of treating or preventing Parkinson's disease. Compositions using Blautia hydrogenotrophica may be particularly effective for treating Parkinson's disease.

In certain embodiments, the compositions of the invention are for use in a method of modulating the microbiota-gut-brain axis in the treatment or prevention of a disease or condition mediated by the microbiota-gut-brain axis. In particular, the compositions of the invention may be used in modulating the microbiota-gut-brain axis in the treatment or prevention of autism spectrum disorders; obsessive compulsive disorder; major depressive disorder; anxiety disorders; stress disorders; schizophrenia spectrum disorders; bipolar disorders; neurocognitive disorders and Parkinson's disease.

In preferred embodiments of the invention, the bacterial strain in the composition is of Blautia stercoris. Closely related strains may also be used, such as bacterial strains that have a 16s rRNA sequence that is at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% identical to the 16s rRNA sequence of a bacterial strain of Blautia stercoris. 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:1 or 2. Preferably, the sequence identity is to SEQ ID NO:2. Preferably, the bacterial strain for use in the invention has the 16s rRNA sequence represented by SEQ ID NO:2.

In preferred embodiments of the invention, the bacterial strain in the composition is of Blautia wexlerae. Closely related strains may also be used, such as bacterial strains that have a 16s rRNA sequence that is at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% identical to the 16s rRNA sequence of a bacterial strain of Blautia wexlerae. 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:3 or 4. Preferably, the sequence identity is to SEQ ID NO:4. Preferably, the bacterial strain for use in the invention has the 16s rRNA sequence represented by SEQ ID NO:4.

In preferred embodiments of the invention, the bacterial strain in the composition is of Blautia hydrogenotrophica. Closely related strains may also be used, such as bacterial strains that have a 16s rRNA sequence that is at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% identical to the 16s rRNA sequence of a bacterial strain of Blautia hydrogenotrophica. 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:7. Most preferably, the bacterial strain in the composition is the Blautia hydrogenotrophica strain deposited under accession number DSM 14294.

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 central nervous system disorders and conditions, in particular those mediated by the microbiota-gut-brain axis. 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 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 mediated by dysfunction of the microbiota-gut-brain axis, comprising administering a composition comprising a bacterial strain of the genus Blautia.

In developing the above invention, the inventors have identified and characterised a bacterial strain that is particularly useful for therapy. The Blautia stercoris strain of the invention is shown to be effective for treating the diseases described herein, such as autism spectrum disorder. Therefore, in another aspect, the invention provides a cell of the Blautia stercoris strain deposited under accession number NCIMB 42381, or a derivative thereof. The invention also provides compositions comprising such cells, or biologically pure cultures of such cells. The invention also provides a cell of the Blautia stercoris strain deposited under accession number NCIMB 42381, or a derivative thereof, for use in therapy, in particular for the diseases described herein.

In especially preferred embodiments, the invention provides a composition comprising the strain deposited under accession number NCIMB 42381, for use in a method of treating or preventing a central nervous system disorder or condition. In especially preferred embodiments, the invention provides a composition comprising the strain deposited under accession number NCIMB 42381, for use in a method of treating or preventing a neurodevelopmental disorder or a neuropsychiatric condition. In especially preferred embodiments, the invention provides a composition comprising the strain deposited under accession number NCIMB 42381, for use in a method of treating or preventing autism spectrum disorder, or preferably autism. In especially preferred embodiments, the invention provides a composition comprising the strain deposited under accession number NCIMB 42381, for use in a method of reducing stereotyped, repetitive, compulsive or anxious behaviour, especially in the treatment of autism.

In developing the above invention, the inventors have identified and characterised a further bacterial strain that is particularly useful for therapy. The Blautia wexlerae strain of the invention is shown to be effective for treating the diseases described herein, such as autism spectrum disorder. Therefore, in another aspect, the invention provides a cell of the Blautia wexlerae strain deposited under accession number NCIMB 42486, or a derivative thereof. The invention also provides compositions comprising such cells, or biologically pure cultures of such cells. The invention also provides a cell of the Blautia wexlerae strain deposited under accession number NCIMB 42486, or a derivative thereof, for use in therapy, in particular for the diseases described herein.

In especially preferred embodiments, the invention provides a composition comprising the strain deposited under accession number NCIMB 42486, for use in a method of treating or preventing a central nervous system disorder or condition. In especially preferred embodiments, the invention provides a composition comprising the strain deposited under accession number NCIMB 42486, for use in a method of treating or preventing a neurodevelopmental disorder or a neuropsychiatric condition. In especially preferred embodiments, the invention provides a composition comprising the strain deposited under accession number NCIMB 42486, for use in a method of treating or preventing autism spectrum disorder, or preferably autism. In especially preferred embodiments, the invention provides a composition comprising the strain deposited under accession number NCIMB 42486, for use in a method of reducing stereotyped, repetitive, compulsive or anxious behaviour, especially in the treatment of autism.

In developing the above invention, the inventors have identified and characterised a bacterial strain that is particularly useful for therapy. The Blautia hydrogenotrophica strain of the invention is shown to be effective for treating the diseases described herein, such as autism spectrum disorder. Therefore, in another aspect, the invention provides a cell of the Blautia hydrogenotrophica strain deposited under accession number DSM 14294, or a derivative thereof. The invention also provides compositions comprising such cells, or biologically pure cultures of such cells. The invention also provides a cell of the Blautia hydrogenotrophica strain deposited under accession number DSM 14294, or a derivative thereof, for use in therapy, in particular for the diseases described herein.

In especially preferred embodiments, the invention provides a composition comprising the strain deposited under accession number DSM 14294, for use in a method of treating or preventing a central nervous system disorder or condition. In especially preferred embodiments, the invention provides a composition comprising the strain deposited under accession number DSM 14294 for use in a method of treating or preventing a neurodevelopmental disorder or a neuropsychiatric condition. In especially preferred embodiments, the invention provides a composition comprising the strain deposited under accession number DSM 14294, for use in a method of treating or preventing autism spectrum disorder, or preferably autism. In especially preferred embodiments, the invention provides a composition comprising the strain deposited under accession number DSM 14294, for use in a method of reducing stereotyped, repetitive, compulsive or anxious behaviour, especially in the treatment of autism.

BRIEF DESCRIPTION OF DRAWINGS

FIGS. 1A-1B: Effect of treatment with MRX006 on C57Bl/6 mice in the 3-chamber test. *p<0.05 novel versus familiar (FIG. 1A) and **p<0.01 object versus animal (FIG. 1B).

FIG. 2: Effect of treatment with MRX006 on C57Bl/6 mice in the forced swim test. Mrx006 significantly different to the vehicle group **p<0.01; Vehicle group significantly different to the naïve group ##p<0.01.

FIG. 3: Effect of treatment with MRX006 on C57Bl/6 mice in the tail suspension test.

FIGS. 4A-4C: Effect of treatment with MRX006 on C57Bl/6 mice in the fear conditioning test. * MRX006 significantly different to the Vehicle group; # Vehicle significantly different to the Naïve group; *p<0.05, #p<0.05, ##p<0.01, ###p<0.001. FIG. 4A, Acquisition; FIG. 4B, Retrieval; FIG. 4C, Extinction.

FIG. 5: Effect of treatment with MRX006 on C57Bl/6 mice in the novel object recognition test. # Significantly different vs. familiar object within groups; #p<0.05.

FIG. 6: Effect of treatment with MRX006 on C57Bl/6 mice in the marble burying test.

FIGS. 7A-7D: Effect of treatment with MRX006 on C57Bl/6 mice in the elevated plus maze test. FIG. 7A, Time spent in closed arms; FIG. 7B, % Time in closed arms; FIG. 7C, Time spent in open arms; FIG. 7D, % Time in open arms.

FIG. 8: Effect of treatment with MRX006 on stress induced hyperthermia in C57Bl/6 mice. # Vehicle group significantly different to naïve group, #p<0.05.

FIG. 9: Effect of treatment with MRX006 on circulating oxytocin levels in C57Bl/6 mice. * Mrx006 significantly different from vehicle group; *p<0.05.

FIGS. 10A-10B: Effect of treatment with MRX006 on corticosterone plasma levels in C57Bl/6 mice. * Significantly different to naïve (FIG. 10A) or vehicle (FIG. 10B) group; *p<0.05.

FIG. 11: Effect of treatment with MRX006 on gut permeability in C57Bl/6 mice.

FIG. 12: Effect of treatment with MRX006 on organ weight and colon length in C57Bl/6 mice.

FIGS. 13A-13F: Effect of treatment with MRX006 on BTBR mice in the three chamber social interaction test. ##p<0.01 relative to respective within group. ###p<0.001 relative to respective within group. *p<0.05 relative to vehicle group. FIG. 13A, Time in chamber, objective vs conspecific; FIG. 13B, Time in chamber, familiar vs novel; FIG. 13C, % Time, % Time spent investigating novel conspecific; FIG. 13D, Interaction time, objective vs conspecific; FIG. 13E, Interaction time, familiar vs novel; FIG. 13F, % Time interacting, % Time spent investigating novel conspecific.

FIG. 14: Effect of treatment with MRX006 on BTBR mice in the forced intruder test.

FIG. 15: Effect of treatment with MRX006 on BTBR mice in the marble burying test. *p<0.05 relative to vehicle group as determined by a priori comparisons.

FIGS. 16A-16B: Effect of treatment with MRX006 on BTBR mice in the grooming test. *p<0.05 relative to vehicle group. **p<0.01 relative to vehicle group as revealed by a priori comparisons.

FIG. 16A, Grooming time, vehicle vs Mrx006; FIG. 16B Grooming time, grooming.

FIGS. 17A-17D: Effect of treatment with MRX006 on BTBR mice in elevated plus maze test. FIG. 17A, % time spent in closed arms; FIG. 17B, % time spent in open arms; FIG. 17C, No. entries to closed arm; FIG. 17D, No. entries to open arm.

FIGS. 18A-18F: Effect of treatment with MRX006 on BTBR mice in the open field arena. *p<0.05 relative to vehicle group. FIG. 18A, Total distance moved; FIG. 18B, Time spent in outer zone; FIG. 18C, Time spent in inner zone; FIG. 18D, Total distance moved; FIG. 18E, Time spent in outer zone; FIG. 18F, Time spent in inner zone.

FIG. 19: Effect of treatment with MRX006 on BTBR mice in the forced swim test.

FIGS. 20A-20B: Effect of treatment with MRX006 on BTBR mice in female urine sniffing test. #p<0.05 relative to water vehicle group. **p<0.01 relative to vehicle group. FIG. 20A, Time spent sniffing, Female urine sniffing test; FIG. 20B, Female urine sniffing test, vehicle vs Mrx006, water vs urine.

FIGS. 21A-21C: Effect of treatment with MRX006 on BTBR mice in the novel object recognition test. FIG. 21A, NOR day 1; FIG. 21B, NOR day 2; FIG. 21C, NOR discrimination Index.

FIGS. 22A-22B: Effect of treatment with MRX006 on ex vivo gastrointestinal permeability in BTBR mice. FIG. 22A, Ex vivo colon; FIG. 22B, Ex vivo ileum.

FIG. 23: Effect of treatment with MRX006 on in vivo gastrointestinal permeability in BTBR mice.

FIG. 24: Effect of treatment with MRX006 on in vivo gastrointestinal motility in BTBR mice. *p<0.05 relative to vehicle group as revealed by a priori comparisons.

FIG. 25: Effect of treatment with MRX006 on stress-induced corticosterone plasma levels in BTBR mice.

FIGS. 26A-26D: Effect of treatment with MRX006 on organ weight and colon length in BTBR mice. FIG. 26A, Adrenal weight % body weight; FIG. 26B, Spleen weight % body weight; FIG. 26C, Caecum weight % body weight; FIG. 26D, Colon length.

FIG. 27: Effect of treatment with MRX006 on weight of BTBR mice over time.

FIG. 28: Chronic treatment with Mrx006 decreased the number of marbles buried in a MIA mice model. #p<0.05 relative to control group; **p<0.01 relative to vehicle MIA group.

FIGS. 29A-29B: Effect of chronic treatment with MRX006 on sociability in MIA mice in the social transmission of food preference test. FIG. 29A, 0 Hour; FIG. 29B, 24 Hour.

FIGS. 30A-30C: Chronic treatment with MRX006 attenuates stress-induced locomotor activity caused by exposure to the open field arena in MIA mice. ##p<0.01 relative to control group, *p<0.05 relative to vehicle MIA group. FIG. 30A, Total distance moved; FIG. 30B, Time spent in outer zone; FIG. 30C, Time spent in inner zone.

FIG. 31: Effect of chronic treatment with MRX006 on depressive-like behaviour in MIA mice in the female urine sniffing test. &p<0.05 relative to respective water group.

FIG. 32: Effect of treatment with MRX006 on in vivo gastrointestinal motility in MIA mice.

FIGS. 33A-33C: Effect of treatment with MRX006 on organ weight and colon length in MIA mice. FIG. 33A, Colon length; FIG. 33B, Caecum weight % body weight; FIG. 33C, Spleen weight % body weight.

FIG. 34: Effect of treatment with MRX006 on circulating cytokine concentrations in BTBR mice.

FIGS. 35A-35B: Effect of treatment with MRX008 on MIA mice in the marble burying test. ##p<0.01 relative to control group. FIG. 35A, control vs. vehicle vs. Mrx008; FIG. 35B, control vs. vehicle.

FIGS. 36A-36B: Effect of chronic treatment with MRX008 on MIA mice in the social transmission of food preference test. FIG. 36A, T0; FIG. 36B, T24.

FIG. 37: Effect of chronic treatment with MRX008 on MIA mice in the forced swimming test.

FIG. 38: Effect of chronic treatment with MRX008 on intestinal permeability.

FIG. 39: Effect of chronic treatment with MRX008 on intestinal motility. ##p<0.01 relative to control group.

FIGS. 40A-40B: Effect of chronic treatment with MRX008 on BTBR mice in the social transmission of food preference test. FIG. 40A, STFP T0; FIG. 40B, STFP 24 HR.

FIG. 41: Effect of chronic treatment with MRX008 on BTBR mice in the forced intruder test.

FIG. 42: Effect of chronic treatment with MRX008 on BTBR mice in the marble burying test.

FIGS. 43A-43D: Effect of chronic treatment with MRX008 on BTBR mice in the elevated plus maze. FIG. 43A, % time spent in closed arms; FIG. 43B, % time spent in open arms; FIG. 43C, No. entries to closed arms; FIG. 43D, No. entries to open arms.

FIGS. 44A-44C: Effect of chronic treatment with MRX008 on BTBR mice in the open field arena. *p<0.05 relative to vehicle group as revealed by a priori pairwise comparisons. FIG. 44A, Distance moved; FIG. 44B, Time spent in outer zone; FIG. 44C, Time spent inner zone.

FIG. 45: Effect of chronic treatment with MRX008 on BTBR mice in the forced swim test.

FIG. 46: Effect of chronic treatment with MRX008 on depressive-like behaviour in BTBR mice in the female urine sniffing test. ##p<0.01 relative to water vehicle group.

FIG. 47: Effect of chronic treatment with MRX008 on in vivo intestinal motility in BTBR mice.

FIGS. 48A-48D: Effect of chronic treatment with MRX008 on selective anatomical markers in BTBR mice. FIG. 48A, Adrenal weight % body weight; FIG. 48B, Spleen weight % body weight; FIG. 48C, Caecum weight % body weight; FIG. 48D, Colon length.

FIGS. 49A-49D: Effect of chronic treatment with MRX006 on expression of oxytoxin, vasopressin and their respective receptors in the hypothalamus of BTBR mice. *p<0.05 relative to the vehicle group. FIG. 49A, OXTR mRNA; FIG. 49B, AVPR1b mRNA; FIG. 49C, OXT mRNA; FIG. 49D, AV mRNA.

FIGS. 50A-50D: Effect of chronic treatment with MRX006 on expression of oxytoxin, vasopressin and their respective receptors in the amygdala of BTBR mice. *p<0.05 relative to the vehicle group. FIG. 50A, OXTR mRNA; FIG. 50B, AVPR1b mRNA; FIG. 50C, OXT mRNA; FIG. 50D, AVP mRNA.

FIGS. 51A-51H: Effect of chronic treatment with Blautia hydrogenotrophica and butyrate on BTBR mice in the open field arena. The data in FIGS. 51B, 51D, 51F and 51H are identical to that in FIGS. 51A, 51C, 51E and 51G, respectively, except the PBS and LYO control numbers have been pooled. p≤0.05: * vs. C57BL/6 (same treatment, where applicable); # vs. PBS same genotype; § BTBR: But vs. PBS or Bact vs. Lyo. PBS is the negative control for butyrate administration; LYO is the negative control for bacterial (Blautia hydrogenotrophica) administration; BUT is the experimental administration of butyrate; BACT is the experimental administration of Blautia hydrogenotrophica.

FIGS. 52A-52B: Effect of chronic treatment with Blautia hydrogenotrophica and butyrate on BTBR mice in the marble burying test. The data in FIG. 52B are identical to that in FIG. 52A, except the PBS and LYO control numbers have been pooled. p≤0.05: * vs. C57BL/6 (same treatment, where applicable); # vs. PBS same genotype; § BTBR: But vs. PBS or Bact vs. Lyo. PBS is the negative control for butyrate administration; LYO is the negative control for bacterial (Blautia hydrogenotrophica) administration; BUT is the experimental administration of butyrate; BACT is the experimental administration of Blautia hydrogenotrophica.

FIGS. 53A-53B: Effect of chronic treatment with Blautia hydrogenotrophica and butyrate on BTBR mice in the digging test. FIG. 53A shows the time spent digging, while FIG. 53B shows the number of digging bouts. p<0.05: * vs. C57BL/6 (same treatment, where applicable); # vs. PBS same genotype; § BTBR: But vs. PBS or Bact vs. Lyo. PBS is the negative control for butyrate administration; LYO is the negative control for bacterial (Blautia hydrogenotrophica) administration; BUT is the experimental administration of butyrate; BACT is the experimental administration of Blautia hydrogenotrophica.

FIGS. 54A-54F: Effect of chronic treatment with Blautia hydrogenotrophica and butyrate on BTBR mice in the self-grooming test. FIG. 54A shows the time spent grooming; FIG. 54C shows the number of grooming bouts, and FIG. 54E shows the time spent grooming per bout. The data in FIGS. 54B, 54D and 54F are identical to FIGS. 54A, 54C, and 54E respectively, except the PBS and LYO control numbers have been pooled. P<0.05: * vs. C57BL/6 (same treatment, where applicable); # vs. PBS same genotype; § BTBR: But vs. PBS or Bact vs. Lyo. PBS is the negative control for butyrate administration; LYO is the negative control for bacterial (Blautia hydrogenotrophica) administration; BUT is the experimental administration of butyrate; BACT is the experimental administration of Blautia hydrogenotrophica.

FIG. 55: Effect of Blautia hydrogenotrophica (1010/day for 14 days) on short chain fatty acids production (RMN 1H) in caecal contents of healthy HIM rats.

FIG. 56: Qpcr evaluation of B. hydrogenotrophica population in faecal samples of IBS-HMA rats treated or not with a composition comprising B. hydrogenotrophica (BlautiX) for 28 days.

FIGS. 57A-57B: Short chain fatty acids (SCFA) concentrations in caecal samples of IBS-HMA rats treated or not with B. hydrogenotrophica (Blautix) for 28 days. FIG. 57A shows concentration of total SCFA. FIG. 57B shows concentration of Acetic acid, Propionic acid and Butyric acid.

FIGS. 58A-58B: Effect of chronic treatment with Blautia hydrogenotrophica and butyrate on BTBR mice in the three chamber test. FIG. 58A shows the effect of administration on sociability (the preference for sniffing an object or another mouse), while FIG. 58B shows the preference for social novelty (i.e. sniffing a new mouse vs. a familiar mouse). p<0.05: S vs. 50%; * vs. C57BL/6 (same treatment, where applicable); # vs. PBS same genotype; § BTBR: But vs. PBS or Bact vs. Lyo. PBS is the negative control for butyrate administration; LYO is the negative control for bacterial (Blautia hydrogenotrophica) administration; BUT is the experimental administration of butyrate; BACT is the experimental administration of Blautia hydrogenotrophica.

FIG. 59: Effect of chronic treatment with MRX006 on expression of oxytoxin and oxytoxin receptor in the hypothalamic cell lines.

FIG. 60: Effect of chronic treatment with MRX006 on ex vivo gastrointestinal permeability and tight junction expression in colon in BALBc mouse model.

FIG. 61: Effect of chronic treatment with MRX006 on ex vivo gastrointestinal permeability and tight junction expression in ileum in BALBc mouse model.

FIG. 62: Effect of chronic treatment with MRX006 on caecal short chain fatty acid production in BALBc mouse model.

FIG. 63: Effect of chronic treatment with MRX006 on cytokine expression from splenocytes in BALBc mouse model.

FIG. 64: Effect of chronic treatment with MRX006 on plasma levels of amino acids in BALBc mouse model.

FIG. 65: Effect of chronic treatment with MRX006 on neurotransmitter levels in the brainstem in BALBc mouse model.

FIG. 66: Effect of chronic treatment with MRX006 on gene expression of hippocampus neurotransmitter receptors in BALBc mouse model.

FIG. 67: Effect of chronic treatment with MRX006 on gene expression of amygdalar neurotransmitter receptors in BALBc mouse model.

FIG. 68: Effect of chronic treatment with MRX006 on gene expression of prefrontal cortex neurotransmitter receptors in BALBc mouse model.

FIG. 69: Effect of chronic treatment with MRX006 on gene expression of inflammatory markers in the hippocampus in BALBc mouse model.

FIG. 70: Effect of chronic treatment with MRX006 on gene expression of inflammatory markers in the amygdalar in BALBc mouse model.

FIG. 71: Effect of chronic treatment with MRX006 on gene expression of inflammatory markers in the prefrontal cortex in BALBc mouse model.

FIG. 72: Effect of chronic treatment with MRX006 on gene expression of hippocampal endocrine markers in BALBc mouse model.

FIG. 73: Effect of chronic treatment with MRX006 on gene expression of amygdalar endocrine markers in BALBc mouse model.

FIG. 74: Effect of chronic treatment with MRX006 on gene expression of prefrontal cortex endocrine markers in BALBc mouse model.

FIG. 75: Effect of chronic treatment with MRX006 on in vivo gastrointestinal permeability in the colon and ileum in MIA mouse model.

FIG. 76: Effect of treatment with MRX006 on social novelty in the three chamber social interaction test in MIA mice.

FIG. 77: Effect of treatment with MRX006 on social preference in the three chamber social interaction test in MIA mice.

FIG. 78: Effect of treatment with MRX006 on MIA mice in the grooming test.

FIG. 79: Effect of treatment with MRX006 on MIA mice in the elevated plus maze test.

FIG. 80: Effect of treatment with MRX006 on MIA mice in the forced swim test.

FIG. 81: Effect of treatment with MRX006 on stress-induced corticosterone plasma levels in MIA mice.

DISCLOSURE OF THE INVENTION

Bacterial Strains

The compositions of the invention comprise a bacterial strain of the genus Blautia. The examples demonstrate that bacteria of this species are useful for treating or preventing autism spectrum disorders and central nervous system disorders mediated by the microbiota-gut-brain axis. The mouse model experiments used in this application for the assessment of the symptoms of autism spectrum disorders are known in the art to be applicable for the assessment of the symptoms other central nervous system disorders including those listed above

The invention provides a composition comprising a bacterial strain of the genus Blautia for use in therapy, for example, for use in treating or preventing a central nervous system disorder or condition, in particular a central nervous system disorder or condition mediated by the microbiota-gut-brain axis. In certain embodiments, the compositions of the invention comprise strains of the genus Blautia and do not contain any other bacterial genera. In certain embodiments, the compositions of the invention comprise a single strain of the genus Blautia and do not contain any other bacterial strains, genera or species.

Examples of Blautia strains for use in the invention include Blautia stercoris, B. faecis, B. coccoides, B. glucerasea, B. hansenii, B. hydrogenotrophica, B. luti, B. producta, B. schinkii and B. wexlerae. Preferred species are Blautia stercoris, B. wexlerae and B. hydrogenotrophica. The Blautia species are Gram-reaction-positive, non-motile bacteria that may be either coccoid or oval and all are obligate anaerobes that produce acetic acid as the major end product of glucose fermentation [26]. Blautia may be isolated from the human gut, although B. producta was isolated from a septicaemia sample. The GenBank accession number for the 16S rRNA gene sequence of Blautia stercoris strain GAM6-1T is HM626177 (disclosed herein as SEQ ID NO: 1). An exemplary Blautia stercoris strain is described in [27]. The type strain of Blautia wexlerae is WAL 14507=ATCC BAA-1564=DSM 19850 [28]. The GenBank accession number for the 16S rRNA gene sequence of Blautia wexlerae strain WAL 14507 T is EF036467 (disclosed herein as SEQ ID NO:3). This exemplary Blautia wexlerae strain is described in [28].

The Blautia stercoris bacterium deposited under accession number NCIMB 42381 was tested in the Examples and is also referred to herein as MRX006 (strain 830). The terms “MRX006”, “MRx0006” “Mrx006”, “Mrx0006” and strain 830 are used interchangeably herein. A 16S rRNA sequence for MRX006 (830 strain) that was tested is provided in SEQ ID NO:2. MRX006 (Strain 830) was deposited with the international depositary authority NCIMB, Ltd. (Ferguson Building, Aberdeen, AB21 9YA, Scotland) by GT Biologics Ltd. (Life Sciences Innovation Building, Aberdeen, AB25 2ZS, Scotland) on 12 Mar. 2015 as “Blautia stercoris 830” and was assigned accession number NCIMB 42381. GT Biologics Ltd. subsequently changed its name to 4D Pharma Research Limited.

The genome of MRX006 (strain 830) comprises a chromosome and plasmid. A chromosome sequence for MRX006 (strain 830) is provided in SEQ ID NO:5. A plasmid sequence for MRX006 (strain 830) is provided in SEQ ID NO:6. These sequences were generated using the PacBio RS II platform.

The Blautia wexlerae bacterium deposited under accession number NCIMB 42486 was tested in the Examples and is also referred to herein as strain MRX008. The terms “MRX008”, “MRx0008” “Mrx008” and “Mrx0008” are used interchangeably herein. A 16S rRNA sequence for the MRX008 strain that was tested is provided in SEQ ID NO:4. Strain MRX008 was deposited with the international depositary authority NCIMB, Ltd. (Ferguson Building, Aberdeen, AB21 9YA, Scotland) by 4D Pharma Research Ltd. (Life Sciences Innovation Building, Aberdeen, AB25 2ZS, Scotland) on 16 Nov. 2015 as “Blautia/Ruminococcus” and was assigned accession number NCIMB 42486.

A further preferred strain of the invention is the Blautia hydrogenotrophica bacterium deposited under accession number DSM 14294. This strain was deposited with the Deutsche Sammlung von Mikroorganismen [German Microorganism Collection] (Mascheroder Weg 1b, 38124 Braunschweig, Germany) under accession number DSM 14294 as “S5a33” on 10 May 2001. The depositor was INRA Laboratoire de Microbiologie CR de Clermont-Ferrand/Theix 63122 Saint Genes Champanelle, France. Ownership of the deposits has passed to 4D Pharma Plc by way of assignment. 4D Pharma Plc has authorised, by way of an agreement, 4D Pharma Research Limited to refer to the deposited biological material in the application and has given its unreserved and irrevocable consent to the deposited material being made available to the public. The deposit under accession number DSM 14294 was published on 11 May 2000.

The Blautia hydrogenotrophica bacterium deposited under accession number DSM 14294 was tested in the Examples and is a preferred strain of the invention.

Bacterial strains closely related to the strain tested in the examples are also expected to be effective for treating or preventing autism spectrum disorders and central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis. In certain embodiments, 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 the 16s rRNA sequence of a bacterial strain of Blautia stercoris. 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:1 or 2. Preferably, the sequence identity is to SEQ ID NO:2. Preferably, the bacterial strain for use in the invention has the 16s rRNA sequence represented by SEQ ID NO:2. In certain embodiments, 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 the 16s rRNA sequence of a bacterial strain of Blautia wexlerae. 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:3 or 4. Preferably, the sequence identity is to SEQ ID NO:4. Preferably, the bacterial strain for use in the invention has the 16s rRNA sequence represented by SEQ ID NO:4.

In certain embodiments, the bacterial strain for use in the invention has a chromosome with sequence identity to SEQ ID NO:5. In preferred embodiments, the bacterial strain for use in the invention has a chromosome 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:5 across at least 60% (e.g. at least 65%, 70%, 75%, 80%, 85%, 95%, 96%, 97%, 98%, 99% or 100%) of SEQ ID NO:5. For example, the bacterial strain for use in the invention may have a chromosome with at least 90% sequence identity to SEQ ID NO:5 across 70% of SEQ ID NO:5, or at least 90% sequence identity to SEQ ID NO:5 across 80% of SEQ ID NO:5, or at least 90% sequence identity to SEQ ID NO:5 across 90% of SEQ ID NO:5, or at least 90% sequence identity to SEQ ID NO:5 across 100% of SEQ ID NO:5, or at least 95% sequence identity to SEQ ID NO:5 across 70% of SEQ ID NO:5, or at least 95% sequence identity to SEQ ID NO:5 across 80% of SEQ ID NO:5, or at least 95% sequence identity to SEQ ID NO:5 across 90% of SEQ ID NO:5, or at least 95% sequence identity to SEQ ID NO:5 across 100% of SEQ ID NO:5, or at least 98% sequence identity to SEQ ID NO:5 across 70% of SEQ ID NO:5, or at least 98% sequence identity to SEQ ID NO:5 across 80% of SEQ ID NO:5, or at least 98% sequence identity to SEQ ID NO:5 across 90% of SEQ ID NO:5, or at least 98% sequence identity to SEQ ID NO:5 across 100% of SEQ ID NO:5.

In certain embodiments, the bacterial strain for use in the invention has a plasmid with sequence identity to SEQ ID NO:6. In preferred embodiments, the bacterial strain for use in the invention has a plasmid 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:6 across at least 60% (e.g. at least 65%, 70%, 75%, 80%, 85%, 95%, 96%, 97%, 98%, 99% or 100%) of SEQ ID NO:6. For example, the bacterial strain for use in the invention may have a plasmid with at least 90% sequence identity to SEQ ID NO:6 across 70% of SEQ ID NO:6, or at least 90% sequence identity to SEQ ID NO:6 across 80% of SEQ ID NO:6, or at least 90% sequence identity to SEQ ID NO:6 across 90% of SEQ ID NO:6, or at least 90% sequence identity to SEQ ID NO:6 across 100% of SEQ ID NO:6, or at least 95% sequence identity to SEQ ID NO:6 across 70% of SEQ ID NO:6, or at least 95% sequence identity to SEQ ID NO:6 across 80% of SEQ ID NO:6, or at least 95% sequence identity to SEQ ID NO:6 across 90% of SEQ ID NO:6, or at least 95% sequence identity to SEQ ID NO:6 across 100% of SEQ ID NO:6, or at least 98% sequence identity to SEQ ID NO:6 across 70% of SEQ ID NO:6, or at least 98% sequence identity to SEQ ID NO:6 across 80% of SEQ ID NO:6, or at least 98% sequence identity to SEQ ID NO:6 across 90% of SEQ ID NO:6, or at least 98% sequence identity to SEQ ID NO:6 across 100% of SEQ ID NO:6.

In certain embodiments, the bacterial strain for use in the invention has a chromosome with sequence identity to SEQ ID NO:5 and a plasmid with sequence identity to SEQ ID NO:6.

Bacterial strains that are biotypes of the bacterium deposited under accession number 42381 are also expected to be effective for treating or preventing autism spectrum disorder and central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis. Bacterial strains that are biotypes of the bacterium deposited under accession number 42486 are also expected to be effective for treating or preventing autism spectrum disorder and central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis. A biotype is a closely related strain that has the same or very similar physiological and biochemical characteristics.

Strains that are biotypes of the bacterium deposited under accession number NCIMB 42381 or 42486 and that are suitable for use in the invention may be identified by sequencing other nucleotide sequences for the bacterium deposited under accession number NCIMB 42381 or 42486. For example, substantially the whole genome may be sequenced and a biotype strain for use in the invention may have 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). For example, in some embodiments, a biotype strain has at least 98% sequence identity across at least 98% of its genome or at least 99% sequence identity across 99% of its genome. Other suitable sequences for use in identifying biotype strains may include hsp60 or repetitive sequences such as BOX, ERIC, (GTG)5, or REP or [29]. Biotype strains may have sequences with at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% sequence identity to the corresponding sequence of the bacterium deposited under accession number NCIMB 42381 or 42486.

In some embodiments, a biotype strain has a sequence with at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% sequence identity to the corresponding sequence of strain MRX006 deposited as NCIMB 42381 and comprises a 16S rRNA sequence that is at least 99% identical (e.g. at least 99.5% or at least 99.9% identical) to SEQ ID NO:2. In some embodiments, a biotype strain has a sequence with at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% sequence identity to the corresponding sequence of strain MRX006 deposited as NCIMB 42381 and has the 16S rRNA sequence of SEQ ID NO:2.

In some embodiments, a biotype strain has a sequence with at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% sequence identity to the corresponding sequence of strain MRX008 deposited as NCIMB 42486 and comprises a 16S rRNA sequence that is at least 99% identical (e.g. at least 99.5% or at least 99.9% identical) to SEQ ID NO:4. In some embodiments, a biotype strain has a sequence with at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% sequence identity to the corresponding sequence of strain MRX008 deposited as NCIMB 42486 and has the 16S rRNA sequence of SEQ ID NO:4.

Alternatively, strains that are biotypes of a bacterium deposited under accession number NCIMB 42381 or 42486 and that are suitable for use in the invention may be identified by using the accession number NCIMB 42381 deposit or the accession number NCIMB 42486 deposit, and restriction fragment analysis and/or PCR analysis, for example by using fluorescent amplified fragment length polymorphism (FAFLP) and repetitive 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 other Blautia stercoris or Blautia wexlerae strains.

In certain embodiments, strains that are biotypes of a bacterium deposited under accession number NCIMB 42381 or 42486 and that are suitable for use in the invention are strains that provide the same pattern as a bacterium deposited under accession number NCIMB 42381 or 42486 when analysed by amplified ribosomal DNA restriction analysis (ARDRA), for example when using Sau3AI restriction enzyme (for exemplary methods and guidance see, for example, [30]). Alternatively, biotype strains are identified as strains that have the same carbohydrate fermentation patterns as a bacterium deposited under accession number NCIMB 42381 or 42486.

Other Blautia stercoris strains that are useful in the compositions and methods of the invention, such as biotypes of the bacterium deposited under accession number NCIMB 42381 or 42486, may be identified using any appropriate method or strategy, including the assays described in the examples. For instance, strains for use in the invention may be identified by culturing in anaerobic YCFA and/or administering the bacteria to an autism spectrum disorder mouse model and then assessing cytokine levels. In particular, bacterial strains that have similar growth patterns, metabolic type and/or surface antigens to the bacterium deposited under accession number NCIMB 42381 or 42486 may be useful in the invention. A useful strain will have comparable immune modulatory activity to the NCIMB 42381 or 42486 strain. In particular, a biotype strain will elicit comparable effects on the autism spectrum disorder models 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 Blautia stercoris strain deposited under accession number NCIMB 42381. This is the exemplary MRX006 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 Blautia stercoris strain deposited under accession number NCIMB 42381, or a derivative thereof. The invention also provides a composition comprising a cell of the Blautia stercoris strain deposited under accession number NCIMB 42381, or a derivative thereof. The invention also provides a biologically pure culture of the Blautia stercoris strain deposited under accession number NCIMB 42381. The invention also provides a cell of the Blautia stercoris strain deposited under accession number NCIMB 42381, or a derivative thereof, for use in therapy, in particular for the diseases described herein.

A particularly preferred strain of the invention is the Blautia wexlerae strain deposited under accession number NCIMB 42486. This is the exemplary MRX008 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 Blautia wexlerae strain deposited under accession number NCIMB 42486, or a derivative thereof. The invention also provides a composition comprising a cell of the Blautia wexlerae strain deposited under accession number NCIMB 42486, or a derivative thereof. The invention also provides a biologically pure culture of the Blautia wexlerae strain deposited under accession number NCIMB 42486. The invention also provides a cell of the Blautia wexlerae strain deposited under accession number NCIMB 42486, or a derivative thereof, for use in therapy, in particular for the diseases described herein.

A derivative of the strain deposited under accession number NCIMB 42381 or 42486 may be a daughter strain (progeny) or a strain cultured (subcloned) from the original. A derivative of the strain deposited under accession number NCIMB 42381 or 42486 may be a daughter strain (progeny) or a strain cultured (subcloned) from the original. A derivative of a strain of the invention may be modified, for example at the genetic level, without ablating the biological activity. In particular, a derivative strain of the invention is therapeutically active. A derivative strain will have comparable immune modulatory activity to the original NCIMB 42381 or 42486 strain. In particular, a derivative strain will elicit comparable effects on the central nervous system disorder or condition 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 derivative of the NCIMB 42381 strain will generally be a biotype of the NCIMB 42381 strain. A derivative of the NCIMB 42486 strain will generally be a biotype of the NCIMB 42486 strain.

References to cells of the Blautia stercoris strain deposited under accession number NCIMB 42381 encompass any cells that have the same safety and therapeutic efficacy characteristics as the strains deposited under accession number NCIMB 42381, and such cells are encompassed by the invention. Thus, in some embodiments, reference to cells of the Blautia stercoris strain deposited under accession number NCIMB 42381 refers only to the MRX006 strain deposited under NCIMB 42381 and does not refer to a bacterial strain that was not deposited under NCIMB 42381. In some embodiments, reference to cells of the Blautia stercoris strain deposited under accession number NCIMB 42381 refers to cells that have the same safety and therapeutic efficacy characteristics as the strains deposited under accession number NCIMB 42381, but which are not the strain deposited under NCIMB 42381.

References to cells of the Blautia wexlerae strain deposited under accession number NCIMB 42486 encompass any cells that have the same safety and therapeutic efficacy characteristics as the strains deposited under accession number NCIMB 42486, and such cells are encompassed by the invention. Thus, in some embodiments, reference to cells of the Blautia wexlerae strain deposited under accession number NCIMB 42486 refers only to the strain deposited under NCIMB 42486 and does not refer to a bacterial strain that was not deposited under NCIMB 42486. In some embodiments, reference to cells of the Blautia wexlerae strain deposited under accession number NCIMB 42486 refers to cells that have the same safety and therapeutic efficacy characteristics as the strains deposited under accession number NCIMB 42486, but which are not the strain deposited under NCIMB 42486.

In certain embodiments, the bacterial strain for use in the invention has a chromosome with sequence identity to SEQ ID NO:5, for example as described above, and a 16S rRNA sequence with sequence identity to SEQ ID NO:1, 2, 3 or 4, for example as described above, preferably with a 16s rRNA sequence that is at least 99% identical to SEQ ID NO: 2 or 4, more preferably which comprises the 16S rRNA sequence of SEQ ID NO:2 or 4.

In certain embodiments, the bacterial strain for use in the invention has a chromosome with sequence identity to SEQ ID NO:5, for example as described above, and is effective for treating or preventing central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

In certain embodiments, the bacterial strain for use in the invention has a chromosome with sequence identity to SEQ ID NO:5, for example as described above, and a 16S rRNA sequence with sequence identity to SEQ ID NO: 1, 2, 3 or 4, for example as described above, and is effective for treating or preventing central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

In certain embodiments, the bacterial strain for use in the invention has a 16s rRNA sequence that is at least 99%, 99.5% or 99.9% identical to the 16s rRNA sequence represented by SEQ ID NO: 2 or 4 (for example, which comprises the 16S rRNA sequence of SEQ ID NO:2 or 4) and a chromosome with at least 95% sequence identity to SEQ ID NO:5 across at least 90% of SEQ ID NO:5, and which is effective for treating or preventing central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

In certain embodiments, the bacterial strain for use in the invention is a Blautia stercoris and has a 16s rRNA sequence that is at least 99%, 99.5% or 99.9% identical to the 16s rRNA sequence represented by SEQ ID NO: 2 or 4 (for example, which comprises the 16S rRNA sequence of SEQ ID NO:2 or 4) and a chromosome with at least 98% sequence identity (e.g. at least 99% or at least 99.5% sequence identity) to SEQ ID NO:5 across at least 98% (e.g. across at least 99% or at least 99.5%) of SEQ ID NO:5, and which is effective for treating or preventing central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

In certain embodiments, the bacterial strain for use in the invention has a plasmid with sequence identity to SEQ ID NO:6, for example as described above, and a 16S rRNA sequence with sequence identity to SEQ ID NO:1, 2, 3 or 4, for example as described above, preferably with a 16s rRNA sequence that is at least 99% identical to SEQ ID NO: 2 or 4, more preferably which comprises the 16S rRNA sequence of SEQ ID NO:2 or 4.

In certain embodiments, the bacterial strain for use in the invention has a plasmid with sequence identity to SEQ ID NO:6, for example as described above, and is effective for treating or preventing central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

In certain embodiments, the bacterial strain for use in the invention has a plasmid with sequence identity to SEQ ID NO:6, for example as described above, and a 16S rRNA sequence with sequence identity to SEQ ID NO: 1, 2, 3 or 4, for example as described above, and is effective for treating or preventing central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

In certain embodiments, the bacterial strain for use in the invention has a 16s rRNA sequence that is at least 99%, 99.5% or 99.9% identical to the 16s rRNA sequence represented by SEQ ID NO: 2 or 4 (for example, which comprises the 16S rRNA sequence of SEQ ID NO:2 or 4) and a plasmid with at least 95% sequence identity to SEQ ID NO:6 across at least 90% of SEQ ID NO:6, and which is effective for treating or preventing central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

In certain embodiments, the bacterial strain for use in the invention is a Blautia stercoris and has a 16s rRNA sequence that is at least 99%, 99.5% or 99.9% identical to the 16s rRNA sequence represented by SEQ ID NO: 2 or 4 (for example, which comprises the 16S rRNA sequence of SEQ ID NO:2 or 4) and a plasmid with at least 98% sequence identity (e.g. at least 99% or at least 99.5% sequence identity) to SEQ ID NO:6 across at least 98% (e.g. across at least 99% or at least 99.5%) of SEQ ID NO:6, and which is effective for treating or preventing central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

In certain embodiments, the bacterial strain for use in the invention has a chromosome with sequence identity to SEQ ID NO:5, for example as described above, a plasmid with sequence identity to SEQ ID NO:6, for example as described above, and a 16S rRNA sequence with sequence identity to SEQ ID NO:1, 2, 3 or 4, for example as described above, preferably with a 16s rRNA sequence that is at least 99% identical to SEQ ID NO: 2 or 4, more preferably which comprises the 16S rRNA sequence of SEQ ID NO:2 or 4.

In certain embodiments, the bacterial strain for use in the invention has a chromosome with sequence identity to SEQ ID NO:5, for example as described above, and a plasmid with sequence identity to SEQ ID NO:6, for example as described above, and is effective for treating or preventing central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

In certain embodiments, the bacterial strain for use in the invention has a chromosome with sequence identity to SEQ ID NO:5, for example as described above, a plasmid with sequence identity to SEQ ID NO:6, for example as described above, and a 16S rRNA sequence with sequence identity to SEQ ID NO: 1, 2, 3 or 4, for example as described above, and is effective for treating or preventing central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

In certain embodiments, the bacterial strain for use in the invention has a 16s rRNA sequence that is at least 99%, 99.5% or 99.9% identical to the 16s rRNA sequence represented by SEQ ID NO: 2 or 4 (for example, which comprises the 16S rRNA sequence of SEQ ID NO:2 or 4), a chromosome with at least 95% sequence identity to SEQ ID NO:5 across at least 90% of SEQ ID NO:5, and a plasmid at least 95% sequence identity to SEQ ID NO:6 across at least 90% of SEQ ID NO:6, and which is effective for treating or preventing central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

In certain embodiments, the bacterial strain for use in the invention is a Blautia stercoris and has a 16s rRNA sequence that is at least 99%, 99.5% or 99.9% identical to the 16s rRNA sequence represented by SEQ ID NO: 2 or 4 (for example, which comprises the 16S rRNA sequence of SEQ ID NO:2 or 4), a chromosome with at least 98% sequence identity (e.g. at least 99% or at least 99.5% sequence identity) to SEQ ID NO:5 across at least 98% (e.g. across at least 99% or at least 99.5%) of SEQ ID NO:5, and a plasmid with at least 98% sequence identity (e.g. at least 99% or at least 99.5% sequence identity) to SEQ ID NO:6 across at least 98% (e.g. across at least 99% or at least 99.5%) of SEQ ID NO:6, and which is effective for treating or preventing central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

In preferred embodiments, the bacterial strains in the compositions of the invention are viable and capable of partially or totally colonising the intestine.

Blautia hydrogenotrophica (previously known as Ruminococcus hydrogenotrophicus) has been isolated from the guts of mammals, is strictly anaerobic, and metabolises H2/CO2 to acetate, which may be important for human nutrition and health. The type strain of Blautia hydrogenotrophica is S5a33=JCM 14656. The GenBank accession number for the 16S rRNA gene sequence of Blautia hydrogenotrophica strain S5a36 is X95624.1 (disclosed herein as SEQ ID NO:7). This exemplary Blautia hydrogenotrophica strain is described in [28] and [31]. The S5a33 strain and the S5a36 strain correspond to two subclones of a strain isolated from a faecal sample of a healthy subject. They show identical morphology, physiology and metabolism and have identical 16S rRNA sequences. Thus, in some embodiments, the Blautia hydrogenotrophica for use in the invention has the 16S rRNA sequence of SEQ ID NO:7.

In certain embodiments, 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 the 16s rRNA sequence of a bacterial strain of Blautia hydrogenotrophica. 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:7. Preferably, the bacterial strain for use in the invention has the 16s rRNA sequence represented by SEQ ID NO:7.

Bacterial strains that are biotypes of the bacterium deposited under accession number DSM 14294 are also expected to be effective for treating or preventing autism spectrum disorder and central nervous system disorders and conditions, in particular central nervous system disorders and conditions mediated by the microbiota-gut-brain axis.

Strains that are biotypes of the bacterium deposited under accession number DSM 14294 and that are suitable for use in the invention may be identified by sequencing other nucleotide sequences for the bacterium deposited under accession number DSM 14294. For example, substantially the whole genome may be sequenced and a biotype strain for use in the invention may have 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). For example, in some embodiments, a biotype strain has at least 98% sequence identity across at least 98% of its genome or at least 99% sequence identity across 99% of its genome. Other suitable sequences for use in identifying biotype strains may include hsp60 or repetitive sequences such as BOX, ERIC, (GTG)5, or REP or [29]. Biotype strains may have sequences with at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% sequence identity to the corresponding sequence of the bacterium deposited under accession number DSM 14294.

In some embodiments, a biotype strain has a sequence with at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% sequence identity to the corresponding sequence of the strain deposited as accession number DSM 14294 and comprises a 16S rRNA sequence that is at least 99% identical (e.g. at least 99.5% or at least 99.9% identical) to SEQ ID NO:7. In some embodiments, a biotype strain has a sequence with at least 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% sequence identity to the corresponding sequence of the strain deposited as accession number DSM 14294 and has the 16S rRNA sequence of SEQ ID NO:7.

Alternatively, strains that are biotypes of a bacterium deposited under accession number DSM 14294 and that are suitable for use in the invention may be identified by using the accession number DSM 14294 deposit, and restriction fragment analysis and/or PCR analysis, for example by using fluorescent amplified fragment length polymorphism (FAFLP) and repetitive 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 other Blautia hydrogenotrophica strains.

In certain embodiments, strains that are biotypes of a bacterium deposited under accession number NCIMB 42381 or 42486 and that are suitable for use in the invention are strains that provide the same pattern as a bacterium deposited under accession number DSM 14294 when analysed by amplified ribosomal DNA restriction analysis (ARDRA), for example when using Sau3AI restriction enzyme (for exemplary methods and guidance see, for example, [30]). Alternatively, biotype strains are identified as strains that have the same carbohydrate fermentation patterns as a bacterium deposited under accession number DSM 14294.

Other Blautia hydrogenotrophica strains that are useful in the compositions and methods of the invention, such as biotypes of the bacterium deposited under accession number DSM 14294, may be identified using any appropriate method or strategy, including the assays described in the examples. For instance, strains for use in the invention may be identified by culturing in anaerobic YCFA and/or administering the bacteria to an autism spectrum disorder mouse model and then assessing cytokine levels. In particular, bacterial strains that have similar growth patterns, metabolic type and/or surface antigens to the bacterium deposited under accession number DSM 14294 may be useful in the invention. A useful strain will have comparable immune modulatory activity to the accession number DSM 14294 strain. In particular, a biotype strain will elicit comparable effects on the autism spectrum disorder models 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 Blautia hydrogenotrophica strain deposited under accession number DSM 14294. This is the exemplary 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 Blautia hydrogenotrophica strain deposited under accession number DSM 14294, or a derivative thereof. The invention also provides a composition comprising a cell of the Blautia hydrogenotrophica strain deposited under accession number DSM 14294, or a derivative thereof. The invention also provides a biologically pure culture of the Blautia hydrogenotrophica strain deposited under accession number DSM 14294. The invention also provides a cell of the Blautia hydrogenotrophica strain deposited under accession number DSM 14294, or a derivative thereof, for use in therapy, in particular for the diseases described herein.

A derivative of the strain deposited under accession number DSM 14294 may be a daughter strain (progeny) or a strain cultured (subcloned) from the original. A derivative of the strain deposited under accession number DSM 14294 may be a daughter strain (progeny) or a strain cultured (subcloned) from the original. A derivative of a strain of the invention may be modified, for example at the genetic level, without ablating the biological activity. In particular, a derivative strain of the invention is therapeutically active. A derivative strain will have comparable immune modulatory activity to the original strain deposited under accession number DSM 14294. In particular, a derivative strain will elicit comparable effects on the central nervous system disorder or condition 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 derivative of the DSM 14294 strain will generally be a biotype of the DSM 14294 strain.

References to cells of the Blautia hydrogenotrophica strain deposited under accession number DSM 14294 encompass any cells that have the same safety and therapeutic efficacy characteristics as the strains deposited under accession number DSM 14294, and such cells are encompassed by the invention. Thus, in some embodiments, reference to cells of the Blautia hydrogenotrophica strain deposited under accession number DSM 14294 refers only to the strain deposited under DSM 14294 and does not refer to a bacterial strain that was not deposited under DSM 14294. In some embodiments, reference to cells of the Blautia hydrogenotrophica strain deposited under accession number DSM 14294 refers to cells that have the same safety and therapeutic efficacy characteristics as the strains deposited under accession number DSM 14294, but which are not the strain deposited under DSM 14294.

In preferred embodiments, the bacterial strains in the compositions of the invention are viable and capable of partially or totally colonising the intestine.

Therapeutic Uses

Modulation of the Microbiota-Gut-Brain Axis

Communication between the gut and the brain (the microbiota-gut-brain axis) occurs via a bidirectional neurohumoral communication system. Recent evidence shows that the microbiota that resides in the gut can modulate brain development and produce behavioural phenotypes via the microbiota-gut-brain axis. Indeed, a number of reviews suggest a role of the microbiota-gut-brain axis in maintaining central nervous system functionality and implicate dysfunction of the microbiota-gut-brain axis in the development of central nervous system disorders and conditions [10],[13],[14],[32].

The bidirectional communication between the brain and the gut (i.e. the-gut-brain axis) includes the central nervous system, neuroendocrine and neuroimmune systems, including the hypothalamus-pituitary-adrenal (HPA) axis, sympathetic and parasympathetic arms of the autonomic nervous system (ANS), including the enteric nervous system (ENS) and the vagus nerve, and the gut microbiota.

As demonstrated in the examples, the compositions of the present invention can modulate the microbiota-gut-brain axis and reduce behavioural symptoms associated with a CNS disorder. Accordingly, the compositions of the invention may be useful for treating or preventing disorders of the central nervous system (CNS), in particular those disorders and conditions associated with dysfunction of the microbiota-gut-brain axis.

The compositions of the invention may also be useful for treating or preventing neurodevelopmental disorders and/or neuropsychiatric conditions. Neurodevelopmental diseases and neuropsychiatric conditions are often associated with the microbiota-gut-brain axis. The compositions of the invention may be useful for treating or preventing neurodevelopmental diseases and/or neuropsychiatric conditions mediated by dysfunction of the microbiota-gut-brain axis. In further preferred embodiments, the compositions of the invention are for use in treating or preventing a neurodevelopmental disorder or a neuropsychiatric condition.

In particular embodiments, the compositions of the invention may be useful for treating or preventing a disease or condition selected from the group consisting of: autism spectrum disorders (ASDs); child developmental disorder; obsessive compulsive disorder (OCD); major depressive disorder; depression; seasonal affective disorder; anxiety disorders; schizophrenia spectrum disorders; schizophrenia; bipolar disorder; psychosis; mood disorder; chronic fatigue syndrome (myalgic encephalomyelitis); stress disorder; post-traumatic stress disorder; dementia; Alzheimer's; Parkinson's disease; and/or chronic pain. In further embodiments, the compositions of the invention may be useful for treating or preventing motor neuron disease; Huntington's disease; Guillain-Barre syndrome and/or meningitis.

The compositions of the invention may be particularly useful for treating or preventing chronic disease, treating or preventing disease in patients that have not responded to other therapies (such as treatment with anti-psychotics and/or anti-depressants), and/or treating or preventing the tissue damage and symptoms associated with dysfunction of the microbiota-gut-brain axis.

In certain embodiments, the compositions of the invention modulate the CNS. In some embodiments, the compositions of the invention modulate the autonomic nervous system (ANS). In some embodiments, the compositions of the invention modulate the enteric nervous system (ENS). In some embodiments, the compositions of the invention modulate the hypothalamic, pituitary, adrenal (HPA) axis. In some embodiments, the compositions of the invention modulate the neuroendocrine pathway. In some embodiments, the compositions of the invention modulate the neuroimmune pathway. In some embodiments, the compositions of the invention modulate the CNS, the ANS, the ENS, the HPA axis and/or the neuroendocrine and neuroimmune pathways. In certain embodiments, the compositions of the invention module the levels of commensal metabolites and/or the gastrointestinal permeability of a subject.

The signalling of the microbiota-gut-brain axis is modulated by neural systems. Accordingly, in some embodiments, the compositions of the invention modulate signalling in neural systems. In certain embodiments, the compositions of the invention modulate the signalling of the central nervous system. In some embodiments, the compositions of the invention modulate signalling in sensory neurons. In other embodiments, the compositions of the invention modulate signalling in motor neurons. In some embodiments, the compositions of the invention modulate the signalling in the ANS. In some embodiments, the ANS is the parasympathetic nervous system. In preferred embodiments, the compositions of the invention modulate the signalling of the vagus nerve. In other embodiments, the ANS is the sympathetic nervous system. In other embodiments, the compositions of the invention modulate the signalling in the enteric nervous system. In certain embodiments, the signalling of ANS and ENS neurons responds directly to luminal contents of the gastrointestinal tract. In other embodiments, the signalling of ANS and ENS neurons responds indirectly to neurochemicals produced by luminal bacteria. In other embodiments, the signalling of ANS and ENS neurons responds to neurochemicals produced by luminal bacteria or enteroendocrine cells. In certain preferred embodiments, the neurons of the ENS activate vagal afferents that influence the functions of the CNS. In some embodiments, the compositions of the invention regulate the activity of enterochromaffin cells.

In certain embodiments, the compositions of the invention modulate fear conditioning in an animal model. In certain embodiments, the compositions of the invention can be used to modulate the development of fear and/or anxiety, and/or modulate the extent to which the fear and/or anxiety becomes extinct in a subject. In certain embodiments, the compositions of the invention can be used to modulate the extent of stress-induced hyperthermia in an animal model. In certain embodiments, the compositions of the invention modulate the level of stress and/or anxiety in a subject.

Autism Spectrum Disorder (ASD)

Autism spectrum disorder is a set of heterogeneous neurodevelopmental conditions, characterised by early-onset difficulties in social interaction, communication and unusually restricted, repetitive behaviour and interests. Symptoms can be recognised from a very early age but ASD is often diagnosed in more able children starting mainstream education. Autism represents the primary type of ASD.

Historically, autism has been diagnosed on the basis of three core domains: impaired social interaction, abnormal communication, and restricted and repetitive behaviours and interests. In the International Classification of Diseases (ICD-10R, WHO 1993) and the Diagnostic and Statistical Manual (DSM-IV, American Psychiatric Association, 2000), autism comes under the umbrella term of Pervasive Developmental Disorder (PDD), with four possible diagnostic subtypes: Asperger Syndrome, Childhood Autism/Autistic Disorder, Atyptical Autism, and PDD—not otherwise specified. In DMS-5, these diagnostic subtypes are combined into a single category of autism spectrum disorder (ASD) and the previous use of three core domains of impairment has been reduced to two main areas, namely social communication and interaction, and repetitive behaviour, which include sensory integration dysfunctions.

ASD is a ‘spectrum disorder’ as it affects each person in a variety of different ways and can range from very mild to severe. The functioning of the affected individual varies substantially depending on language abilities, level of intelligence, co-morbidity, composition of symptoms and access to services. Cognitive functioning, learning, attention and sensory processing are usually impaired.

DSM-IV states that the diagnosis of autism requires the presence of at least six symptoms, including a minimum of two measures of qualitative impairment in social interaction, one symptom of qualitative impairment in communication, and one symptom of restricted and repetitive behaviour. DMS-5 redefines diagnosis of ASD into two symptom domains: (i) social interaction and social communication deficits; and (ii) restricted, repetitive patterns of behaviour, interests or activities.

Co-morbid medical conditions are highly prevalent in ASDs. Co-morbid include anxiety and depression, seizures, attention deficits, aggressive behaviours, sleep problems, gastrointestinal disorders, epilepsy, mental retardation, intellectual disabilities and feeding difficulties.

The examples demonstrate that the compositions of the invention achieve a reduction in disease incidence and disease severity in an animal model of autism spectrum disorder and so they may be useful in the treatment or prevention of autism spectrum disorders.

ASD is a central nervous system disorder that is partially triggered by environmental factors. Therefore, dysfunction of the microbiota-gut-brain axis may be responsible for development and persistence of ASDs. Accordingly, in preferred embodiments, the composition of the invention are for use in treating or preventing autism spectrum disorders. In some embodiments, the compositions of the invention are for use in treating or preventing autism. In some embodiments, the autism is Pervasive Developmental Disorder (PDD). In another embodiment, the PDD is Asperger Syndrome, Childhood Autism/Autistic Disorder, Atyptical Autism and/or PDD—not otherwise specified. Accordingly, in some embodiments, the compositions of the invention are for use in treating or preventing autism spectrum disorders, autism, pervasive developmental disorder; Asperger Syndrome; Childhood Autism/Autistic Disorder, Atypical Autism and/or PDD—not otherwise specified.

The compositions of the invention may be useful for modulating the microbiota-gut-brain axis of a subject. Accordingly, in preferred embodiments the compositions of the invention are for use in preventing an ASD in a patient that has been identified as at risk of an ASD, or that has been diagnosed with an ASD at a prenatal or an early developmental stage; in childhood and/or in adulthood. The compositions of the invention may be useful for preventing the development of ASDs.

The compositions of the invention may be useful for managing or alleviating ASDs. Treatment or prevention of ASDs may refer to, for example, an alleviation of the severity of symptoms or a reduction in the frequency of exacerbations or the range of triggers that are a problem for the patient.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate at least one core symptom of ASDs.

In some embodiments, the compositions of the invention prevent, reduce or alleviate at least one of the two symptom domains of ASD classified in the DMS-5. In some embodiments, the compositions of the invention prevent, reduce or alleviate social interaction and/or social communication deficits. In some embodiments, the compositions of the invention prevent, reduce or alleviate restrictive, repetitive patterns of behaviour, interests or activities. In some embodiments, the compositions of the invention prevent, reduce or alleviate social interaction, social communication deficits and/or restrictive, repetitive patterns of behaviour, interests or activities.

In some embodiments, the compositions of the invention prevent, reduce or alleviate repetitive behaviour, stereotyped behaviour, compulsive behaviour, routine behaviour, sameness behaviour and restricted behaviour. In some embodiments, the compositions of the invention improve social awareness, social information processing, capacity for social communication, social anxiety/avoidance, and autistic preoccupations and traits in a subject with ASDs.

In some embodiments, the compositions of the invention prevent, reduce or alleviate additional symptoms associated with the core symptoms of ASDs. In some embodiments, the compositions of the invention prevent, reduce or alleviate irritability (including aggression, deliberate self-injury and temper tantrums), agitation, crying, lethargy, social withdrawal, stereotypic behaviour, hyperactivity, non-compliance, inappropriate speech, anxiety, depression, and/or over or under-controlled behaviour in a subject with ASDs. In some embodiments, the compositions of the invention improve cognitive functioning, learning, attention and/or sensory processing in a subject with ASD.

In other embodiments, the compositions of the invention improve secondary outcome measures in a subject with ASDs. In some embodiments, the secondary outcome measures include additional symptom and/or functional rating scales, behavioural scales and miscellaneous measures of interest.

In some embodiments, the compositions of the invention cause in a positive change in the diagnostic and/or symptomatic scale for the assessment of core symptoms of a subject with ASDs. In some embodiments, the diagnostic and/or symptomatic scale is the Autism Diagnostic Interview—Revised (ASI-R). In some embodiments, the diagnostic or symptomatic scale is the Autism Diagnostic Observation Schedule-Generic (ADOS-G) now ADOS-2. In other embodiments, the diagnostic or symptomatic scale is the Autism Diagnostic Interview Revised (ADI-R). In other embodiments, the diagnostic or symptomatic scale is the Diagnostic Interview for Social and Communication Disorders (DISCO). In yet other embodiments, the diagnostic or symptomatic scale is the Childhood Autism Rating Scale (CARS and CARS2).

In some embodiments, the compositions of the invention cause a positive change in generic measures of the efficacy endpoints of ASDs. In certain embodiments, the generic measures include, but are not limited to the Aberrant Behaviour Checklist (ABC), the Child Behaviour Checklist (CBCL), the Vineland-II Adaptive Behaviour Scales (VABS), the Social Responsiveness Scale (SRS), and/or the Repetitive Behaviour Scale—Revised (RBS-R).

In some embodiments, the compositions of the invention improve the Clinical Global Impression—Global Improvement (CGI-I) scale for assessing psychiatric and neurological disorders. In some embodiments, the compositions of the invention display a positive effect on global functioning of the subject with ASDs.

Additional scales would be known to a person skilled in the art. In some embodiments, the compositions of the invention would improve the outcome of diagnostic and/or symptomatic scales known to a person skilled in the art.

In certain embodiments, the compositions of the invention prevent, reduce or alleviate the incidence of comorbidities of ASDs. In some embodiments, the compositions of the invention prevent, reduce or alleviate the incidence of anxiety and depression, seizures, attention deficits, aggressive behaviours, sleep problems, gastrointestinal disorders (including irritable bowel syndrome (IBS)), epilepsy, mental retardation, intellectual disabilities and/or feeding difficulties. In certain embodiments, the compositions of the invention prevent, reduce or alleviate gastrointestinal comorbidities, such as abdominal pain, diarrhoea and flatulence.

In some embodiments, the compositions of the invention prevent, reduce or alleviate the symptoms of certain psychiatric and behavioural disorders that may present clinically with similarities to autism. Accordingly, in some embodiments, the compositions of the invention, prevent, reduce or alleviate attention deficit disorder (ADHD); affective/anxiety disorders; attachment disorders; oppositional defiant disorder (ODD); obsessive compulsive disorder (OCD) and/or psychoses including schizophrenia (cognitive impairment).

In some embodiments, the compositions of the invention are particularly effective at preventing, reducing or alleviating ASDs when used in combination with another therapy for treating ASDs. Such therapies include anti-psychotic, anti-anxiety and anti-depressant drugs. Such drugs include risperidone (Risperdal®); olanzapine (Zyprexa®); fluoxetine (Prozac®); sertraline (Zoloft®); fluvoxamine (Luvox®); clomipramine (Anafranil®); haloperidol (Haldol®); thioridazine; fluphenazine; chlorpromazine; ziprasidone (Geogon®); carbamazepine (Tegretol®); lamotrigine (Lamictal®); topiramate (Topomax®); valproic acid (Depakote®); methylphenidate (Ritalin®); diazepam (Valium®) and lorazepam (Ativan®).

The EMA Guidelines on the clinical development of medicinal products for the treatment of autism spectrum disorder state that, due to the heterogeneity of the diseases, it may not be possible to achieve a significant effect on all core symptoms with a single compound, and so short term efficacy has to be demonstrated on at least one core symptom. The live biotherapeutic strains used in the Examples have shown effective treatment of at least one core symptom of autistic spectrum disorder, so these strains and related Blautia strains are expected to be effective against human disease.

Obsessive Compulsive Disorder (OCD)

OCD is a heterogeneous, chronic and disabling disorder belonging to the anxiety disorders. According to the DSM-IV definition, the essential features of OCD are recurrent obsessions and/or compulsions (criterion A) that are severe and time consuming (more than one hour a day) or cause marked distress or significantly interfere with the subject's normal routine, occupational functioning, usual social activities or relationships (criterion C). As some point during the course of the disorder, the person has recognised that the obsessions or compulsions are excessive or unreasonable (criterion B).

Obsessions are defined as recurrent and persistent thoughts, impulses or images that are experienced as intrusive and inappropriate and cause marked anxiety or distress. The thoughts, impulses or images are not simply excessive worries about real-life problems, they are recognised by the patient as a product of his own mind (e.g. fear for contamination, symmetry obsession). The person attempts to ignore, suppress or neutralise the obsessions with some other thoughts or actions.

Compulsions are defined as repetitive behaviours (e.g. hand washing, ordering, hoarding, checking) or mental acts (e.g. praying, counting, repeating words silently) that the person feels driven to perform in response to an obsession or according to rules that must be applied rigidly.

OCD is often associated with co-morbidity rates of other psychiatric diseases including major depressive disorder, other anxiety disorders (generalised anxiety disorder, social anxiety disorder, panic disorder), substance abuse and eating disorders (anorexia and bulimia).

OCD is a psychiatric disorder that may develop or persist due to dysfunction of the microbiota-gut-brain axis. Accordingly, in preferred embodiments, the compositions of the invention are for use in treating or preventing OCD in a subject.

In certain embodiments, the compositions of the invention prevent, reduce or alleviate the essential symptomatic features of OCD. In certain embodiments, the compositions of the invention prevent, reduce or alleviate recurrent obsessions and/or compulsions in a subject. In certain embodiments, the obsessions are recurrent or persistent thoughts, impulses or images that are experiences as intrusive and inappropriate and cause marked anxiety or distress. In certain embodiments, the compulsions are repetitive behaviours that the subject feels driven to perform in response to an obsession or according to rules that must be applied rigidly.

In certain embodiments, the compositions of the invention improve symptoms of OCD in a subject accordingly to the Y-BOCS and/or the NIMH-OC diagnostic and/or symptomatic scales. In some embodiments, the Y-BOCS scale is used to monitor improvement of primary endpoints. In some embodiments, the NIMH-OC scale is used to monitor improvement of secondary parameters.

In some embodiments, the compositions of the invention improve the Clinical Global Impression—Global Improvement (CGI-I) scale for assessing psychiatric and neurological disorders. In some embodiments, the compositions of the invention display a positive effect on global social functioning (relationships, work, etc.) of the subject with ASDs. In some embodiments, the global scale is the Sheehan disability scale.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate at least one comorbidity of OCD. The comorbidities of OCD include major depressive disorder, other anxiety disorders (generalised anxiety disorder, social anxiety disorder, panic disorder), substance abuse and eating disorders (anorexia and bulimia) Gilles de la Tourette syndrome, ADHD (Attention-Deficit/Hyperactivity Disorder) and developmental disorders.

In some embodiments, the compositions of the invention are particularly effective at preventing, reducing or alleviating OCD when used in combination with another therapy for treating OCD. Such therapies include serotonin and dopamine reuptake inhibitors; clomipramine and anti-psychotics.

Major Depressive Disorder (MDD)

MDD is associated with substantial psychosocial dysfunction and high individual mental strain as well as with excess morbidity and mortality (the risk of suicide is considerable). The term major depressive disorder encompasses clinical depression, major depression, unipolar depression, unipolar disorder, recurrent depression and simply depression. The term major depressive disorder covers mood disorders; dysthymia; chronic depression; seasonal affective disorder and borderline personality disorder.

According to the DMS-5 criteria, MDD symptoms include a depressed mood, or loss of interest or pleasure in daily activities for more than two weeks; and impaired social, occupational and educational function. Specific symptoms, at least five of the following nine, present nearly every day: depressed mood or irritable most of the day; decreased interest or pleasure in most activities, most of each day; significant weight change or change in appetite; change in sleep (insomnia or hypersomnia); change in activity (psychomotor agitation or retardation); fatigue or loss of energy; guilt or worthlessness (feelings of worthlessness or excessive or inappropriate guilt); reduced concentration (diminished ability to think or concentrate, or more indecisiveness; and suicidality (thoughts of death or suicide, or subject has a suicide plan). In addition, MDD is associated with anxiety symptoms including irrational worry; preoccupation with unpleasant worries; trouble relaxing and/or feeling tense. MDD episodes can be mild, moderate or severe.

MDD episodes are often associated with comorbidity with other psychiatric disorders or with somatic disorders like Parkinson's disease, Alzheimer's disease, cerebrovascular disorders, cancer and chronic pain syndromes. MDD is frequently associated with a wide spectrum of other mental disorders as comorbidities including generalised anxiety disorder; anxiety disorder; substance use disorders; post-traumatic stress disorder (PTSD); personality disorders; pain; stress; irritable bowel syndrome; insomnia; headaches and interpersonal problems.

Major depressive disorder is a psychiatric disorder that may develop or persist due to dysfunction of the microbiota-gut-brain axis. Accordingly, in preferred embodiments, the compositions of the invention are for use in treating or preventing MDD in a subject.

In certain embodiments, the compositions of the invention are for use in treating or preventing acute major depressive episodes and/or the prevention of new episodes (recurrence prevention). In certain embodiments, the compositions of the invention prevent, reduce or alleviate the occurrence of mild, moderate or severe MDD episodes.

In certain embodiments, the compositions of the invention prevent, reduce or alleviate one or more of the symptoms of MDD as classified by the DMS-5 criteria listed herein. In a preferred embodiment, the compositions of the invention prevent, reduce or alleviate a depressed mood in a subject. In a preferred embodiment, the compositions of the invention prevent, reduce or alleviate a decreased interest or pleasure in most activities in a subject. In some embodiments, the compositions of the invention reduce the occurrence of symptoms of MDD within a 2-week period.

In some embodiments, the compositions of the invention improve the symptoms of MDD according to a symptomatic or diagnostic scale. Such scales for assessing symptomatic improvement include the Hamilton Rating Scale of Depression (HAMD) and the Montgomery Asberg Depression Rating Scale. In addition, the Zung Self-Rating Depression Scale (SDS) and Zung Self-Rating Anxiety Scale (SAS) are also suitable symptomatic improvement scales.

In some embodiments, the compositions of the invention improve the Clinical Global Impression—Global Improvement (CGI-I) scale for assessing psychiatric and neurological disorders. In some embodiments, the compositions of the invention display a positive effect on global social and occupational functioning of the subject with MDD.

In certain embodiments, the compositions of the invention are for use in treating or preventing treatment resistant MDD.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate at least one comorbidity of MDD. The comorbidities of MDD include generalised anxiety disorder; anxiety disorder; substance use disorders; post-traumatic stress disorder (PTSD); personality disorders; pain; stress; IBS; insomnia; headaches and interpersonal problems.

In some embodiments, the compositions of the invention are particularly effective at preventing, reducing or alleviating MDD when used in combination with another therapy for treating MDD. Such therapies include antidepressants, augmentation strategies (e.g. combination therapy, lithium and other mood stabilizers, thyroid hormones and atypical antipsychotics) or even second generation antipsychotics.

Anxiety Disorders

Anxiety disorders are a group of mental disorders characterised by feelings of anxiety and fear. There are a number of anxiety disorders including generalised anxiety disorder (GAD); specific phobia; social anxiety disorder; separation anxiety disorder; agroraphobia; panic disorder and selective mutism.

GAD is diagnosed according to DMS-5 in six criterion. The first criterion is too much anxiety or worry over more than six months wherein the anxiety or worry is present most of the time in regards to many activities. The second criterion is that the subject is unable to manage the symptoms of the first criterion. The third criterion is that at least three (one in children) of the following occurs: restlessness; tires easily; problems concentrating; irritability; muscle tension and problems with sleep. The final three criterion are that the symptoms results in significant social, occupational and functional impairment; the symptoms are not due to medications, drugs, or other physical health problems; and the symptoms do not fit better with another psychiatric problem such as panic disorder. All other anxiety disorders may be considered as differential diagnoses of GAD.

GAD is frequently associated with a wide spectrum of other mental disorders as comorbidities including depression; substance use disorders; stress; IBS; insomnia; headaches; pain; cardiac events; interpersonal problems and ADHD.

Anxiety disorders are psychiatric disorders that may develop or persist due to dysfunction of the microbiota-gut-brain axis. Accordingly, in preferred embodiments, the compositions of the invention are for use in treating or preventing anxiety disorders in a subject. In certain embodiments, the anxiety disorder is generalised anxiety disorder (GAD); specific phobia; social anxiety disorder; separation anxiety disorder; agoraphobia; panic disorder and selective mutism.

In certain embodiments, the compositions of the invention prevent, reduce or alleviate one or more of the symptoms of GAD in a subject as classified by the DMS-5 criteria listed herein. According to DMS-5, the same symptoms are associated with other anxiety disorders. Therefore, in certain embodiments, the compositions of the invention prevent, reduce or alleviate one or more of the symptoms of anxiety disorders in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate the anxiety or worry of the subject. In certain embodiments, the compositions of the invention reduce the occurrence of symptoms within a six month period. In certain embodiments, the composition of the invention prevents, reduces or alleviates restlessness; fatigue; loss of concentration; irritability; muscle tension; and/or problems with sleep. In some embodiments, the compositions of the invention prevent, reduce or alleviate social, occupational and functional impairment associated with anxiety disorders.

In some embodiments, the compositions of the invention improve the symptoms of anxiety disorders according to a symptomatic or diagnostic scale. In certain embodiments, the scale for assessing symptomatic improvement includes the Hamilton Anxiety Rating Scale (HAM-A). In some embodiments, the HAM-A total scale is used to assess primary endpoint. In other embodiments, the HAM-A psychic anxiety factor may be useful as a secondary endpoint.

In some embodiments, the compositions of the invention improve the Clinical Global Impression—Global Improvement (CGI-I) scale for assessing psychiatric and neurological disorders. In some embodiments, the compositions of the invention display a positive effect on global social, occupational and functional impairment of the subject with anxiety disorder. In some embodiments, the global scale is the Sheehan disability scale.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate at least one comorbidity of GAD and anxiety disorders. The comorbidities of GAD include depression; substance use disorders; stress; IBS; insomnia; headaches; pain; cardiac events; interpersonal problems and ADHD.

In some embodiments, the compositions of the invention are particularly effective at preventing, reducing or alleviating anxiety disorders when used in combination with another therapy for treating anxiety disorders. Such therapies include selective serotonin reuptake inhibitors (venlafaxine, duloxetine, escitalopram and paroxetine); benzodiazepines (alprazolam, lorazepam and clonazepam); pregabalin (Lyrica®) and gabapentin (Neurontin®); serotonin receptor partial agonists (buspirone and tandospirone); atypical serotonergic antidepressants (such as imipramine and clomipramine); monoamine oxidase inhibitors (MAOIs) (such as moclobemide and phenelzine); hydroxyzine; propranolol; clonidine; guanfacine and prazosin.

Post-Traumatic Stress Disorder (PTSD)

PTSD is a severe and disabling disorder, an essential feature of which is the inclusion of a traumatic event as a precipitating factor of this disorder.

The symptoms of PTSD are grouped into four main clusters according to the DMS-V criteria: (i) intrusion: examples include nightmares, unwanted thoughts of the traumatic events, flashbacks, and reacting to traumatic reminders with emotional distress or physiological reactivity; (ii) avoidance: examples include avoiding triggers for traumatic memories including places, conversations, or other reminders; (iii) negative alterations in cognitions and mood: examples include distorted blame of self or others for the traumatic event, negative beliefs about oneself or the world, persistent negative emotions (e.g., fear, guilt, shame), feeling alienated, and constricted affect (e.g., inability to experience positive emotions); (iv) alterations in arousal and reactivity: examples include angry, reckless, or self-destructive behaviour, sleep problems, concentration problems, increased startle response, and hypervigilance.

Symptoms that resolve within 4 weeks of the traumatic event meet the criteria for an Acute Stress Disorder. The DSM distinguishes between acute (duration of symptoms for less than three months) and chronic PTSD (duration of symptoms longer than 3 months). If the symptoms begin more than 6 months after the stressor, the disorder is defined as delayed onset PTSD.

PTSD carries high comorbidities with major depressive disorder and substance use disorders.

PTSD is a psychiatric disorder that may develop or persist due to dysfunction of the microbiota-gut-brain axis. Accordingly, in preferred embodiments, the compositions of the invention are for use in treating or preventing PTSD in a subject. According to a similar pathogenesis, in certain embodiments, the compositions of the invention are for use in treating or preventing stress disorders. In certain embodiments, the compositions of the invention treat acute stress disorder. In some embodiments, the compositions of the invention treat acute and/or chronic PTSD. In some embodiments, the compositions of the invention treat delayed onset PTSD.

In certain embodiments, the compositions of the invention prevent, reduce or alleviate one or more of the symptoms of PTSD (or stress disorder) in a subject as classified by the DMS-5 criteria listed herein. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate intrusive thoughts in a subject with PTSD. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate avoidance behaviour in a subject with PTSD. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate negative alterations in cognitions and mood in a subject with PTSD. In preferred embodiments, the compositions of the invention prevent alterations in arousal and reactivity in a subject with PTSD.

In some embodiments, the compositions of the invention improve the symptoms of PTSD and stress disorders according to a symptomatic or diagnostic scale. In certain embodiments, the scale for assessing symptomatic improvement is the Clinical-Administered PTSD (CAPS) scale.

In some embodiments, the compositions of the invention improve the Clinical Global Impression—Global Improvement (CGI-I) scale for assessing psychiatric and neurological disorders. In some embodiments, the compositions of the invention display a positive effect on global social, occupational and functional impairment of the subject with PTSD and stress disorders. In some embodiments, the global scale is the Sheehan disability scale.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate at least one comorbidity of PTSD and stress disorders. The comorbidities of PTSD and stress disorders include MDD, substance use disorders; stress and anxiety.

In some embodiments, the compositions of the invention are particularly effective at preventing, reducing or alleviating PTSD and stress disorders when used in combination with another therapy for treating PTSD and stress disorders. Such therapies include serotoninergic agents, tricyclic antidepressants, mood stabilisers, adrenergic inhibiting agents, antipsychotics, benzodiazepines, sertraline (Zoloft®), fluoxetine (Prozac®) and/or paroxetine (Paxil®).

Schizophrenia Spectrum and Psychotic Disorders

These diseases affect a subject's ability to think clearly, make good judgements, respond emotionally, communicate effectively, understand reality, and behave appropriately. Psychotic diseases include schizophrenia (symptoms listed below); schizoaffective disorder (the subject has symptoms of both schizophrenia and a mood disorder, such as depression or bipolar disorder); schizophreniform disorder (displays the symptoms of schizophrenia, but the symptoms last for a shorter time: between 1 and 6 months); brief psychotic disorder (subjects display a sudden, short period of psychotic behaviour, often in response to a very stressful event, such as a death in the family—recovery is usually less than a month); delusional disorder (delusions last for at least 1 month); shared psychotic disorder; substance-induced psychotic disorder; psychotic disorder due to another medical condition; paraphrenia (displaying symptoms similar to schizophrenia and starting late in life, when people are elderly). The most well-known psychotic disorder is schizophrenia and the majority of psychotic disorders display similar symptoms to schizophrenia.

Schizophrenia is a severe psychiatric disease with a heterogeneous course and symptom profile. Schizophrenia presents clinically with so-called positive and negative symptoms. The positive symptoms include delusions, hallucinations, disorganised speech, and disorganised or catatonic behaviours. Negative symptoms include affective flattening, restriction in the fluency and productivity of thought and speech and in the initiation of goal directed behaviour. The positive symptoms appear to reflect an excess or distortion of normal functions, whereas negative symptoms appear to reflect a diminution or loss of normal function. In addition, cognitive deficits (defects of working memory, information processing, attention/vigilance, learning, reasoning and social cognition) are common. Cognitive deficits generally show poor improvement with current antipsychotic treatment. Schizophrenic patients also suffer from mood symptoms. Besides these predominant symptoms, schizophrenia is associated with a comorbidity with other psychiatric symptoms such as manic and depressive symptoms, anxiety or obsessive-compulsive symptoms, substance abuse and dependence, and personality disorder.

According to the DMS-5, for the diagnosis of schizophrenia, a subject must have at least two of the following symptoms: delusions; hallucinations; disorganised speech; disorganised or catatonic behaviour and negative symptoms. At least one of the symptoms must be the presence of delusions, hallucinations or disorganised speech. Continuous signs of disturbance must persist for at least 6 months, during which the subject must experience at least 1 month of active symptoms, with social or occupational deterioration problems occurring over a significant amount of time.

Schizophrenia spectrum and psychotic disorders are psychiatric disorders that may develop or persist due to dysfunction of the microbiota-gut-brain axis. Therefore, in preferred embodiments, the compositions of the invention are for use in treating or preventing schizophrenia spectrum and/or psychotic disorders in a subject. In certain embodiments, the schizophrenia spectrum and psychotic disorder is selected from schizophrenia; schizoaffective disorder; schizophreniform disorder; brief psychotic disorder; delusional disorder; shared psychotic disorder; substance-induced psychotic disorder; psychotic disorder due to another medical condition and paraphrenia. In preferred embodiments, the compositions of the invention are for use in treating or preventing schizophrenia. In certain embodiments, the schizophrenia is selected from paranoid, disorganised, catatonic, undifferentiated and residual schizophrenia.

In certain embodiments, the compositions of the invention prevent, reduce or alleviate one or more of the symptoms of schizophrenia in a subject as classified by the DMS-5 criteria listed herein. These embodiments apply to the prevention, reduction or alleviation of symptoms of other schizophrenia spectrum and psychotic disorders. In certain embodiments, the compositions of the invention prevent, reduce or alleviate negative symptoms of schizophrenia. In certain embodiments, the compositions of the invention prevent, reduce or alleviate positive symptoms of schizophrenia. In certain embodiments, the compositions of the invention prevent, reduce or alleviate negative and positive symptoms of schizophrenia. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate delusions, hallucinations, disorganised speech, and disorganised or catatonic behaviours in a subject with schizophrenia. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate affective flattening, restriction in the fluency and productivity of thought and speech and in the initiation of goal directed behaviour in a subject with schizophrenia. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate the cognitive defects and/or mood disorders in a subject with schizophrenia.

In certain embodiments, the compositions of the invention reduce the occurrence of positive and/or negative symptoms of schizophrenia in a subject within a 6 month period. In certain embodiments, the compositions of the invention improve social and/or occupational functionality in a subject with schizophrenia spectrum or psychotic disorder.

In some embodiments, the compositions of the invention improve the symptoms of schizophrenia spectrum or psychotic disorders according to a symptomatic or diagnostic scale. In certain embodiments, the scale for assessing symptomatic improvement is the Positive and Negative Symptom Scale (PANSS) and Brief Psychiatric Rating Scale (BPRS). In certain embodiments, the Scale for Assessment of Negative Symptoms (SANS) is used.

In some embodiments, the compositions of the invention improve the Clinical Global Impression—Global Improvement (CGI-I) scale for assessing psychiatric and neurological disorders. In some embodiments, the compositions of the invention display a positive effect on global social and occupational impairment of the subject with schizophrenia spectrum or psychotic disorders.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate at least one comorbidity of schizophrenia spectrum or psychotic disorder. In certain embodiments, the comorbidity is as manic and depressive symptoms, anxiety or obsessive-compulsive symptoms, substance abuse and dependence, and personality disorder.

In certain embodiments, the compositions of the invention are for use in treating or preventing treatment resistant of refractory schizophrenia.

In some embodiments, the compositions of the invention are particularly effective at preventing, reducing or alleviating schizophrenia spectrum or psychotic disorders when used in combination with another therapy for treating PTSD and stress disorders. In certain embodiments, such therapies include first generation antipsychotics including chlorpromazine, fluphenazine, haloperidol and/or perphenazine. In certain embodiments, such therapies include second generation therapies including aripiprazole (Abilify®); asenapine (Saphris®); brexpiprazole (Rexulti®); cariprazine (Vraylar®); clozapine (Clozaril®); iloperidone (Fanapt®); lurasidone (Latuda®); olanzapine (Zyprexa®); paliperidone (Invega); quetiapine (Seroquel®); risperidone (Risperdal®); ziprasidone (Geodon®).

Bipolar Disorder

Bipolar disorder in general is a chronic disease. Mania is the cardinal symptom of bipolar disorder. There are several types of bipolar disorder based upon the specific duration and pattern of manic and depressive episodes. In DMS-5, a distinction is made between bipolar I disorder, bipolar II disorder, cyclothymic disorder, rapid-cycling bipolar disorder and bipolar disorder NOS.

According to the DSM, mania is a distinct period of abnormally and persistently elevated, expansive, or irritable mood. The episode must last a week, and the mood must have at least three of the following symptoms: high self-esteem; reduced need for sleep; increase rate of speech; rapid jumping of ideas; easily distracted; an increased interest in goals or activities; psychomotor agitation; increased pursuit of activities with a high risk of danger.

Bipolar I disorder involves one or more manic or mixed (mania and depression) episodes and at least one major depressive episode (see above for symptoms of MDD episodes). Bipolar II disorder has one or more major depressive episodes accompanied by at least one hypomanic episode. There are no manic or mixed episodes. Hypomania is a lesser form of mania. The symptoms are responsible for significant social, occupational and functional impairments. Cyclothymia is characterized by changing low-level depression along with periods of hypomania. The symptoms must be present for at least two years in adults or one year in children before a diagnosis can be made. Symptom free periods in adults and children last no longer than two months or one month, respectively. Rapid cycling bipolar disorder is a severe form of bipolar disorder. It occurs when a person has at least four episodes of major depression, mania, hypomania, or mixed states within a year. Not-otherwise specified (NOS) bipolar disorder classified bipolar symptoms that do not clearly fit into other types. NOS is diagnosed when multiple bipolar symptoms are present but not enough to meet the label for any of the other subtypes.

Bipolar disorder is associated with the following comorbidities: ADHD; anxiety disorders; substance disorders; obesity and metabolic syndrome.

Bipolar disorder is a psychiatric disorder that may develop or persist due to dysfunction of the microbiota-gut-brain axis. Therefore, in preferred embodiments, the compositions of the invention are for use in treating or preventing bipolar disorder in a subject. In certain embodiments, the bipolar disorder is bipolar I disorder. In certain embodiments, the bipolar disorder is bipolar II disorder. In certain embodiments, the bipolar disorder is cyclothymic disorder. In certain embodiments, the bipolar disorder is rapid-cycling bipolar disorder. In certain embodiments, the bipolar disorder is bipolar disorder NOS.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate one or more of the symptoms of bipolar disorder in a subject. In certain embodiments, the compositions of the invention prevent, reduce or alleviate the occurrence of manic episodes in a subject. In certain embodiments, the compositions of the invention prevent, reduce or alleviate the occurrence of an abnormally and persistently elevated, expansive, or irritable mood. In certain embodiments, the compositions of the invention prevent, reduce or alleviate one or more of the following symptoms: high self-esteem; reduced need for sleep; increase rate of speech; rapid jumping of ideas; easily distracted; an increased interest in goals or activities; psychomotor agitation; increased pursuit of activities with a high risk of danger. In certain embodiments, the compositions of the invention prevent, reduce or alleviate the occurrence of one or more manic or mixed episodes in a subject. In certain embodiments, the compositions of the invention reduce the occurrence of at least one major depressive episode in a subject. In certain embodiments, the compositions of the invention prevent, reduce or alleviate the occurrence of at least one major depressive episode accompanied by at least one hypomanic episode.

In preferred embodiments, the compositions of the invention treat the acute phase of bipolar disorder and/or prevent the occurrence of further episodes. In certain embodiments, the compositions of the invention treat the acute phase of manic/depressive episodes in a subject with bipolar disorder and prevent occurrence of further manic/depressive episodes.

In some embodiments, the compositions of the invention improve the symptoms of bipolar disorder according to a symptomatic or diagnostic scale. In certain embodiments, the scale for assessing symptomatic improvement of manic episodes is the Manic State Rating Scale and the Young Mania Rating Scale. In certain embodiments, the scale is the Bech-Rafaelsen Mania Scale (BRMAS). In certain embodiments, scales for assessing symptomatic improvement of the switch from manic to depressive episodes include the Hamilton Depression Rating Scale, the Montgomery-Asberg Rating Scale, and the Bech-Rafaelsen Depression Scale.

In some embodiments, the compositions of the invention improve the Clinical Global Impression—Global Improvement (CGI-I) scale for assessing psychiatric and neurological disorders. In some embodiments, the compositions of the invention display a positive effect on global social, occupational and functional impairments of the subject with bipolar disorder.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate at least one comorbidity of bipolar disorder. In certain embodiments, the comorbidity is selected from ADHD, anxiety disorders, substance disorder, obesity and metabolic syndrome.

In certain embodiments, the compositions of the invention are for use in treating or preventing manic-depressive illness and bipolar disorder unresponsive to lithium and divalproex.

In some embodiments, the compositions of the invention are particularly effective at preventing, reducing or alleviating bipolar disorder when used in combination with another therapy for treating bipolar disorder. In certain embodiments, such therapies include lithium carbonate, anticonvulsant drugs (including valproate, divalproex, carbamazepine and lamotrigine) and antipsychotic drugs (including aripiprazole, olanzapine, quetiapine and risperidone).

Neurocognitive Disorders and Alzheimer's Disease

In DSM-5, the term dementia was replaced with the terms major neurocognitive disorder and mild neurocognitive disorder. Neurocognitive disorder is a heterogeneous class of psychiatric diseases. The most common neurocognitive disorder is Alzheimer's disease, followed by vascular dementias or mixed forms of the two. Other forms of neurodegenerative disorders (e.g. Lewy body disease, frontotemporal dementia, Parkinson's dementia, Creutzfeldt-Jakob disease, Huntington's disease, and Wernicke-Korsakoff syndrome) are accompanied by dementia.

The symptomatic criteria for dementia under DSM-5 are evidence of significant cognitive decline from a previous level of performance in one or more cognitive domains selected from: learning and memory; language; executive function; complex attention; perceptual-motor and social cognition. The cognitive deficits must interfere with independence in everyday activities. In addition, the cognitive deficits do not occur exclusively in the context of a delirium and are not better explained by another mental disorder (for example MDD or schizophrenia).

In addition to the primary symptom, subjects with neurocognitive disorders display behavioural and psychiatric symptoms including agitation, aggression, depression, anxiety, apathy, psychosis and sleep-wake cycle disturbances.

Neurocognitive disorders are psychiatric disorders that may develop or persist due to dysfunction of the microbiota-gut-brain axis. Therefore, in preferred embodiments, the compositions of the invention are for use in treating or preventing neurocognitive disorders in a subject. In preferred embodiments, the neurocognitive disorder is Alzheimer's disease. In other embodiments, the neurocognitive disorder is selected from vascular dementias; mixed form Alzheimer's disease and vascular dementia; Lewy body disease; frontotemporal dementia; Parkinson's dementia; Creutzfeldt-Jakob disease; Huntington's disease; and Wernicke-Korsakoff syndrome.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate one or more of the symptoms of neurocognitive disorders in a subject. In certain embodiments, the compositions of the invention prevent, reduce or alleviate the occurrence of cognitive decline in a subject. In certain embodiments, the compositions of the invention improve the level of performance of a subject with neurocognitive disorders in one or more cognitive domains selected from: learning and memory; language; executive function; complex attention; perceptual-motor and social cognition. In some embodiments, the compositions of the invention prevent, reduce or alleviate the occurrence of one or more behavioural and psychiatric symptoms associated with neurocognitive disorders selected from agitation, aggression, depression, anxiety, apathy, psychosis and sleep-wake cycle disturbances.

In certain embodiments, the compositions of the invention prevent, reduce or alleviate symptomatic disease by intervention in suspected pathogenic mechanisms at a preclinical stage. In certain embodiments, the compositions of the invention improve disease modification, with slowing or arrest of symptom progression. In some embodiments, the slowing or arrest of symptom progression correlates with evidence in delaying the underlying neuropathological process. In preferred embodiments, the compositions of the invention improve symptoms of neurocognitive disorders comprising enhanced cognitive and functional improvement. In preferred embodiments, the compositions of the invention improve the behavioural and psychiatric symptoms of dementia (BPSD). In preferred embodiments, the compositions of the invention improve the ability of a subject with neurocognitive disorder to undertake everyday activities.

In preferred embodiments, the compositions of the invention improve both cognition and functioning in a subject with Alzheimer's disease. In some embodiments, the composition of the invention improve the cognitive endpoint in a subject with Alzheimer's disease. In some embodiments, the compositions of the invention improve the functional endpoint in a subject with Alzheimer's disease. In preferred embodiments, the compositions of the invention improve the cognitive and functional endpoint in a subject with Alzheimer's disease. In yet further preferred embodiments, the compositions of the invention improve the overall clinical response (the global endpoint) in a subject with Alzheimer's disease.

In some embodiments, the compositions of the invention improve the symptoms of neurocognitive disorders according to a symptomatic or diagnostic test. In certain embodiments, the tests for assessing symptomatic improvement of Alzheimer's disease (and other neurocognitive disorders) are selected from objective cognitive, activities of daily living, global assessment of change, health related quality of life tests and tests assessing behavioural and psychiatric symptoms of neurocognitive disorders.

In certain embodiments, the objective cognitive tests for assessment of symptomatic improvement use the Alzheimer's disease Assessment Scale cognitive subscale (ADAS-cog) and the classic ADAS scale. In certain embodiments, symptomatic improvement of cognition is assessed using the Neurophysiological Test Battery for Use in Alzheimer's Disease (NTB).

In some embodiments, the global assessment of change test uses the Clinical Global Impression—Global Improvement (CGI-I) scale for assessing psychiatric and neurological disorders. In some embodiments, the global scale is the Clinician's Interview Based Impression of Change plus (CIBIC-plus). In some embodiments, the global scale is the Alzheimer's Disease Cooperative Study Unit Clinician's Global Impression of Change (ADCS-CGIC).

In certain embodiments, the health related quality of life measures are the Alzheimer's Disease-Related QOL (ADRQL) and the QOL-Alzheimer's Disease (QOL-AD).

In certain embodiments, the tests assessing behavioural and psychiatric symptoms of neurocognitive disorders are selected from the Behavioural pathology in Alzheimer's Disease Rating Scale (BEHAVE-AD); the Behavioural Rating Scale for Dementia (BRSD); the Neuropsychiatric Inventory (NPI); and the Cohen-Mansfield Agitation Inventory (CMAI).

In some embodiments, the compositions of the invention are particularly effective at preventing, reducing or alleviating neurocognitive disorders when used in combination with another therapy for treating neurocognitive disorders. In certain embodiments, such therapies include acetylcholinesterase inhibitors including donepezil (Aricept®), galantamine (Razadyne®) and rivastigmine (Exelon®), and memantine.

Parkinson's Disease

Parkinson's disease is a common neurodegenerative disease neuropathologically characterised by degeneration of heterogeneous populations of neural cells (dopamine-producing cells). The clinical diagnosis of Parkinson's disease requires bradykinesia and at least one of the following core symptoms: resting tremor; muscle rigidity and postural reflex impairment. Other signs and symptoms that may be present or develop during the progression of the disease are autonomic disturbances (sialorrhoea, seborrhoea, constipation, micturition disturbances, sexual functioning, orthostatic hypotension, hyperhydrosis), sleep disturbances and disturbances in the sense of smell or sense of temperature. Depressive symptoms and cognitive dysfunction comorbidities develop in many Parkinson's disease patients, as well as neurocognitive disorders related to Lewy Bodies.

Parkinson's disease is a psychiatric disorder that may develop or persist due to dysfunction of the microbiota-gut-brain axis. Therefore, in preferred embodiments, the compositions of the invention are for use in treating or preventing Parkinson's disease in a subject.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate one or more of the symptoms of Parkinson's disease in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate one or more core symptoms of Parkinson's disease in a subject. In certain embodiments, the compositions of the invention prevent, reduce or alleviate bradykinesia in a subject. In certain embodiments, the compositions of the invention prevent, reduce or alleviate resting tremor; muscle rigidity and/or postural reflex impairment in a subject. In certain embodiments, the compositions of the invention prevent, reduce or alleviate one or more symptoms associated with Parkinson's disease progression selected from autonomic disturbances (sialorrhoea, seborrhoea, constipation, micturition disturbances, sexual functioning, orthostatic hypotension, hyperhydrosis), sleep disturbances and disturbances in the sense of smell or sense of temperature.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate depressive symptoms comorbid with Parkinson's disease. In certain embodiments, the compositions of the invention improve verbal memory and/or executive functions. In certain embodiments, the compositions of the invention improve attention, working memory, verbal fluency and/or anxiety.

In other preferred embodiments, the compositions of the invention prevent, reduce or alleviate cognitive dysfunctions comorbid with Parkinson's disease.

In certain embodiments, the compositions of the invention prevent, reduce or alleviate Parkinson's disease progression. In certain embodiments, the compositions of the invention prevent, reduce or alleviate later motor complications. In certain embodiments, the compositions of the invention prevent, reduce or alleviate late motor fluctuations. In certain embodiments, the compositions of the invention prevent, reduce or alleviate neuronal loss. In certain embodiments, the compositions of the invention improve symptoms of Parkinson's disease dementia (PDD). In certain embodiments, the compositions of the invention prevent, reduce or alleviate impairment of executive function, attention and/or working memory. In certain embodiments, the compositions of the invention improve dopaminergic neurotransmission. In certain embodiments, the compositions of the invention prevent, reduce or alleviate impaired dopaminergic neurotransmission.

In some embodiments, the compositions of the invention improve the symptoms of Parkinson's disease according to a symptomatic or diagnostic scale. In certain embodiments, the tests for assessing symptomatic improvement of motor function in Parkinson's disease is the Unified Parkinson's Disease Rating Scale. In particular, UPDRS II considers the activity of daily life and UPDRS III considers motor-examination.

In some embodiments, the compositions of the invention improve the symptoms associated the PDD according to a symptomatic or diagnostic test and/or scale. In certain embodiments, the test or scale is selected from the Hopkins Verbal Learning Test—Revised (HVLT-R); the Delis-Kaplan Executive Function System (D-KEFS) Color-Word Interference Test; the Hamilton Depression Rating Scale (HAM-D 17; depression); the Hamilton Anxiety Rating Scale (HAM-A; anxiety) and the Unified Parkinson's Disease Rating Scale (UPDRS; PD symptom severity).

In some embodiments, the compositions of the invention improve the Clinical Global Impression—Global Improvement (CGI-I) scale for assessing psychiatric and neurological disorders. In some embodiments, the compositions of the invention display a positive effect on global social and occupational impairment of the subject with Parkinson's disease.

In some embodiments, the compositions of the invention are particularly effective at preventing, reducing or alleviating neurocognitive disorders when used in combination with another therapy for treating neurocognitive disorders. In certain embodiments, such therapies include dopamine agonists (including L-Dopa+); monoamine oxidase inhibitors, catecholamine-O-methyl transferase inhibitors; anticholinergics and glutamate modulators.

Other Central Nervous System Disorders

In preferred embodiments, the compositions of the invention are for use in treating or preventing a central nervous system disorder associated with dysfunction of the microbiota-gut-brain axis. In addition to the embodiments above, the compositions of the invention are for use in treating or preventing psychosis; chronic fatigue syndrome (myalgic encephalomyelitis) and/or chronic pain. In further embodiments, the compositions of the invention may be useful for treating or preventing motor neuron disease; Huntington's disease; Guillain-Barre syndrome and/or meningitis.

Huntington's Disease

Huntington's disease is an inherited brain condition, caused by an inherited faulty gene, which damages certain nerve cells in the brain. This brain damage gets progressively worse over time and can affect movement, cognition (perception, awareness, thinking, judgement) and behaviour. Early features of the disease can include personality changes, mood swings, fidgety movements, irritability and altered behaviour.

In certain embodiments, the compositions of the invention are for use in treating or preventing Huntington's disease. In certain embodiments, the compositions of the invention manage the symptoms of Huntington's disease, such as irritability or excessive movement. In certain embodiments, the compositions of the invention treat the depression associated with Huntington's disease and/or improve symptoms such as social withdrawal, lack or interest and sleep disturbance. In certain embodiments, the compositions of the invention improve memory and ability to concentrate on tasks. In certain embodiments, the compositions of the invention treat disabling abnormal movements. In certain embodiments, the compositions of the invention treat behavioural problems, antisocial behaviour, irritability and psychosis associated with Huntington's disease. In certain embodiments, the compositions of the invention induce neuroprotection and prevent nerve damage. In certain embodiments, the compositions of the invention increase the levels of dopamine and/or the levels of dopamine-containing cells.

Neurochemical Factors, Neuropeptides and Neurotransmitters and the Microbiota-Gut-Brain Axis

As outlined above, the microbiota-gut-brain axis is modulated by a number of different physiological systems. The microbiota-gut-brain axis is modulated by a number of signalling molecules. Alterations in the levels of these signalling molecules results in defects in central nervous system development and/or functionality. Indeed, many of the molecules disclosed in this section have been implicated in the functionality of the microbiota-gut-brain axis and the pathogenesis of central nervous system disorders or conditions ([14], [32], [10], [33]). The experiments performed by the inventors indicate that behavioural changes can be triggered by administration of Blautia strains. This effect may be mediated by an effect on levels of the signalling molecules, in particular those listed in this section. These alterations may be responsible for the therapeutic benefits associated with Blautia strains. Accordingly, due to the fact that the central nervous system disorders and conditions disclosed herein display a similar fundamental biochemical and physiological pathogenesis (i.e. via the microbiota-gut-brain axis), a similar therapeutic benefit of Blautia strains may be also achieved for these disorders and conditions. Administration of Blautia stercoris may be particularly effective for triggering behavioural changes associated with central nervous system disorders or conditions. In certain embodiments, administration of Blautia wexlerae may be particularly effective for triggering behavioural changes associated with central nervous system disorders or conditions.

The signalling of the microbiota-gut-brain axis is modulated by levels of neurochemical factors, neuropeptides and neurotransmitters. Accordingly, in certain embodiments, the compositions of the invention modulates levels of neurochemical factors, neuropeptides and neurotransmitters. Accordingly, in certain preferred embodiments, the compositions of the invention directly alter CNS biochemistry. In preferred embodiments, the compositions of the invention modulate the levels of brain-derived neurotrophic factor (BDNF). In certain embodiments, the compositions of the invention modulate the levels of monoamines. In certain embodiments, the monoamines are serotonin (5-hydroxytryptamine (5-HT)), dopamine, norepinephrine and/or epinephrine. In certain embodiments, the monoamines are catecholamines. In certain embodiments, the catecholamines are dopamine, norepinephrine and epinephrine. In certain embodiments, the monoamines are tryptamines. In certain embodiments, the tryptamines are serotonin and melatonin. In certain embodiments, the compositions of the invention modulate the levels of acetylcholine.

In certain preferred embodiments, the compositions of the invention modulate the levels of oxytoxin. Oxytocin is associated with emotional, social, cognitive and neuroendocrine physiologies as well as autoregulation. In particular, oxytocin release is involved in anxiolysis; positive mood; maternal behaviour, pair bonding; sexual behaviour; social memory; olfactory memory; anorexiant effects; attenuation of the HPA axis response to stress; autoexcitation during birth and suckling as well as other physiological and psychological processes. In certain embodiments, the compositions of the invention increase the levels of oxytocin. In certain embodiments, the compositions of the invention decrease the levels of oxytocin. In certain embodiments, the compositions of the invention increase or decrease oxytocin signalling. In certain embodiments, the compositions of the invention modulate the levels of oxytocin receptors. In certain embodiments, the compositions of the invention modulate the flux of calcium ions into or out of neuronal, muscle and gastrointestinal cells. In preferred embodiments, the compositions of the invention treat and prevent neurodevelopmental and neuropsychiatric disorders and diseases associated with the microbiota-gut-brain axis by modulating the levels of oxytocin.

In certain embodiments, the compositions of the invention modulate the levels of brain monoamines and metabolites thereof. In preferred embodiments, the monoamine is serotonin. In certain embodiments, the compositions of the invention modulate the serotonergic and/or kynurenine routes of tryptophan metabolism. In certain embodiments, the compositions of the invention modulate the levels of serotonin metabolites, such as 5-Hydroxyindoleacetic acid (5-HIAA). In certain embodiments, the compositions of the invention modulate the levels of dopamine metabolites, such as Homovanillic acid (HVA). Modulation of these neurotransmitters and neurochemical factors is useful for treating stress, depression and anxiety-related disorders.

The signalling of the microbiota-gut-brain axis is modulated by levels of γ-aminobutyric acid (GABA). Accordingly, in preferred embodiments, the compositions of the invention modulate the levels of GABA. GABA is an inhibitory neurotransmitter that reduces neuronal excitability. In certain embodiments, the compositions of the invention increase the levels of GABA. In certain embodiments, the compositions of the invention decrease the levels of GABA. In certain embodiments, the compositions of the invention alter GABAergic neurotransmission. In certain embodiments, the compositions of the invention modulate the level of GABA transcription in different regions of the central nervous system. In certain embodiments, the commensal derived GABA crosses the blood-brain barrier and affects neurotransmission directly. In certain embodiments, the compositions of the invention lead to a reduction of GABA in the hippocampus, amygdala and/or locus coeruleus. In certain embodiments, the compositions of the invention lead to an increase of GABA in cortical regions.

The levels of neuroactive molecules, such as serotonin, melatonin, GABA, histamines and acetylcholine are linked to the pathophysiology of central nervous system diseases such as dementia, Alzheimer's disease and Huntington's disease.

The signalling of the microbiota-gut-brain axis is modulated by levels of histamines. Accordingly, in certain embodiments, the compositions of the invention modulate the levels of histamines. In certain embodiments, the histamines has an immunoregulatory effect. In certain embodiments, histamine levels enable translocation of bacteria from the lumen into systemic circulation. Therefore, in some embodiments, the compositions of the invention alter gastrointestinal tract permeability and/or barrier function. In certain other embodiments, the histamine acts as a neurotransmitter linked to central processes.

The signalling of the microbiota-gut-brain axis is modulated by the HPA axis. Accordingly, in certain embodiments, the compositions of the invention modulate HPA activity. In certain embodiments, the compositions of the invention attenuate the HPA stress response. In certain preferred embodiments, the compositions of the invention modulate inflammatory responses associated with HPA activity. In certain embodiments, the compositions of the invention modulate the levels of glucocorticoids. In certain preferred embodiments, the compositions of the invention modulate the levels of corticosterone and adrenaline. In certain embodiments, the compositions of the invention modulate the levels of corticotrophin-releasing factor and/or vasopressin. In certain embodiments, the compositions of the invention modulate the levels of vasopressin and/or other neurohypophysial or antidiuretic hormones.

Alterations in HPA axis activity are associated with anxiety and stress disorders. The signalling of the microbiota-gut-brain axis is modulated by alterations in the immune response and inflammatory factors and markers. Accordingly, in certain embodiments, the compositions of the invention may modulate the immune response. In certain embodiments, the compositions of the invention modulate the systemic levels of circulating neuroimmune signalling molecules. In certain preferred embodiments, the compositions of the invention modulate pro-inflammatory cytokine production and inflammation. In certain embodiments, the compositions of the invention modulate the inflammatory state. In certain embodiments, the compositions of the invention modulate the splenocyte proliferative response. In certain embodiments, the compositions of the invention modulate the systemic and/or plasma levels of C-reactive protein; IL-1 family cytokines; IL-1β; IL-2; IL-4; IL-6; IL-8; IL-10; IL-12p40; IL-17; IL-17A; IL-21; IL-23; TNF-α and IFN-γ. In some embodiments the compositions of the invention module the levels of anti-inflammatory cytokines, for example IL-10. In preferred embodiments, the compositions of the invention increase the levels of IL-10. In some embodiments, the compositions of the invention modulate the levels of TNF-α. In preferred embodiments, the compositions of the invention modulate the levels of IFN-γ. In some embodiments, the compositions of the invention modulate the IFN-γ:IL-10 ratio. In certain preferred embodiments, the compositions of the invention decrease the IFN-γ:IL-10 ratio. In preferred embodiments, the compositions of the invention decrease the levels of the pro-inflammatory cytokines TNF-α and IFN-γ. Increased circulating levels of cytokines are closely associated with various neuropsychiatric disorders, including depression, anxiety, schizophrenia and ASD. Evidence of inflammatory state alteration is highlighted in disorders such as schizophrenia, major depressive disorder and bipolar disorder.

In certain embodiments, the compositions of the invention modulates the levels of tolerance-mediating dendritic cells and reciprocally regulate pro and anti-inflammatory cytokine responses. In certain embodiments, the compositions of the invention decrease the systemic level of myeloperoxidase (a marker for inflammation and oxidation). Therapeutic modulators of the immune system and of inflammatory responses are useful for treating autism spectrum disorders and mood disorders.

In certain embodiments, the compositions of the invention modulate the immune response to an infection or vaccination. In certain embodiments, the compositions of the invention modulate the level of inflammation in response to infection or vaccination. In certain preferred embodiments, the compositions of the invention modulate maternal immune activation in response to an infection or vaccination during pregnancy. Accordingly, the compositions of the invention can be administered during pregnancy in order to treat or prevent a central nervous system disorder in the offspring.

The signalling of the microbiota-gut-brain axis is modulated by levels commensal metabolites. Accordingly, in certain embodiments, the compositions of the invention modulate the systemic levels of microbiota metabolites. In certain preferred embodiments, the compositions of the invention modulate the level of short chain fatty acids (SCFAs). In certain embodiments the level of SCFAs is increased or decreased. In some embodiments, the SCFA is butyric acid (BA) (or butyrate). In some embodiments, the SCFA is propionic acid (PPA). In some embodiments, the SCFA is acetic acid. In certain embodiments, the compositions of the invention modulate the ability of SCFAs to cross the blood-brain barrier. In certain embodiments, the compositions of the invention modulate the level of Polysaccharide A (PSA). In certain embodiments, the compositions of the invention modulate the levels of the potent pro-inflammatory endotoxin lipopolysaccharide (LPS). LPS leads to the production of inflammatory cytokines that alter physiological brain activity and modulate neuropeptide synthesis. LPS has an important influence on the modulation of the CNS, increasing the activity of areas devoted to the control of emotions (e.g. the amygdala). In certain embodiments, the compositions of the invention modulate the level of tryptophan and/or its metabolites. In certain embodiments, the compositions of the invention modulate the levels of 4-ethylphenylsulphate (4EPS; a uremic toxic associated with ASD-related behavioural abnormalities). In preferred embodiments, the compositions of the invention decrease the levels of 4-ethylphenylsulphate in a subject. The signals generated by the stimulation of neuronal signalling pathways caused by intraluminal gut stimuli strongly modulate brain activity, including pain perception, immune-response modulation, emotional control and other homeostatic functions. Accordingly, a composition able to modulate levels of these factors would have broad therapeutic applications for treating or preventing CNS disorders.

The signalling of the microbiota-gut-brain axis is modulated by levels gastrointestinal permeability. Accordingly, in some embodiments, the compositions of the invention alter the integrity of the gastrointestinal tract epithelium. In certain embodiments, the compositions of the invention modulate the permeability of the gastrointestinal tract. In certain embodiments, the compositions of the invention modulate the barrier function and integrity of the gastrointestinal tract. In certain embodiments, the compositions of the invention modulate gastrointestinal tract motility. In certain embodiments, the compositions of the invention modulate the translocation of commensal metabolites and inflammatory signalling molecules into the bloodstream from the gastrointestinal tract lumen.

The signalling of the microbiota-gut-brain axis is modulated by microbiome composition in the gastrointestinal tract. Accordingly, in certain embodiments, the compositions of the invention modulates the microbiome composition of the gastrointestinal tract. In certain embodiments, the compositions of the invention prevents microbiome dysbiosis and associated increases in toxic metabolites (e.g. LPS). In certain embodiments, the compositions of the invention modulate the levels of Clostridium in the gastrointestinal tract. In preferred embodiments, the compositions of the invention reduce the level of Clostridium in the gastrointestinal tract. In certain embodiments, the compositions of the invention reduce the levels of Campylobacter jejuni. In certain embodiments, the compositions of the invention modulate the proliferation of harmful anaerobic bacteria and the production of neurotoxins produced by these bacteria. In certain embodiments, the compositions of the invention modulate the microbiome levels of Lactobacillus and/or Bifidobacterium. In certain embodiments, the compositions of the invention modulate the microbiome levels of Sutterella, Prevotella, Ruminococcus genera and/or the Alcaligenaceae family. In certain embodiments, the compositions of the invention increase the level of Lactobacillus plantarum and/or Saccharomyces boulardii.

In certain embodiments, the compositions of the invention prevent the dysregulation of the composition of the microbiome by extensive antibiotic use. In certain preferred embodiments, the compositions of the invention maintain a functional maternal microbiome composition upon administration of antibiotics during pregnancy. Accordingly, the compositions of the invention can be administered during pregnancy in order to treat or prevent a central nervous system disorder in the offspring.

Modulation of the microbiome has been shown to be effective at improving psychiatric disorder-related behaviours, including anxiety, depression, autism spectrum disorder, obsessive-compulsive disorder and memory abilities (including spatial and non-spatial memory), as well as other CNS-related disorders including Parkinson's disease. Certain studies have suggested that probiotics can reduce psychological stress, somatisation, depression and anger-hostility. The levels of Lactobacillus are associated with depression and have been implicated in pain signalling associated with gastrointestinal discomfort.

In certain embodiments, the compositions of the invention prevent, reduce or alleviate at least one of the behavioural symptoms associated with a central nervous system disorder described herein. In preferred embodiments, the compositions of the invention improve the overall clinical response in a subject.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate stereotyped, repetitive behaviour in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate the occurrence of unusually restrictive behaviours and/or interests. In certain embodiments, the compositions of the invention prevent, reduce or alleviate recurrent obsessions and/or compulsions in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate deficits in social behaviour in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate avoidance behaviour in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate deficits in communication behaviour in a subject.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate negative alterations in cognitions and mood in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate anxiety-related behaviour in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate stress-related behaviour in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate depression-related behaviour in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate aggressive behaviour in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate the occurrence of an abnormally and persistently elevated, expansive, or irritable mood.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate intrusive thoughts in a subject. In preferred embodiments, the compositions of the invention prevent alterations in arousal and reactivity in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate delusions, hallucinations, disorganised speech, and disorganised or catatonic behaviours in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate affective flattening, restriction in the fluency and productivity of thought and speech and in the initiation of goal directed behaviour in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate one or more of the following symptoms: high self-esteem; reduced need for sleep; increase rate of speech; rapid jumping of ideas; easily distracted; an increased interest in goals or activities; psychomotor agitation; increased pursuit of activities with a high risk of danger.

In preferred embodiments, the compositions of the invention improve spatial and/or non-spatial memory deficits in a subject. In preferred embodiments, the compositions of the invention improve both cognition and functioning in a subject. In preferred embodiments, the compositions of the invention improve locomotor activity in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate bradykinesia in a subject. In preferred embodiments, the compositions of the invention prevent, reduce or alleviate resting tremor; muscle rigidity and/or postural reflex impairment in a subject.

In preferred embodiments, the compositions of the invention prevent, reduce or alleviate at least one comorbidity associated with a CNS disorder disclosed herein.

In preferred embodiments, the compositions of the invention improve the scores of a subject on at least one of the symptomatic and/or diagnostic scales for CNS disorders described herein. In certain other embodiments, the symptomatic and/or diagnostic scale is selected from the General Health Questionnaire (GHQ); the Depression Anxiety and Stress Scale (DASS); the Leiden Index of Depression Sensitivity-Revised (LEIDS-r); the Positive and Negative Symptom Scale (PANSS); the State-Trait Anxiety Inventory (STAI); the Development Behavior Checklist (DBC); the Beck Depression Inventory (BDI); the Beck Anxiety Inventory (BAI); the Hopkins Symptom Checklist (HSCL-90); the Hospital Anxiety and Depression Scale (HADS); the Perceived Stress Scale (PSS); the Coping Checklist (CCL) (also used to counter the stress of daily life); and the questionnaire-based Profile of Mood State (POMS).

In certain embodiments, the compositions of the invention may improve the symptomatic and/or diagnostic scale when assessing therapeutic efficacy in other animal models of CNS disorders known to a person skilled in the art. In addition to the behavioural assays disclosed in the examples, the compositions of the invention may improve reciprocal social interactions; olfactory communication; ultrasonic vocalisation; motor stereotypes (such as circling and vertical jumping), repetitive behaviour such as self-grooming and diffing; and perseverance in spatial tasks.

In addition, the compositions of the invention will be useful in treating and/or preventing CNS disorders in other animal models of CNS disorders. Other mouse models include inbred mice strains (including BALB/cJ and C58/J) and also genetically modified mice strains (including NEUREXIN1, NEUROLIGIN3, NEUROLIGIN4, SHANK2, SHANK3, CNTNAP2, Tsc1/2 and Fmr1 gene mutant mice strains).

Butyrate is a short chain fatty acid that acts as a histone deacetylase inhibitor, is capable of signalling through G-protein coupled receptors and is implicated in the regulation of metabolic pathways.

The Examples demonstrate that Blautia hydrogenotrophica increases the intestinal levels of butyrate when administered in oral compositions. The Examples also demonstrate that Blautia hydrogenotrophica is useful for treating central nervous system disorders and conditions. This effect of Blautia hydrogenotrophica may be mediated by butyrate.

Butyrate has been linked to histone deacetylation in the hippocampus and frontal cortex of the brain [34] and has been implicated in Huntington's disease, Parkinson's disease, Alzheimer's disease and autism [35].

In certain embodiments, the Blautia hydrogenotrophica strain for use in the invention is a butyrate producer. In certain embodiments, the Blautia hydrogenotrophica strain for use in the invention synthesise butyrate by the acetyl-CoA, glutarate, 4-aminobutyrate and/or lysine pathways. In certain embodiments, the Blautia hydrogenotrophica strain for use in the invention metabolises complex polysaccharides (e.g. starch and xylan) to produce acetyl-CoA, which can subsequently be used to synthesise butyrate. In certain embodiments, the Blautia hydrogenotrophica strain for use in the invention produces butyrate by bacterial fermentation in the colon.

In certain preferred embodiments, the compositions of the invention comprising Blautia hydrogenotrophica modulate the levels of butyrate. In certain embodiments, compositions of the invention comprising Blautia hydrogenotrophica increase the levels of butyrate.

In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica is a histone deacetylase (HDAC) inhibitor. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica inhibits histone deacetylation in the hippocampus and frontal cortex of the brain. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica increases histone acetylation and promotes the expression of pro-survival, pro-regenerative and pro-plasticity genes. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica rescues histone acetylation, prevents neuronal cell death and extends lifespan (for example in Huntington's disease). In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica protects neurones from cell death (for example in Parkinson's disease). In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica restores histone acetylation and increases the expression of learning associated genes (e.g. for treating or preventing Alzheimer's disease). In certain embodiments, these epigenetic modifications may be potential psychiatric treatments.

In certain embodiments, the HDAC inhibitor activity of the composition of the invention comprising Blautia hydrogenotrophica influences the transcription of numerous genes involved in neuronal survival, plasticity and regeneration. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica increases the acetylation around the promoters of neurotrophic factors. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica increases the acetylation around the promoter of BDNF, GDNF and NGF. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica increases the expression of BDNF, GDNF and NGF. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica increases the expression of immediate early genes involved in plasticity, including c-Fos and Homer1a. In certain embodiments, the expression of these factors is altered in the brain.

In certain embodiments, the deacetylase inhibitory activity of the composition of the invention comprising Blautia hydrogenotrophica maintains acetylation of non-histone proteins. In certain embodiments, the acetylation affects the enzymatic and metabolic activity of many proteins. For example, HDAC inhibitors have been shown to maintain acetylation and activation of the transcription factor Sp1 during oxidative stress, enhancing the protective adaptive response to promote cell survival. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica prevents oxidative stress in vivo.

In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica restores blood-brain barrier (BBB) integrity and/or tight junction protein expression (e.g. claudin 5 and/or occluding) in the frontal cortex, hippocampus and striatum. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica restores and/or maintains BBB integrity. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica promotes and/or maintains tight junction expression. Therefore, in certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica can establish and/or maintain a barrier to inflammatory mediators, neurochemical factors, neuropeptides and neurotransmitters associated with central nervous system disorders, in particular neurodevelopmental disorders and/or neuropsychiatric conditions.

In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica inhibits neuro-inflammation. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica increases the levels of IL-1RA (an inhibitor of the pro-inflammatory IL-1β). In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica decreases the levels pro-inflammatory IL-1β and/or TNFα. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica increases IL-4 expression, which increases the levels of IL-1RA.

In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica is an anti-inflammatory agent. In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica inhibits nuclear factor κB (NF-κB) activation. Accordingly, the composition of the invention comprising Blautia hydrogenotrophica may modulate the expression of early immune inflammatory response genes, including IL-1B, TNFα, IL-2, IL-6, IL-8, IL-12, inducible nitric acid synthase, cyclooxygenase-2, intercellular adhesion molecule-1, T cell receptor-α and IHC class II molecules.

In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica affects mitochondrial activity. Accordingly, in certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica treats and/or prevents Alzheimer's disease, Parkinson's disease, Huntington's disease, mitochrondial encephalopathy and/or adrenoleukodystrophy.

In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica affects the signalling through GPCRs. Accordingly, in certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica treats and/or prevents Parkinson's disease.

In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica affects histone acetylation. Accordingly, in certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica treats and/or prevents Alzheimer's disease, Parkinson's disease, and/or Huntington's disease.

In certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica affects microbiome homeostasis. Accordingly, in certain embodiments, the composition of the invention comprising Blautia hydrogenotrophica treats and/or prevents central nervous system disorders and conditions.

In certain embodiments, the compositions of the invention comprising Blautia hydrogenotrophica may trigger improvement in behavioural changes associated with central nervous system disorders or conditions.

In certain embodiments, the effects of Blautia hydrogenotrophica may be independent of butyrate. For example, the Examples demonstrate that administration of Blautia hydrogenotrophica, but not butyrate alone, significantly increases horizontal and vertical activity of mice and time spent in the centre of the open field model suggesting a role in reducing anxiety. Specifically, Blautia hydrogenotrophica displays efficacy in reducing anxiety-like and stereotyped behaviour, while the efficacy of butyrate is limited to reducing stereotyped behaviour.

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 oil (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 endoscopic gastrostomy (PEG), or a port, such as a chest wall port that provides access to the stomach, jejunum 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 pregnant 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 born.

The compositions of the invention may be administered to a patient that has been diagnosed with a central nervous system disorder or condition, in particular a central nervous system disorder or condition mediated by the microbiota-gut-brain axis, or that has been identified as being at risk of a central nervous system disorder or condition, in particular central nervous system disorder or condition mediated by the microbiota-gut-brain axis. The compositions may also be administered as a prophylactic measure to prevent the development of central nervous system disorders or conditions, in particular central nervous system disorders or conditions mediated by the microbiota-gut-brain axis 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 have reduced or absent colonisation by Blautia, in particular Blautia stercoris or Blautia wexlerae. For example, the patient may have reduced or absent colonisation by Blautia, in particular Blautia stercoris, Blautia wexlerae or Blautia hydrogenotrophica.

The compositions of the invention may be administered as a food product, such as a nutritional supplement.

Generally, the compositions of the invention are for the treatment of humans, although they may be used to treat animals 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 animals. If administered to animals, 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 [36], [ ], and [38].

Alternatively, the composition of the invention may comprise a live, active bacterial culture.

In some embodiments, the bacterial strain in the composition of the invention has not been inactivated, for example, has not been heat-inactivated. In some embodiments, the bacterial strain in the composition of the invention has not been killed, for example, has not been heat-killed. In some embodiments, the bacterial strain in the composition of the invention has not been attenuated, for example, has not been heat-attenuated. For example, in some embodiments, the bacterial strain in the composition of the invention has not been killed, inactivated and/or attenuated. For example, in some embodiments, the bacterial strain in the composition of the invention is live. For example, in some embodiments, the bacterial strain in the composition of the invention is viable. For example, in some embodiments, the bacterial strain in the composition of the invention is capable of partially or totally colonising the intestine. For example, in some embodiments, the bacterial strain in the composition of the invention is viable and capable of partially or totally colonising the intestine.

In some embodiments, the composition comprises a mixture of live bacterial strains and bacterial strains that have been killed.

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 degradation until delivery at the target location through, for example, rupturing 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, self-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, references [39] and [40].

The composition may be administered orally and may be in the form of a tablet, capsule or powder. Encapsulated products are preferred because Blautia are anaerobes. Other ingredients (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 beneficial effect upon a patient. A therapeutically effective amount of a bacterial strain may be sufficient to result in delivery to and/or partial or total colonisation of the patient's intestine.

A suitable daily dose of the bacteria, for example for an adult human, may be from about 1×103 to about 1×1011 colony forming units (CFU); for example, from about 1×107 to about 1×1010 CFU; in another example from about 1×106 to about 1×1010 CFU.

In certain embodiments, the composition contains the bacterial strain in an amount of from about 1×106 to about 1×1011 CFU/g, respect to the weight of the composition; for example, from about 1×108 to about 1×1010 CFU/g. The dose may be, for example, 1 g, 3 g, 5 g, and 10 g.

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 transgalacto-oligosaccharides.

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-oligosaccharides, inulin, isomalt-oligosaccharides, pectins, xylo-oligosaccharides (or XOS), chitosan-oligosaccharides (or COS), beta-glucans, arable gum modified and resistant 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 molecule to which three glucose molecules are bonded.

In certain embodiments, the compositions of the invention are used in combination with another therapeutic compound for treating or preventing the central nervous system disorder. In some embodiments, the compositions of the invention are administered with nutritional supplements that modulate central neurotransmitters and neuropeptides. In preferred embodiments, the nutritional supplements comprise or consist of nutritional vitamins. In certain embodiments, the vitamins are vitamin B6, magnesium, dimethylglycine (vitamin B16) and vitamin C. In certain embodiments, the compositions of the invention are administered in combination with another probiotic. In certain preferred embodiments, the probiotic comprises or consists of Trichuris suis ova.

The compositions of the invention may comprise pharmaceutically acceptable excipients or carriers. Examples of such suitable excipients may be found in the reference [41]. Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art and are described, for example, in reference [42]. Examples of suitable carriers include lactose, starch, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like. Examples of suitable diluents include ethanol, 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 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, natural sugars such as glucose, anhydrous lactose, free-flow lactose, beta-lactose, corn sweeteners, natural and synthetic gums, such as acacia, tragacanth or sodium alginate, carboxymethyl cellulose and polyethylene glycol. Examples of suitable lubricants include sodium oleate, sodium stearate, magnesium stearate, 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. Antioxidants 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 supplement. 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, the composition of the invention is formulated as a milk-based product. The term “milk-based product” means any liquid or semi-solid milk- 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 recombined 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 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 some embodiments, the compositions of the invention comprise one or more bacterial strains of the genus Blautia and do not contain bacteria from any other genera, or which comprise only de minimis or biologically irrelevant amounts of bacteria from another genera. Thus, in some embodiments, the invention provides a composition comprising one or more bacterial strains of the genus Blautia, which does not contain bacteria from any other genera or which comprises only de minimis or biologically irrelevant amounts of bacteria from another genera, for use in therapy.

In some embodiments, the compositions of the invention comprise one or more bacterial strains of the species Blautia stercoris or Blautia wexlerae and do not contain bacteria from any other species, or which comprise only de minimis or biologically irrelevant amounts of bacteria from another species.

Thus, in some embodiments, the invention provides a composition comprising one or more bacterial strains of the species Blautia stercoris or Blautia wexlerae, which does not contain bacteria from any other species or which comprises only de minimis or biologically irrelevant amounts of bacteria from another species, for use in therapy.

In some embodiments, the compositions of the invention comprise one or more bacterial strains of the species Blautia hydrogenotrophica and do not contain bacteria from any other species, or which comprise only de minimis or biologically irrelevant amounts of bacteria from another species. Thus, in some embodiments, the invention provides a composition comprising one or more bacterial strains of the species Blautia hydrogenotrophica, which does not contain bacteria from any other species or which comprises only de minimis or biologically irrelevant amounts of bacteria from another species, for use in therapy.

In some embodiments, the compositions of the invention comprise one or more bacterial strains of the species Blautia stercoris or Blautia wexlerae and do not contain bacteria from any other Blautia species, or which comprise only de minimis or biologically irrelevant amounts of bacteria from another Blautia species. Thus, in some embodiments, the invention provides a composition comprising one or more bacterial strains of the species Blautia stercoris or Blautia wexlerae, which does not contain bacteria from any other Blautia species or which comprises only de minimis or biologically irrelevant amounts of bacteria from another Blautia species, for use in therapy.

In some embodiments, the compositions of the invention comprise one or more bacterial strains of the species Blautia hydrogenotrophica and do not contain bacteria from any other Blautia species, or which comprise only de minimis or biologically irrelevant amounts of bacteria from another Blautia species. Thus, in some embodiments, the invention provides a composition comprising one or more bacterial strains of the species Blautia hydrogenotrophica, which does not contain bacteria from any other Blautia species or which comprises only de minimis or biologically irrelevant amounts of bacteria from another Blautia species, for use in therapy.

In certain embodiments, the compositions of the invention contain a single bacterial strain or species and do not contain any other bacterial strains or species. Such compositions may comprise only de minimis or biologically irrelevant amounts of other bacterial strains or species. Such compositions may be a culture that is substantially free from other species of organism.

In some embodiments, the invention provides a composition comprising a single bacterial strain of the genus Blautia, which does not contain bacteria from any other strains or which comprises only de minimis or biologically irrelevant amounts of bacteria from another strain for use in therapy.

In some embodiments, the invention provides a composition comprising a single bacterial strain of the species Blautia stercoris or Blautia wexlerae, which does not contain bacteria from any other strains or which comprises only de minimis or biologically irrelevant amounts of bacteria from another strain for use in therapy.

In some embodiments, the invention provides a composition comprising a single bacterial strain of the species Blautia hydrogenotrophica, which does not contain bacteria from any other strains or which comprises only de minimis or biologically irrelevant amounts of bacteria from another strain for use in therapy.

In some embodiments, the compositions of the invention comprise more than one bacterial strain. For example, in some embodiments, the compositions of the invention comprise more than one strain from within the same species (e.g. more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40 or 45 strains), and, optionally, do not contain bacteria from any other species. In some embodiments, the compositions of the invention comprise less than 50 strains from within the same species (e.g. less than 45, 40, 35, 30, 25, 20, 15, 12, 10, 9, 8, 7, 6, 5, 4 or 3 strains), and, optionally, do not contain bacteria from any other species. In some embodiments, the compositions of the invention comprise 1-40, 1-30, 1-20, 1-19, 1-18, 1-15, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 2-50, 2-40, 2-30, 2-20, 2-15, 2-10, 2-5, 6-30, 6-15, 16-25, or 31-50 strains from within the same species and, optionally, do not contain bacteria from any other species. The invention comprises any combination of the foregoing.

In some embodiments, the composition comprises a microbial consortium. For example, in some embodiments, the composition comprises the Blautia bacterial strain as part of a microbial consortium. For example, in some embodiments, the Blautia bacterial strain is present in combination with one or more (e.g. at least 2, 3, 4, 5, 10, 15 or 20) other bacterial strains from other genera with which it can live symbiotically in vivo in the intestine. For example, in some embodiments, the composition comprises a bacterial strain of Blautia in combination with a bacterial strain from a different genus. In some embodiments, the microbial consortium comprises two or more bacterial strains obtained from a faeces sample of a single organism, e.g. a human. In some embodiments, the microbial consortium is not found together in nature. For example, in some embodiments, the microbial consortium comprises bacterial strains obtained from faeces samples of at least two different organisms. In some embodiments, the two different organisms are from the same species, e.g. two different humans. In some embodiments, the two different organisms are an infant human and an adult human. In some embodiments, the two different organisms are a human and a non-human mammal.

In some embodiments, the composition of the invention additionally comprises a bacterial strain that has the same safety and therapeutic efficacy characteristics as strain MRX006, but which is not MRX006 deposited as NCIMB 42381, or which is not a Blautia stercoris. In some embodiments, the composition of the invention additionally comprises a bacterial strain that has the same safety and therapeutic efficacy characteristics as strain MRX008, but which is not MRX008 deposited as NCIMB 42486, or which is not a Blautia wexlerae.

In some embodiments, the composition of the invention additionally comprises a bacterial strain that has the same safety and therapeutic efficacy characteristics as the Blautia hydrogenotrophica strain deposited as DSM 14294, but which is not the Blautia hydrogenotrophica strain deposited as DSM 14294, or which is not a Blautia hydrogenotrophica.

In some embodiments, the composition of the invention does not comprise a bacterial strain of the genus Bacillus. In some embodiments, the composition of the invention does not comprise Bacillus subtilis and/or does not comprise Bacillus coagulans. In some embodiments, the CNS disorder to be treated by the composition of the invention is not bipolar disorder. In some embodiments, the patient to be treated by the composition of the invention does not have a fungal infection. In some embodiments, the patient to be treated by the composition of the invention does not suffer from candidiasis. In some embodiments, the patient to be treated by the composition of the invention has not been diagnosed as having a fungal infection and/or has not been diagnosed as suffering from candidiasis. In preferred such embodiments, the patient to be treated by the composition of the invention has never been diagnosed as having a fungal infection and/or has never been diagnosed as suffering from candidiasis.

In some embodiments in which the composition of the invention comprises more than one bacterial strain, species or genus, the individual bacterial strains, species or genera may be for separate, simultaneous or sequential administration. For example, the composition may comprise all of the more than one bacterial strain, species or genera, or the bacterial strains, species or genera may be stored separately and be administered separately, simultaneously or sequentially. In some embodiments, the more than one bacterial strains, species or genera are stored separately but are mixed together prior to use.

In some embodiments, the bacterial strain for use in the invention is obtained from human adult faeces. In some embodiments in which the composition of the invention comprises more than one bacterial strain, all of the bacterial strains are obtained from human adult faeces or if other bacterial strains are present they are present only in de minimis amounts. The bacteria may have been cultured subsequent to being obtained from the human adult faeces and being used in a composition of the invention.

As mentioned above, in some embodiments, the one or more Blautia bacterial strains is/are the only therapeutically active agent(s) in a composition of the invention. In some embodiments, the bacterial strain(s) in the composition is/are the only therapeutically active agent(s) in a composition of the invention.

The compositions for use in accordance with the invention may or may not require marketing approval.

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 said bacterial strain is spray dried. In certain embodiments, the invention provides the above pharmaceutical composition, wherein the bacterial strain is lyophilised or spray dried and wherein it is live. In certain embodiments, the invention provides the above pharmaceutical composition, wherein the bacterial strain is lyophilised or spray dried and wherein it is viable. In certain embodiments, the invention provides the above pharmaceutical composition, wherein the bacterial strain is lyophilised or spray dried and wherein it is capable of partially or totally colonising the intestine. In certain embodiments, the invention provides the above pharmaceutical composition, wherein the bacterial strain is lyophilised or spray dried and wherein it is viable and capable of partially or totally colonising the intestine.

In some cases, the lyophilised or spray dried bacterial strain 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, diluents or carriers.

In certain embodiments, the invention provides a pharmaceutical composition comprising: a bacterial strain as used in 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: autism spectrum disorders (ASDs); child developmental disorder; obsessive compulsive disorder (OCD); major depressive disorder; depression; seasonal affective disorder; anxiety disorders; schizophrenia spectrum disorders; schizophrenia; bipolar disorder; psychosis; mood disorder; chronic fatigue syndrome (myalgic encephalomyelitis); stress disorder; post-traumatic stress disorder; dementia; Alzheimer's; Parkinson's disease; and/or chronic pain; motor neuron disease; Huntington's disease; Guillain-Barre syndrome and/or meningitis.

In certain embodiments, the invention provides pharmaceutical composition comprising: a bacterial strain as used in the invention; and a pharmaceutically acceptable excipient, carrier or diluent; wherein the bacterial strain is in an amount sufficient to treat or prevent a central nervous system disorder or condition, in particular central nervous system disorder or condition mediated by the microbiota-gut-brain axis. In preferred embodiments, said disease or condition is selected from the group consisting of: autism spectrum disorders (ASDs); child developmental disorder; obsessive compulsive disorder (OCD); major depressive disorder; depression; seasonal affective disorder; anxiety disorders; schizophrenia spectrum disorders; schizophrenia; bipolar disorder; psychosis; mood disorder; chronic fatigue syndrome (myalgic encephalomyelitis); stress disorder; post-traumatic stress disorder; dementia; Alzheimer's; Parkinson's disease; and/or chronic pain. In further embodiments, the compositions of the invention may be useful for treating or preventing motor neuron disease; Huntington's disease; Guillain-Barre syndrome and/or meningitis.

In certain embodiments, the invention provides the above pharmaceutical composition, wherein the amount of the bacterial strain is from about 1×103 to about 1×1011 colony forming units per gram with respect to a weight of the composition.

In certain embodiments, the invention provides the above pharmaceutical composition, wherein the composition is administered at a dose of 1 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, subcutaneous, 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, magnesium stearate, mannitol and sorbitol.

In certain embodiments, the invention provides the above pharmaceutical composition, comprising a diluent selected from the group consisting of ethanol, 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, free-flow lactose, beta-lactose, corn sweetener, acacia, tragacanth, sodium alginate, carboxymethyl cellulose, polyethylene glycol, sodium oleate, sodium stearate, magnesium stearate, 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 p-hydroxybenzoic acid.

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 atmosphere 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: 1 month, 3 months, 6 months, 1 year, 1.5 years, 2 years, 2.5 years or 3 years.

In some embodiments, the composition of the invention is provided in a sealed container comprising a composition as described herein. In some embodiments, the sealed container is a sachet or bottle. In some embodiments, the composition of the invention is provided in a syringe comprising a composition as described herein.

The composition of the present invention may, in some embodiments, be provided as a pharmaceutical formulation. For example, the composition may be provided as a tablet or capsule. In some embodiments, the capsule is a gelatine capsule (“gel-cap”).

In some embodiments, the compositions of the invention are administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, and/or buccal, lingual, or sublingual administration by which the compound enters the blood stream directly from the mouth.

Pharmaceutical formulations suitable for oral administration include solid plugs, solid microparticulates, semi-solid and liquid (including multiple phases or dispersed systems) such as tablets; soft or hard capsules containing multi- or nano-particulates, liquids (e.g. aqueous solutions), emulsions or powders; lozenges (including liquid-filled); chews; gels; fast dispersing dosage forms; films; ovules; sprays; and buccal/mucoadhesive patches.

In some embodiments the pharmaceutical formulation is an enteric formulation, i.e. a gastro-resistant formulation (for example, resistant to gastric pH) that is suitable for delivery of the composition of the invention to the intestine by oral administration. Enteric formulations may be particularly useful when the bacteria or another component of the composition is acid-sensitive, e.g. prone to degradation under gastric conditions.

In some embodiments, the enteric formulation comprises an enteric coating. In some embodiments, the formulation is an enteric-coated dosage form. For example, the formulation may be an enteric-coated tablet or an enteric-coated capsule, or the like. The enteric coating may be a conventional enteric coating, for example, a conventional coating for a tablet, capsule, or the like for oral delivery. The formulation may comprise a film coating, for example, a thin film layer of an enteric polymer, e.g. an acid-insoluble polymer.

In some embodiments, the enteric formulation is intrinsically enteric, for example, gastro-resistant without the need for an enteric coating. Thus, in some embodiments, the formulation is an enteric formulation that does not comprise an enteric coating. In some embodiments, the formulation is a capsule made from a thermogelling material. In some embodiments, the thermogelling material is a cellulosic material, such as methylcellulose, hydroxymethylcellulose or hydroxypropylmethylcellulose (HPMC). In some embodiments, the capsule comprises a shell that does not contain any film forming polymer. In some embodiments, the capsule comprises a shell and the shell comprises hydroxypropylmethylcellulose and does not comprise any film forming polymer (e.g. see [43]). In some embodiments, the formulation is an intrinsically enteric capsule (for example, Vcaps® from Capsugel).

In some embodiments, the formulation is a soft capsule. Soft capsules are capsules which may, owing to additions of softeners, such as, for example, glycerol, sorbitol, maltitol and polyethylene glycols, present in the capsule shell, have a certain elasticity and softness. Soft capsules can be produced, for example, on the basis of gelatine or starch. Gelatine-based soft capsules are commercially available from various suppliers. Depending on the method of administration, such as, for example, orally or rectally, soft capsules can have various shapes, they can be, for example, round, oval, oblong or torpedo-shaped. Soft capsules can be produced by conventional processes, such as, for example, by the Scherer process, the Accogel process or the droplet or blowing process.

Culturing Methods

The bacterial strains for use in the present invention can be cultured using standard microbiology techniques as detailed in, for example, references [44], [ ] and [46].

The solid or liquid medium used for culture may be YCFA agar or YCFA medium. YCFA medium may include (per 100 ml, approximate values): Casitone (1.0 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 (0.1 mg), hemin (1 mg), biotin (1 μg), cobalamin (1 μg), p-aminobenzoic acid (3 μg), folic acid (5 μg), and pyridoxamine (15 μg).

Bacterial Strains for Use in Vaccine Compositions

The inventors have identified that the bacterial strains of the invention are useful for treating or preventing central nervous system disorders or conditions, in particular central nervous system disorders or conditions mediated by the microbiota-gut-brain axis. This is likely to be a result of the effect that the bacterial strains of the invention have on the host central, autonomic and/or enteric nervous system; the activity of the HPA pathway; the neuroimmune and neuroendocrine pathways; and the level of commensal metabolites in the host gastrointestinal tract and/or gastrointestinal permeability of the host. Therefore, the compositions of the invention may also be useful for preventing central nervous system disorders or conditions, in particular central nervous system disorders or conditions mediated by the microbiota-gut-brain axis, when administered as vaccine compositions. In certain such embodiments, the bacterial strains of the invention are viable. In certain such embodiments, the bacterial strains of the invention are capable of partially or totally colonising the intestine. In certain such embodiments, the bacterial strains of the invention are viable and capable of partially or totally colonising the intestine. In other 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 present 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 [47] and [48]-[54], etc.

The term “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%.

In certain embodiments the term “modulate” means increase or activate. In alternative embodiments, the term “modulate” means decrease or suppress.

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 definition of the invention.

References to a percentage sequence identity between two nucleotide sequences means that, when aligned, that percentage of nucleotides 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. [55]. A preferred alignment 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 Smith-Waterman homology search algorithm is disclosed in ref [56].

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

The present study aimed to assess the effect of live biotherapeutics on the treatment of psychiatric and neurological disorders in two different mouse models that display behavioural characteristics associated with neurodevelopmental and psychiatric disorders. In particular, the study focuses on autistic-related behaviour in (i) C57BL/6 wt mouse model, (ii) a BTBR inbred, genetically modified mouse model and (iii) a maternal immune activation (MIA) mouse model. The effects of chronic MRX006 versus vehicle treatment across anxiety, depression, and cognitive and social domains of behaviour in the three mouse models were investigated. In addition, physiological and anatomical analyses were performed as well as detection of circulating cytokine and oxytocin levels.

The EMA Guidelines on the clinical development of medicinal products for the treatment of autism spectrum disorder state that, due to the heterogeneity of the diseases, it may not be possible to achieve a significant effect on all core symptoms with a single compound, and so short term efficacy has to be demonstrated on at least one core symptom. The live biotherapeutics tested in the Examples have shown effective treatment of at least one core symptom of autistic spectrum disorder, so these strains and related Blautia strains are expected to be effective against human disease. Similarly, other central nervous system disorders or conditions display complex pathology with multiple different symptoms, and have few approved treatments. Therefore, it is understood that an effective treatment does not need to treat all symptoms of a central nervous system disorder or condition. A treatment would be considered therapeutically useful if it treated one of the symptoms associated with a central nervous system disorder or condition.

Example 1—Assessing Anxiety, Depression, Cognitive and Social Domains of Behaviour in C57BL/6 Mice Example 1a—Materials and Methods for the C57BL/6 Mice Experiments Mice

Male C57BL/6 mice were purchased from Harlan UK. The animals were housed in a temperature- and humidity-controlled room on a 12 hr dark cycle (lights on from 7:00-19:00 hr). All experiments were conducted in accordance with the European Directive 2010/63/EEC, the requirements of S.I. No 543 of 2012, and approved by the Animal Experimentation Ethics Committee of University College Cork.

Strain

MRX006: Blautia stercoris bacterium deposited under accession number NCIMB 42381.

Biotherapeutic was provided in glycerol stock. Live biotherapeutics were grown in the facility in anaerobic conditions.

Live Biotherapeutic Administration

Dosing with MRX006 or vehicle (control) commenced when the mice were 7 weeks old. These mice were treated once daily with MRX006 or phosphate buffer solution (PBS) for 4 weeks before the beginning of the behavioural battery. Mice were further treated once daily during the behavioural battery. MRX006 (1×109 CFU oral administration) was dissolved in PBS prior to administration.

Administration Schedule

The treatment groups for the study are shown below. The vehicle for oral administration is PBS. Daily oral administration occurs via oral gavage.

Group Treatment Number 1 Naïve (no gavage) 12 2 Control (PBS, oral gavage) 12 3 MRX006 (oral gavage in PBS) 12

The Naïve group was not handled until the beginning of the behavioural battery.

Fecal Collection

Fresh fecal samples were collected from individual mice every week until the end of the study. At least 20 mg of fresh faeces were placed in a microcentrifuge tube, place immediately on ice and then stored at −80° C.

Experimental Design and Methods

As outlined above, dosing with MRX006 commenced when the mice were 7 weeks old. The initial dosing took place for 4 weeks before the behavioural experiments. The behavioural battery occurred in the following order: marble burying and 3 chamber tests at week 5; the elevated plus maze and tail suspension tests at week 6; the open field and novel object recognition tests at week 8; the stress-induced hyperthermia test at week 9; the fear conditioning test at week 10; and the forced swim test at week 11. The fluorescein gut permeability assay was performed at week 9. Finally, in weeks 12, the mice were culled and dissected for brain, proximal and distal colon, adrenal and spleen regions, along with plasma samples.

Graphical Design and Statistical Analysis

All graphs were generated on graphpad prism software (version 5). Data were analysed using IBM SPSS Statistic 22.0 (EEUU). Data distribution was analysed using the Kolmogorov-Smirnov normality test. Data comparing vehicle group versus MRX006 groups were analysed using two-way ANOVA and Fisher's least significant difference (LSD) post hoc test. Data comparing vehicle group versus naïve mice were analysed by unpaired Student t test. Non-normally distributed data were analysed by the Kruskal-Wallis and non-parametric Mann-Whitney U test. P<0.05 was the criterion for statistical significance.

Example 1b—Assessment of Social Interaction Behaviour—the Three Chamber Tests

Rationale

The 3-Chamber Social Interaction Test (3-CSIT) is a well validated ethologically relevant model that assesses social interaction between sex-matched conspecifics and allows for readouts of social novelty and social preference in mice. The test is used to characterize and demonstrate changes in this behavioural readout. The test allows mice to freely explore between an inanimate object or sex-matched conspecific mice.

In addition, the 3-chamber test (3-CHT) is a test used to assess cognition in the form of general sociability and interest in social novelty in rodent models. Rodents normally prefer to spend more time with another rodent (sociability) than with an object. Moreover, rodents prefer to investigate a novel mouse versus a familiar mice (social novelty). Based on these inclinations, the 3-CHT can help identify rodents with deficits in sociability and/or social novelty.

Methods

Animals are placed in a rectangular apparatus divided into three chambers (left and right and a smaller centre chamber) by partitions with small circular openings allowing easy access to all compartments. The test is composed of three sequential 10 min trials: (1) habituation (the test animal is allowed to explore the three empty chambers); (2) sociability (an unfamiliar animal is placed in an inner mesh wire cage in either the left or right chambers); (3) social novelty preference (a novel animal is placed into the previously empty inner cage in the chamber, opposite the now familiar animal). Naive animals should have no preference for either chamber in the habituation phase, a preference for the mouse in the sociability phase, and a preference for the novel mouse in the social novelty phase. An increase in the discrimination ratio would suggest an increase in social behaviour. All animals are age- and sex-matched, with each chamber cleaned and lined with fresh bedding after each 30 minute trial. For each of the three stages, behaviour is recorded by a video camera mounted above the apparatus.

Results

Student t test within groups revealed that all groups spent more time investigating a mouse versus an object (**p<0.01) suggesting no deficits in sociability (FIG. 1). Daily gavage did not affect sociability. Interestingly, MRX006 enhanced the time spent investigating a novel versus a familiar mice, suggesting increased social novelty (p<0.05; FIG. 1).

Conclusions

Chronic treatment with MRX006 enhanced preference for social novelty in C57BL/6 mice in the three chamber test.

Example 1c—Assessing Depression-Like Behaviour—the Forced Swim Test (FST)

Rationale

The forced swim test (FST) is the most widely used experimental paradigm to assess antidepressant activity. Naïve animals will display escape behaviour in the form of swimming, climbing and diving before adapting an immobile floating posture. The duration of immobility is indicative of behavioural despair. Antidepressant drugs decrease the time spent immobile in this test.

Methods

Mice are forced to swim for 6 min in a glass cylinder (24×21 cm) filled with 23-25° C. tap water to a depth of 17 cm. The FST was videotaped from a ceiling camera. The behavioural parameter scored is immobility during the last 4 min of the 6-min test.

Results

Daily gavage increased immobility time suggesting depressive-like behaviour compared to naïve mice (t Student test, t (190=4.565; p<0.01; FIG. 2). Chronic treatment with MRX006 significantly reduced immobility suggesting antidepressant-like effects compared to the vehicle group (F (2.29)=14.992; **p<0.01).

Conclusions

Chronic treatment with MXR006 induced antidepressant-like behaviour in the forced swim test.

Example 1d—Assessing Depression-Like Behaviour—the Tail Suspension Test

Rationale

The tail suspension test is a well-characterized test used to assess antidepressant-like behaviour. The time spend immobile is an index of depression-like behaviour. Treatment with antidepressant drugs decreases the time spent immobile.

Methods

Mice are suspended to an elevated bar (60 cm) by a piece of adhesive tape attached 1 cm before the tip of their tail for a period of 6 min. The mice are suspended in such a way that they cannot escape or hold on to nearby surfaces. During this test, six minutes in duration, the resulting escape oriented behaviours are quantified. The behavioural parameter scored is time spent immobile. The test was video-recorded by a tripod camera and the time of immobility was scored manually by an investigator blind to the experimental conditions.

Results

Daily handling for gavage (Student t test, t (20)=0.9405; p=0.3582) and chronic treatment with MRX006 (one-way ANOVA, F (2.30)=2.014; p=0.152) did not induce any significant effect in the tail suspension test (FIG. 3).

Conclusion

Chronic treatment with MRX006 did not induce any observable anti-depressant-like behaviour in C57BL/6 mice in the tail suspension test.

Example 1e—Assessing Cognition—the Fear Conditioning Test

Rationale

Contextual fear conditioning is used as a measure of hippocampus-dependent memory. Fear conditioning is a form of an associative learning which measures the freezing response displayed by the animal to an unconditioned stimuli (US), such as a shock with a conditioned stimulus (CS), a particular tone or light or smell. The measurement of freezing levels was used to assess the animal response to the US and CS stimuli. This test measures how efficiently the mice forget what they have acquired on the acquisition day. The test assesses the anxiety of mice towards conditioned stimuli associated with unconditioned stimuli and the speed at which the mice show reduced anxiety and/or stress (freezing levels) in the presence of conditioned stimuli in the repeated absence of paired unconditioned stimuli.

Methods

The apparatus for this test consisted of chambers with a light above. Each chamber is located inside a larger chamber, which protects from outside light and noise. On the first day (training or acquisition stage), mice were placed into the chamber and their freezing behaviour was recorded for 3 min (baseline), followed by up to 6 light/tone [conditioned stimulus (CS—30 s)] and footshock [unconditioned stimulus (US—2 s)] pairings with an interval of up to 2 min. Pairings consisted of the cue [e.g., a combined light (˜260 lx) and tone exposure (80 dB)] for 20 s and an electric footshock during the last 2 s of the cue. Foot shock increases freezing behaviour. The intensity of the current was 0.6 mA. The minimal current that induces a freezing response was used. Two minutes after the last pairing, mice were returned to their normal housing conditions. At 24 and 48 h after conditioning (days 2 (retrieval stage) and 3 (extinction stage), respectively), the same experimental procedure was repeated in absence of footshocks to test for memory retention and extinction of the conditioned fear memory (extinction phase). Contextual memory retention is characterized by freezing behaviour when the animal is placed in the context (i.e. the footshock chamber) in the absence of a foot shock.

Results

Freezing levels were measured in the acquisition phase (exposure to both CS and US coupled together), retrieval and extinction phase (CS not coupled with US) (see FIG. 4). In this study, daily gavage induced a significant increase in freezing levels (phase 1 #p<0.05; phase 2 ###p<0.001; phase 3-4 ##p<0.01) in the acquisition phase as compared to naïve animals. MRX006 chronic treatment did not alter freezing levels when compared to the vehicle group during the acquisition phase. The acquisition phase was followed by the retrieval phase which took place 24 hours after the training session (CS not followed by US). The data showed a significant increase in freezing levels in phase 1 (exposure to the first tone) in the vehicle group compared to the naïve group (p<0.05). Interestingly, a significant reduction in the freezing levels was observed at phase 1 in MRX006-treated mice (*p<0.05) when compared to the vehicle group. Overall, in the retrieval phase, MRX006-treated displayed a trend for decreased freezing levels when compared to the vehicle group, suggesting that chronic treatment with MRX006 may enhance learning. The retrieval phase was followed by the extinction phase (24 hours later, CS and no US). This test measures how efficiently the mice forget what they have acquired on the acquisition day. In phase 3 of extinction phase the vehicle group displayed increased freezing levels when compared to the naïve group (p<0.05). Chronic treatment with MRX006 did not induce any significant change in the extinction phase.

Conclusion

Chronic treatment with MRX006 reduced freezing levels in the retrieval phase suggesting MRX006 may enhance learning. Overall, chronic treatment with MRX006 did not observably alter significantly fear conditioned behaviour.

Example 1f—Assessing Cognition—the Novel Object Recognition (NOR) Test

Rationale

The protocol used was adapted from Bevins and Besheer (2006), and used to test cognition, particularly recognition memory. This test is based on the spontaneous tendency of rodents to spend more time exploring a novel object than a familiar one. The choice to explore the novel object reflects the use of learning and recognition memory. In addition, improved memory is a reflection of reduced depression-like behaviour.

Methods

The protocol used was adapted from Bevins and Besheer (2006). It was conducted over 3 days. On Day 1, the animals were allowed to acclimate to the testing environment for 10 minutes, which was a large container equipped with an overhead camera. No bedding was used and the container was wiped with 70% ethanol between each animal. On Day 2, the animals were allowed to acclimate to the test apparatus for 10 minutes. Following this period, the animal was removed from the container and two identical objects were introduced to the environment. The animal was returned to the container and allowed to explore for a further 10 minutes. The objects were cleansed before each trial with a 70% ethanol solution. Following the training period, the rodent was removed from the environment for a delay period of 24 hours. On Day 3, the rodent was returned to the container, which this time contained only 1 familiar object from the day previous and 1 novel object. Activity of the animal with the 2 objects was recorded for 5 minutes. The amount of time that the rodent spent exploring each object was recorded by manual observation and a discrimination index (DI) value corresponding to time spent interacting with the novel object over total interaction time was generated. A decrease in DI compared to control rats indicates a deficit in this type of memory.

Results

Daily handling for gavage did not affect recognition memory in C57Bl/6 mice (FIG. 5). Indeed, both groups show a pattern for preference exploring more a novel versus a familiar object (although did not reach statistically significance). Mice chronically treated with MRX006 display significantly increased preference exploring a novel object versus a familiar one (p<0.05) (FIG. 5).

Conclusion

Chronic treatment with MRX006 resulted in C57Bl/6 mice spending significantly more time investigating a novel versus a familiar object suggesting enhance recognition memory.

Example 1g—Assessing Anxiety-Like Behaviour—the Marble Burying Test

Rationale

The marble burying test is a useful model of neophobia, anxiety and obsessive compulsive behaviour. It is also used to test novel antidepressants, anxiolytics and antipsychotics. Mice pre-treated with pharmacological agents such as anxiolytics show decreased marble burying behaviour, compared to the control mice.

Methods

Mice were individually placed into a novel polypropylene cage (35×28×18.5 cm, L×W×H), containing standard rodent (hard wood) bedding (5 cm) and 20 marbles on top of it (five rows of marbles regularly spaced 2 cm away from the walls and 2 cm apart). Experiments were conducted under a light intensity of 1000 lux. 30 minutes later, mice were removed from these cages and the number of marbles buried for more than ⅔rds of their surface was scored.

Results

Student t test (vehicle versus naïve; t (20)=0.1308; p=0.8973) and one way ANOVA analysis (F (2.38)=0.992; p=0.384) revealed that neither chronic treatment with MRX006 nor daily gavage altered the number of marbles buried suggesting no alterations in anxiety-like baselines (FIG. 6).

Conclusions

Chronic treatment with MRX006 did not alter observably anxiety-like behaviour in C57Bl/6 mice in the marble burying test.

Example 1h—Assessing Anxiety-Like Behaviour—the Elevated Plus Maze Test

Rationale

The elevated plus maze (EPM) is a widely used test to assess anxiety-like behaviours in rodents. The EPM assesses general anxiety behaviour, with less anxious mice spending more time in the open arms of the maze. An increase in open arm activity (duration) reflects anti-anxiety behaviour.

Methods

The set up consisted of a grey plastic cross-shaped maze 1 meter elevated from the floor, comprising two open (aversive) and two closed (safe) arms (50×5×15 cm walls). Experiments occurred under red light (7 lux). Mice were placed into the centre of the maze facing an open arm (to avoid direct entrance into a closed arm) and were allowed to explore the arena for a duration of six minutes. Experiments were videotaped using a ceiling camera to allow for measuring several behavioural parameters. The apparatus was cleaned with 70% (vol/vol) ethanol after each subject to prevent olfactory cues from the previous mouse. Time spent in the open/closed arms, time spent in the center, and the number of transitions were analysed manually. The percentage of time spent and the number of entries in each arm was measured for anxiety-like behaviour and locomotor activity, respectfully. Entrance into an arm was defined as all four paws inside the arm. An increase in open arm activity (duration) reflects anti-anxiety behaviour.

Results

Student t test analysis revealed that daily gavage did not affect the time spent in open arms (FIG. 7). One-way ANOVA analysis revealed that chronic treatment with MRX006 did not alter the behaviour in the elevated plus maze when compared to the control group (FIG. 7). Specifically, chronic treatment with Mrx006 did not alter the time spent in open arms and in closed arms.

Conclusions

Chronic treatment with MRX006 did not observably alter behaviour of C57Bl/6 mice in the elevated plus maze.

Example 1i—Assessing Anxiety Levels in the Stress Induced Hyperthermia (SIH) Test

Rationale

The SIH paradigm is a well-characterised index of anxiety. In this test, the stress is triggered simply by the measurement of rectal temperature.

Methods

Briefly, animals were singly housed 1 d before the test. Rectal temperature was measured twice with a 15 min interval using a lubricated temperature-sensitive probe. Due to the stress experienced during the first temperature measurement, the temperature of the second measurement (T2) is higher than that of the first (T1). This difference in temperature (ΔT=T2−T1) is defined as the SIH response. The SIH response is reduced by different classes of anxiolytics.

Results

Daily handling for gavage increased ΔT suggesting anxiety-like behaviour (Student t test, t (19)=2.121, p=0.047). Chronic treatment with MRX006 did not induce changes in ΔT when compared to the vehicle group (one-way ANOVA, F (2.29)=1.215; p=0.312) (FIG. 8).

Conclusions

Chronic treatment with MRX006 did not observably alter the stress induced change in temperature in the stress induced hyperthermia test in C57Bl/6 mice.

Example 1j—Physiological Analysis—Plasma Oxytocin Levels

Methods and Results

Oxytocin levels were measured in naïve, vehicle and MRX006 treated mice (FIG. 9). Oxytocin peptide, which acts in the central nervous systems of males and females is critical for a variety of complex social behaviours including affiliation, sexual behaviour, social recognition, aggression and trust. Radioimmunoassay (RIA) is a sensitive method for measuring very small quantities of peptides and metabolites in the blood. Samples were prepared for RIA and dispatched for oxytocin measure through RIA technique (RIAgnosis, Sinzing, Germany). Chronic treatment with MRX006 in C57Bl/6 mice showed a reduction of oxytocin levels (p<0.05).

Example 1k—Physiological Analysis—Stress-Induced Corticosterone Plasma Levels

Methods and Results

Corticosterone is a major rodent hormone released in response to stress. In this study corticosterone level changes were measured at baseline and following the forced swim test (acute stress exposure) in naïve, vehicle and MRX6-treated mice (FIG. 10). Corticosterone measurements were carried out at different time points, namely at T0 (before forced swim test), and 30 min (T30), 90 min (T90) and 120 min (T120) post-exposure to forced swim test. The data showed a significant increase in stress-induced corticosterone levels in the vehicle group compared to the naïve group at T30 (p<0.05). Interestingly, a significant increase in stress-induced corticosterone levels was observed at T30 in MRX6-treated mice when compared to the control group. No significant changes were observed in the other time points. These results may suggest an increased sensitivity to stress in both the vehicle and the MRX006 treated group.

Example 1l—Physiological Analysis—In Vivo Gastrointestinal Permeability Assay

Rationale

This procedure is used to assess in vivo intestinal motility. Gut permeability was quantified between the different chronic treatments through the quantification of Fluorescein isothiocynate (FITC) in the blood post oral administration of the fluorescein derivative. It is an established method to quantify the gut permeability, based on the principal of leaking of the orally administered fluorescein derivative through the gut into the peripheral system.

Methods

Test mice were singly-housed and fasted overnight. The following morning (˜9 am), mice were administered FITC dextran ((600 mg/kg) by oral gavage. Two hours later, 100 μl of blood sample was collected in heparin-coated capillary tubes and transferred to a darkened eppendorf and placed on ice. Samples were centrifuged 3500×g for 15 minutes, plasma was aspirated and samples were stored at −80° D for long storage.

Undiluted plasma was used to quantify FITC concentration. 25 μl of FITC was pipetted in duplicated in 384 well plate (Greiner bio one). FITC was measured with a Victor spectrometer between the ranges of 490 nm-520 nm. For a standard curve, a serial dilution of FITC was prepared in PBS (pH7.4). An increase in absorbance is indicative of a decrease in barrier integrity.

Results

The data showed a trend towards an increase in intestine permeability with daily gavages (p=0.051) but it failed to reach significance (FIG. 11). Overall intestine permeability between the MRX006 groups remained unaltered.

Conclusions

Chronic treatment with MRX006 displayed no observable effect on gut permeability.

Example 1m—Physiological Analysis—Organ Weight and Colon Length

Daily handling for gavage and chronic treatment with MRX006 did not induce changes in caecum weight, spleen weight, and adrenal weight (FIG. 12).

Conclusions from the C57B/6 Mouse Model

Chronic treatment with MRX006 induced antidepressant-like effects in the forced swim test, a widely used test to screen antidepressant-like activity. In addition, chronic treatment with MRX006 enhanced social behaviour in C57Bl/6 mice, which spend more time interacting with novel versus familiar mice in the 3-chamber test, indicating that MRX006 reversed social impairments, a core symptom of autism spectrum disorders. Furthermore, chronic treatment with MRX006 tended to reduce freezing levels in the fear conditioning test indicating that administration of this strain improves cognitive functions of memory and reduces anxiety in C57Bl/6 mice.

Further studies are required to characterise the effects of MRX006 when acutely or sub-chronically administered on oxytocin levels and corticosterone levels. The evidence indicates that MRX006 modulates signalling of the hypothalamic pituitary axis (HPA).

Therefore, administration of MRX006 causes antidepressant effects, enhances social novelty and pro-cognitive effects.

Example 2—the BTBR Mouse Model

The BTBR mouse model uses inbred, genetically modified mice that display a robust autistic-like phenotype. Deficits in social behaviours, increased repetitive behaviours and increased anxiety-related behaviours have been reported in this strain (Meyza and Blanchard, 2017). Due to this robust behavioural phenotype, the BTBR mouse is an ideal animal model to assess the efficacy of novel therapeutic agents for the treatment of autistic-related behaviours. Alleviation of such symptoms by a live biotherapeutic can also be indicative of efficacy of the biotherapeutic in the treatment of other psychiatric or neurological diseases.

Example 2a—Materials and Methods for BTBR Mouse Model

Mice

Male BTBR mice were bred in house. The animals were housed in a temperature- and humidity-controlled room on a 12 hr dark cycle (lights on from 7:00-19:00 hr). All experiments were conducted in accordance with the European Directive 2010/63/EEC, the requirements of S.I. No 543 of 2012, and approved by the Animal Experimentation Ethics Committee of University College Cork.

Strain

MRX006: Blautia stercoris bacterium deposited under accession number NCIMB 42381.

Biotherapeutic was provided in glycerol stock. Live biotherapeutics were grown in the facility in anaerobic conditions.

Live Biotherapeutic Administration

Dosing with MRX006 or vehicle commenced when the mice were 8 weeks old. These mice were treated once daily with MRX006 or phosphate buffer solution (PBS) for 3 weeks before the beginning of the behavioural battery. Mice were further treated once daily during the behavioural battery. MRX006 (1×107 to 1×109 CFU oral administration) was dissolved in PBS prior to administration.

Administration Schedule

The treatment groups for the study are shown below. The vehicle for oral administration is PBS. Daily oral administration occurs via oral gavage.

Group Treatment Number 1 Control (PBS, oral gavage) 10 2 MRX006 (oral gavage in PBS) 10

Fecal Collection

Fresh fecal samples were collected from individual mice every week until the end of the study. At least 20 mg of fresh faeces were placed in a microcentrifuge tube, place immediately on ice and then stored at −80° C.

Experimental Design and Methods

As outlined above, dosing with MRX006 commenced when the mice were 8 weeks old. The initial dosing took place for 3 weeks before the behavioural experiments. The behavioural battery occurred in the following order: marble burying test at week 4; the elevated plus maze at week 5; the open field and novel object recognition tests, and the social transmission of food preference tests at week 6; the female urine sniffing and social interaction tests at week 7, and the forced swimming test at week 9. The carmine red gastrointestinal motility assay and gastrointestinal permeability assay tail bleeds occurred during weeks 8 and 9 respectively. Finally, in weeks 10 to 11, the mice were killed for splenocyte stimulation and ex vivo measurement of FITC in the ileum and colon.

The effects of live biotherapeutic treatment in the BTBR model on stereotyped, social and depression-like behaviours, along with gastrointestinal parameters (permeability and motility) are outlined in the following examples.

Group 1, listed in the table above, represents the control BTBR mice, which would be expected to show phenotypes associated with autistic spectrum disorders. Any effect of treatment on the behavioural symptoms of autistic spectrum disorders would be identified by differences between Group 1 and Group 2.

Graphical Design and Statistical Analysis

All graphs were generated on graphpad prism software (version 5). Data were analysed using IBM SPSS Statistic 22.0 (EEUU). Data distribution was analysed using the Kolmogorov-Smirnov normality test. Data comparing vehicle group versus the MRX006 group were analysed using one-way ANOVA and Fisher's least significant difference (LSD) post hoc test. If ANOVA did not reveal a significant effect of treatment, a priori pairwise comparisons test against the control group was conducted. Non-normally distributed data were analysed by the Kruskal-Wallis and non-parametric Mann-Whitney U test. P<0.05 was the criterion for statistical significance.

Example 2b—Assessment of Social Behaviours—the Three Chamber Social Interaction Test

Rationale

The 3-Chamber Social Interaction Test (3-CSIT) is a well validated ethologically relevant model that assesses social interaction between sex-matched conspecifics and allows for readouts of social novelty and social preference in mice. The test allows mice to freely explore between an inanimate object or sex-matched conspecific mice.

Methods

Animals are placed in a rectangular apparatus divided into three chambers (left and right and a smaller centre chamber) by partitions with small circular openings allowing easy access to all compartments. The test is composed of three sequential 10 min trials: (1) habituation (the test animal is allowed to explore the three empty chambers); (2) sociability (an unfamiliar animal is placed in an inner mesh wire cage in either the left or right chambers); (3) social novelty preference (a novel animal is placed into the previously empty inner cage in the chamber, opposite the now familiar animal). Naive animals should have no preference for either chamber in the habituation phase, a preference for the mouse in the sociability phase, and a preference for the novel mouse in the social novelty phase. An increase in the discrimination ratio would suggest an increase in social behaviour. All animals are age- and sex-matched, with each chamber cleaned and lined with fresh bedding after each 30 minute trial. For each of the three stages, behaviour is recorded by a video camera mounted above the apparatus.

Results

For assessing sociability, student's t-test within groups revealed an increased preference for a novel conspecific (CS) mouse relative to an object for the MRX006 group [t(22)=5.281; P<0.0001](FIG. 13A). For assessing social novelty, student's t-test within groups revealed an increased preference for a novel conspecific in the vehicle group only [t(22)=3.452; P<0.001]. ANOVA of interaction time with the novel conspecific did not reveal an effect of treatment [F(3,47)=2.492; P=0.43; FIG. 13B]. However, a priori pairwise comparisons test revealed that treatment with MRX006 (t(22)=0.7497; P=0.4614) decreased interaction time with a novel conspecific when compared to the vehicle group. ANOVA of percentage time spent investigating the novel conspecific revealed an effect of treatment [F(3,47)=2.942; P=0.0433; FIG. 13C]. Post-hoc comparisons revealed treatment with MRX006 decreased the percentage time investigating a novel conspecific (p<0.05).

Two way ANOVA analysis [Object/Conspecific (CS): [F(1,47)=21.164; P<0.0001; Treatment: F(1,47)=0.56; P=0.815; Object/CS×Treatment: F(1,47)=5.414; P=0.025] followed by post hoc analysis revealed that MRX006 treated mice spent more time investigating the conspecific versus the object (p<0.01; FIG. 13D). Two-way ANOVA analysis [Familiar vs novel: F(1,47)=3.454; P=0.070; Treatment: F(1,47)=0.360; P=0.552; F/Nx Treatment: F(1,47)=8.627; P=0.005] followed by post hoc comparisons revealed that mice treated with MRX006 spent significantly less time investigating a novel versus a familiar mouse (p<0.05; FIG. 13E). By contrast, vehicle mice spent more time investigating the novel versus the familiar conspecific (p<0.05; FIG. 13E). Percentage analysis revealed that MRX006 treated mice spent less time interacting with the novel conspecific when compared with the vehicle group (t=2.480 df=22; P=0.0213; FIG. 13F).

Conclusions

Chronic treatment with MRX006 reduces social novelty and decreases social cognition of BTBR mice in the three chamber test.

Example 2c—Assessment of Social Behaviours—Forced Intruder Test

Rationale

This procedure evaluates social interaction behaviour between rodents. By placing an intruder mouse into the resident mouse's home-cage, one can assess social interaction and aggressive behaviour.

Methods

Each session consisted of placing an intruder mouse into a resident mouse's home-cage for a period of 10 minutes. Experiments were videotaped using a ceiling camera to allow for measuring several behavioural parameters. The amount of time that the animals spent interacting was recorded.

Results

ANOVA of interaction time did not reveal an effect of treatment [F(3,45)=2.327; P=0.088; FIG. 14]. Similarly, a priori pairwise comparisons test revealed that treatment with MRX006 (t=1.425 df=22; P=0.1682) did not affect social interaction behaviour when compared to the vehicle group.

Conclusions

Chronic treatment with MRX006 does not influence social behaviour of BTBR mic in the social interaction test.

Example 2d—Assessment of Stereotyped Behaviours—the Marble Burying Test

Rationale

This test assesses for repetitive, compulsive and anxious behaviour. A higher number of marbles buried is indicative of greater anxious or stereotyped behaviours. Indeed, Mice treated with pharmacological agents such as anxiolytics show decreased marble burying behaviour, compared to the control mice.

Methods

Mice were individually placed into a novel polypropylene cage (35×28×18.5 cm, L×W×H), containing standard rodent (hard wood) bedding (5 cm) and 20 marbles on top of it (five rows of marbles regularly spaced 2 cm away from the walls and 2 cm apart). Experiments were conducted under a light intensity of 1000 lux. 30 minutes later, mice were removed from these cages and the number of marbles buried for more than ⅔rds of their surface was scored.

Results

There was no effect of treatment as determined by ANOVA on the number of marbles buried [F(3,45)=1.64; P=0.193]. However, a priori pairwise comparisons test revealed that MRX006 (t=2.276 df=21, p<0.05) decreased the number of marbles buried (FIG. 15).

Conclusions

Treatment with MRX006 reduces repetitive behaviour in BTBR mice in the marble burying test.

Example 2e—Assessment of Stereotyped Behaviours—the Grooming Test

Rationale

This test is used as an index for stereotyped and repetitive behaviour. An increase in time spent grooming is indicative of increased stereotyped or repetitive behaviour.

Methods

Mice were individually placed into a novel glass beaker (500 mL), which was covered with a Plexiglas top. Experiments were conducted under a light intensity of 60 lux. Experiments were videotaped using a hand-held camera attached to a tripod stand. Grooming behaviour was recorded for 20 minutes.

Results

There was a significant effect of live biotherapeutics as determined by ANOVA [F(3,47)=4.174; P=0.011] on grooming activity. Post-hoc comparisons revealed that treatment with MRX006 significantly reduced the amount of time spent grooming relative to the vehicle group (p<0.05) (FIG. 16). Similarly, a priori pairwise comparisons test revealed that MRX006 (t=2.895 df=22, p<0.01) decreased the time spent grooming compared to the vehicle group.

Conclusions

Chronic treatment with MRX006 reduces repetitive behaviours in BTBR mice in the grooming test.

Example 2f—Assessment of Anxiety-Like Behaviours—the Elevated Plus Maze

Rationale

The elevated plus maze (EPM) is a widely used test to assess anxiety-like behaviours in rodents. The EPM assesses general anxiety behaviour, with less anxious mice spending more time in the open arms of the maze. An increase in open arm activity (duration) reflects anti-anxiety behaviour.

Methods

The set up consisted of a grey plastic cross-shaped maze 1 meter elevated from the floor, comprising two open (aversive) and two closed (safe) arms (50×5×15 cm walls). Experiments occurred under red light (7 lux). Mice were placed into the centre of the maze facing an open arm (to avoid direct entrance into a closed arm) and were allowed to explore the arena for a duration of five minutes. Experiments were videotaped using a ceiling camera to allow for measuring several behavioural parameters. The percentage of time spent and the number of entries in each arm was measured for anxiety-like behaviour and locomotor activity, respectfully. Entrance into an arm was defined as all four paws inside the arm.

Results

ANOVA analysis revealed no effects of live biotherapeutic treatment on percentage of time spent in closed arms [F(3.47)=0.885; P=0.457; FIG. 17A). Kruskal Wallis non-parametric analysis of percentage time spent in open arms revealed no effect of treatment [chi-squared=1.220; df=3; P=0.748; FIG. 17B]. ANOVA of the number of entries into the closed arms revealed no effect of treatment [F(3,44)=1.82; P=0.159; FIG. 17C]. Kruskal Wallis non-parametric analysis of number of the entries into the open arms revealed no effect of treatment [chi-squared=2.045; df=3; P=0.563; FIG. 17D].

Conclusions

Chronic treatment with MRX006 has no effect on anxiety-like behaviour in BTBR mice in the elevated plus maze.

Example 2g—Assessment of Anxiety-Like Behaviours—the Open Field Arena

Rationale

The open field arena is used to assess the response of exposure to a novel stressful environment and locomotor activity. Naïve mice naturally spend most of their time alongside the walls of the arena, as it is less exposed than the centre of the arena. An increase in duration of time spent in the centre represents a decrease in anxiety-like behaviour.

Methods

Mice were individually placed into an open field arena (43×35×25, L×w×h) and allowed to explore for 10 minutes. Experiments occurred under a light intensity of 60 lux. Experiments were videotaped using a ceiling camera to allow for measuring several behavioural parameters using Ethovision software. The distance travelled was scored for locomotor activity.

Results

ANOVA of distance moved did not reveal an effect of treatment on locomotor activity in the open field arena [F(3,47)=0.317; P=0.813; FIG. 18A and FIG. 18D]. ANOVA of time spent in the outer zone did not reveal an effect of treatment [F(3,46)=2.217; P=0.100; FIG. 18B]. However, a priori pairwise comparisons test revealed that MRX006 treatment (t=2.791 df=21; p<0.05; FIG. 18E) decreased the time spent in the outer zone of open field arena. ANOVA of time spent in the inner zone did not reveal an effect of treatment [F(3,46)=2.217; P=0.100; FIG. 18C]. However, apriori pairwise comparisons test revealed that MRX006 treatment (t=2.791 df=21; p<0.05; FIG. 18F) increased the time spent in the inner zone of open field arena.

Conclusions

Chronic treatment with MRX006 reduces anxiety-like behaviour in BTBR mice in the open field arena test.

Example 2h—Assessment of Depression-Like Behaviour—the Forced Swim Test

Rationale

The forced swim test (FST) is the most widely used experimental paradigm to assess antidepressant activity. Naïve animals will display escape behaviour in the form of swimming, climbing and diving before adapting an immobile floating posture. The duration of immobility is indicative of behavioural despair. Antidepressant drugs decrease the time spent immobile in this test.

Methods

Mice are forced to swim for 6 min in a glass cylinder (24×21 cm) filled with 23-25° C. tap water to a depth of 17 cm. The FST was videotaped from a ceiling camera. The behavioural parameter scored is immobility during the last 4 min of the 6-min test.

Results

ANOVA of immobility time did not reveal an effect of treatment [F(3,46)=1.309; P=0.284; FIG. 19].

Conclusions

Chronic treatment with MXR006 does not influence immobility time of BTBR mice in the forced swimming test.

Example 2i—Assessment of Depression-Like Behaviour—the Female Urine Sniffing Test

Rationale

The female urine sniffing test (FUST) is used to assess anhedonic-like behaviour in rodents. A reduction in sniffing time suggests social avoidance/anhedonia while an increase represents an increase in social behaviour/hedonic behaviour.

Methods

Experimental mice are singly housed one week prior to the test. During the test, a cotton tip applicator, dipped in sterile water, is placed into the home cage and mice are allowed to sniff/investigate for three minutes. Following this three minute test, the cotton tip applicator is removed. 45 minutes later, a new cotton tip applicator is dipped into female urine (collected from female mice of the same strain that are in the estrous stage of their cycle), and placed into the cage. Mice are allowed to sniff/investigate this for a further three minutes. The amount of time spent sniffing the water and urine is recorded.

Results

For the vehicle group, student's t-test revealed a significant increase in the time spent sniffing urine relative to the time spent sniffing water [t(16)=2.611; P=0.0189; FIG. 20A]. For exposure to water, ANOVA of time spent sniffing did not reveal an effect of treatment in the water group [F(3,35)=0.875; P=0.464]. For exposure to urine, ANOVA of time spent sniffing did not reveal an effect of treatment [F(3,34)=2.153; P=0.114]. However, a priori comparison revealed that chronic treatment with MRX006 (t=3.602 df=16; P=0.0024) increased the time spent sniffing urine when compared to the vehicle group.

Two-way ANOVA analysis [Urine: [F(1,36)=44.118; P<0.0001]; Treatment: [F(1,36)=12.335; P=0.001]; Urine x treatment: [F(1,36)=9.236; P=0.005] followed by post hoc tests revealed that mice treated with MRX006 spent more time sniffing urine compared with the vehicle group (*p<0.01; FIG. 20B). Importantly, the vehicle mice spent more time sniffing urine than water as expected (#p<0.05).

Conclusions

Chronic treatment with MRX006 significantly increases the time spent sniffing female urine in BTBR mice in the female sniffing urine test.

Example 2j—Assessment of Depression-Like Behaviour—the Novel Object Recognition Test

Rationale

The protocol used was adapted from Bevins and Besheer (2006), and used to test recognition memory.

Improved memory is a reflection of reduced depression-like behaviour.

Methods

The protocol used was adapted from Bevins and Besheer (2006). It was conducted over 3 days. On Day 1, the animals were allowed to acclimate to the testing environment for 10 minutes, which was a large container equipped with an overhead camera. No bedding was used and the container was wiped with 70% ethanol between each animal. On Day 2, the animals were allowed to acclimate to the test apparatus for 10 minutes. Following this period, the animal was removed from the container and two identical objects were introduced to the environment. The animal was returned to the container and allowed to explore for a further 10 minutes. The objects were cleansed before each trial with a 70% ethanol solution. Following the training period, the rodent was removed from the environment for a delay period of 24 hours. On Day 3, the rodent was returned to the container, which this time contained only 1 familiar object from the day previous and 1 novel object. Activity of the animal with the 2 objects was recorded for 5 minutes. The amount of time that the rodent spent exploring each object was recorded by manual observation and a discrimination index (DI) value corresponding to time spent interacting with the novel object over total interaction time was generated. A decrease in DI compared to control rats indicates a deficit in this type of memory.

Results

Student t-test within groups did not reveal a side-preference for either object A or B on day one of the novel object recognition test (FIG. 21A). Student t-test within groups did not reveal a preference for the novel object relative to the familiar object. For the novel object, ANOVA of interaction time did not reveal an effect of treatment [F(3,46)=0.122; P=0.946; FIG. 21B]. In addition, no effect of treatment on discrimination index was revealed by ANOVA analysis [F(3,47)=0.535; P=0.661; FIG. 21C].

Conclusions

Chronic treatment with MRX006 has no effect on cognitive behaviour in BTBR mice in the novel object recognition test.

Example 2k—In Vivo Gastrointestinal Permeability Assay

Rationale

This procedure is used to assess in vivo intestinal motility.

Methods

Test mice were singly-housed and fasted overnight. The following morning (˜9 am), mice were administered FITC dextran ((600 mg/kg) by oral gavage. Two hours later, 100 μl of blood sample was collected in heparin-coated capillary tubes and transferred to a darkened eppendorf and placed on ice. Samples were centrifuged 3500×g for 15 minutes, plasma was aspirated and samples were stored at −80° D for long storage.

Undiluted plasma was used to quantify FITC concentration. 25 μl of FITC was pipetted in duplicated in 384 well plate (Greiner bio one). FITC was measured with a Victor spectrometer between the ranges of 490 nm-520 nm. For a standard curve, a serial dilution of FITC was prepared in PBS (pH7.4). An increase in absorbance is indicative of a decrease in barrier integrity.

Results

Intestinal barrier function was assessed through oral administration of the fluorescent compound, fluorescein isothiocyante (FITC), followed by subsequent tail bleeds to assess levels of FITC in plasma. ANOVA of FITC concentrations did not reveal a significant effect of treatment [F(3,47)=1.366; P=0.266; FIG. 23].

Conclusion

Chronic treatment with MRX006 did not influence intestinal permeability in BTBR mice.

Example 2l—Ex Vivo Gastrointestinal Permeability Assay

Rationale and Methods

The permeability of the ileum and colon was assessed ex vivo using Ussing chambers. Colon and ileum were excised from mice and collected into 5 mL tubes containing Kreb's buffer. Both colon and ileum were cut along the mesenteric line and mounted onto the Ussing chamber apparatus. For colon, 4 mLs of Krebs solution containing D-glucose were added into both sides of the Ussing chamber apparatus. For ileum, 4 mLs of Krebs solution containing D-mannitol was added into the muscosal side, while an equal volume of Krebs with D-glucose was added to the serosal side. The chambers were oxygenated with carbogen gas (95% O2 and 5% CO2) and kept at 37° C. to maintain tissue integrity. 2.5 mg/mL FITC-dextran was added to the mucosal chamber. Samples were taken from the serosal chamber at timepoints 0 min (baseline), 60 min, 90 min and 120 mins. 25 μl of FITC was pipetted in duplicated in 384 well plate (Greiner bio one). FITC was measured with a Victor spectrometer between the ranges of 490 nm-520 nm. For a standard curve, a serial dilution of FITC was prepared in PBS (pH7.4).

Results

In the ex vivo intestinal permeability assay, repeated measures ANOVA revealed an effect of time for both the colon [F(3,87)=64.197; P<0.0001] and the ileum [F(3,87)=34.572; P<0.0001]. Repeated measures ANOVA did not reveal an effect of treatment with respect to time for either the colon [F(9,87)=1.184; P=0.316; FIG. 23A] or ileum [F(9,87)=0.810; P=0.609; FIG. 22B].

Conclusions

Chronic treatment with MRX006 does not influence the permeability of the colon or ileum.

Example 2m—In Vivo Gastrointestinal Motility Assay

Rationale

This procedure is used to assess in vivo intestinal motility.

Methods

Mice are singly housed prior to the commencement of the test. Mice were orally gavaged with a non-absorbable, coloured dye (Carmine Red). The time to excretion of the first coloured faecal bolus was recorded and used as an index of peristaltic motility of the whole intestine.

Results

Mice were administered a non-absorbable, coloured dye (Carmine Red) by oral gavage. The time to excretion of the first coloured faecal bolus was recorded and used as an index of peristaltic motility of the whole intestine. ANOVA of motility time revealed no effect of treatment [F(3,47)=2.097; P=0.114]. However, a priori pairwise comparisons test revealed that mice treated with MRX006 (t=2.270 df=22, p<0.05; FIG. 24) display altered intestinal motility when compared to the vehicle group.

Conclusions

Chronic treatment with MRX006 showed reduced intestinal motility in BTBR mice.

Example 2n—Stress-Induced Circulating Corticosterone Determination

Rationale

Exposure to the FST results in a robust activation of the HPA axis, with an increase in the levels of the stress hormone, corticosterone. Plasma corticosterone concentrations taken prior to, and after exposure to the FST, were used as an index of stress-induced activation of the hypothalamic pituitary adrenal (HPA) axis.

Methods

On the day of the FST, mice were removed from their home-cage and moved to a surgical room where a basal blood sample was taken. A scalpel blade was used to remove the very tip (1 mm) of the tail. Blood was the collected using a heparinised capillary tube and then transferred to a microcentrifuge tube. Blood samples were also taken 30, 60, 90 and 120 minutes following exposure to the FST to assess for peak and recovery corticosterone levels. Blood was kept on ice and then centrifuged at 2,500×g for 15 minutes at 4° C. Plasma corticosterone was assessed by ELISA, following vendor instructions (ENZO Corticosterone ELISA, ADI-900-097, Enzo Life Sciences).

Results

Repeated measures ANOVA revealed a significant effect of time [F(4,164)=127.127; P<0.0001; FIG. 25]. Post-hoc comparisons revealed a significant increase in circulating corticosterone at the 30-minute time point for all groups. Repeated measures ANOVA did not reveal a significant effect of treatment with respect to time [F(12,164)=0.561; P=0.871].

Conclusions

Chronic treatment with MRX006 does not influence stress-induced corticosterone levels in BTBR mice exposed to the forced swimming test.

Example 2o—Organ Weight and Colon Length

ANOVA of organ weight as a percentage of body weight did not reveal an effect of treatment for the adrenals [F(3,44)=1.480; P=0.234; FIG. 26A}, spleen [F(3,43)=0.779; P=0.513; FIG. 26B] or caecum [F(3,44)=0.441; P=0.725; FIG. 26C]. ANOVA of colon length did not reveal an effect of treatment [F(3,46)=0.826; P=0.487; FIG. 26D].

Conclusions

Treatment with MRX006 does not influence selective anatomical markers.

Example 2p—Weight Monitoring

Animal body weights were assessed once per week over the duration of the experiment to determine whether any of the bacterial strains were influencing this particular parameter. Repeated measures ANOVA revealed a significant effect of time [F(11,484)=111.217; P<0.0001; FIG. 27]. Repeated measures ANOVA did not reveal an effect of treatment with respect to time [F(33,484)=0.581; P=0.971].

Conclusions

Chronic treatment with MRX006 does not influence body weight in BTBR mice.

Conclusions from the BTBR Mouse Model

The main findings of this study were that treatment with MRX006 attenuated stereotyped and anxiety-related behaviours. Specifically, MRX006 reduced the number of marbles buried in the marble burying test as well as reducing the amount of time that animals spent grooming. Moreover, treatment with this live biotherapeutic increased the amount of time spent in the centre of the open field, corresponding with a decrease in the amount of time spent in the periphery, which is indicative of an anxiolytic-like effect. However, no effects on anxiety-like behaviour were observed in the elevated plus maze test. The ability of MRX006 to improve stereotyped and anxiety related behaviours in BTBR mice is promising and indicates that it may be an effective therapeutic.

MRX006 also increased the time spent sniffing urine from female mice. The female urine sniffing test was originally designed as a test to assess hedonic-like behaviour in rodents, with increases in the time spent sniffing urine interpreted as an increase in reward seeking behaviour (Malkesman et al., 2010). Given that BTBR mice are not reported to display a depressive-like phenotype, it is unlikely that the observed increase in time spent sniffing urine in the current experiments following treatment with MRX006 reflects an improvement in hedonic behaviour. Rather, it may be that MRX006 is increasing the ability of these mice to recognise and process social information (i.e. female pheromones). However, no differences in social behaviour among the groups were observed in the 3 chamber test and social interaction test. Treatment with MRX006 reduced the amount of time that mice spent investigating a novel conspecific mouse relative to a familiar conspecific.

With the exception of intestinal motility, the live biotherapeutic assessed in the current study did not affect the several physiological parameters measured. For instance, no effect of the live biotherapeutic was observed in stress-induced corticosterone secretion, anatomical weight, intestinal permeability or total body weight. In the intestinal motility assay, treatment with MRX006 prolonged the time taken for the first red pellet to appear following oral gavage with carmine red dye. Such results suggest that MRX006 prolongs intestinal motility.

Example 3—the Maternal Immune Activation (MIA) Mouse Model

The MIA mouse model uses an environmental immune challenge in pregnant mice in order to trigger the core symptoms of autism spectrum disorder in the offspring. MIA mice typically display stereotyped behaviour (as shown by the grooming and marble burying tests) and deficits in social communication (as shown by the social play, 3-chamber social interaction, and social transmission of food preference tests). The offspring display the three core symptoms of autism (reduced communication; reduced sociability; and increased repetitive or stereotyped behaviour) and therefore provide a suitable model in which to determine whether administration of a therapeutic can alleviate the behavioural phenotypes associated with autistic spectrum disorders and indeed in a number of other neurological disorders. It is well established that alteration of behavioural phenotypes in animal models is indicative of a potentially clinically relevant intervention, irrespective of an understanding of the underlying biological or physiological mechanism (Crawley 2012).

Example 3a—Materials and Methods for MIA Mouse Model

Mice

Maternal immune activation (environmental ASD mouse model) protocol was conducted as previously described (Hsiao, McBride et al. 2013). Briefly, pregnant C57BL/6N mice (ENVIGO, UK) were injected i.p. on E12.5 with saline or 20 mg/kg poly(I:C) according to methods described in (Hsiao, McBride et al. 2013). These mice are listed in the experiments below as MIA mice. Male mice started behaviour at 8 weeks old. The animals were housed in a temperature- and humidity-controlled room on a 12 hr dark cycle (lights on from 7:00-19:00 hr). All experiments were conducted in accordance with the European Directive 2010/63/EEC, the requirements of S.I. No 543 of 2012, and approved by the Animal Experimentation Ethics Committee of University College Cork.

Strain

MRX006: Blautia stercoris bacterium deposited under accession number NCIMB 42381.

Live biotherapeutics were grown in the facility in anaerobic conditions.

Live Biotherapeutic Administration

Dosing with MRX006 or vehicle commenced when the mice were 8 weeks old. These mice were treated once daily with MRX006 or phosphate buffer solution (PBS) for 3 weeks before the beginning of the behavioural battery. Mice were further treated once daily for 5 weeks during the behavioural battery. MRX006 (1×107 to 1×109 CFU oral administration) was dissolved in PBS prior to administration.

Administration Schedule

The treatment groups for the study are shown below. The vehicle for oral administration is PBS. Daily oral administration occurs via oral gavage.

Group Treatment Number 1 Control (PBS, oral gavage) 9 2 Vehicle MIA (PBS, oral gavage) 15 3 MRX006 MIA (oral gavage in PBS) 13

Fecal Collection

Fresh fecal samples were collected from individual mice every week until the end of the study. At least 20 mg of fresh faeces were placed in a microcentrifuge tube, place immediately on ice and then stored at −80° C.

Experimental Design and Methods

As outlined above, dosing with MRX006 or vehicle commenced when the mice were 8 weeks old. The behavioural battery occurred in the following order: the open field arena at week 4, the marble burying test at week 5; social transmission of food preference test at week 6, and the female urine sniffing test at week 7. The carmine red gastrointestinal motility assay and gastrointestinal permeability assay tail bleeds occurred during weeks 7 and 8 respectively. Finally, in week 9, the mice were killed for splenocyte stimulation and ex vivo measurement of FITC in the ileum and colon.

The effects of live biotherapeutic treatment in the MIA model on stereotyped, social and depression-like behaviours, along with gastrointestinal parameters (permeability and motility) are outlined in the following examples.

Group 2, listed in the table above, represents the maternal immune activation mice, the mothers of which were treated with poly (I:C) during pregnancy. These mice would be expected to show phenotypes associated with autistic spectrum disorders compared to the control mice (Group 1)—this control ensures that the poly (I:C) administration did cause the expected behavioural symptoms in the maternal mouse offspring. Any effect of treatment on the behavioural symptoms of autistic spectrum disorders would be identified by differences between Group 2 and Group 3.

Graphical Design and Statistical Analysis

All graphs were generated on graphpad prism software (version 5). Data were analysed using IBM SPSS Statistic 22.0 (EEUU). Data distribution was analysed using the Kolmogorov-Smirnov normality test. Data comparing vehicle group versus the MRX006 group were analysed using one-way ANOVA and Fisher's least significant difference (LSD) post hoc test. If ANOVA did not reveal a significant effect of treatment, a priori pairwise comparisons test against the control group was conducted. Non-normally distributed data were analysed by the Kruskal-Wallis and non-parametric Mann-Whitney U test. P<0.05 was the criterion for statistical significance.

Example 3b—Assessment of Stereotyped Behaviours—the Marble Burying Test

Rationale

This test assesses for repetitive, compulsive and anxious behaviour. A higher number of marbles buried is indicative of greater anxious or stereotyped behaviours. Indeed, Mice treated with pharmacological agents such as anxiolytics show decreased marble burying behaviour, compared to the control mice.

Methods

Mice were individually placed into a novel polypropylene cage (35×28×18.5 cm, L×W×H), containing standard rodent (hard wood) bedding (5 cm) and 20 marbles on top of it (five rows of marbles regularly spaced 2 cm away from the walls and 2 cm apart). Experiments were conducted under a light intensity of 1000 lux. 30 minutes later, mice were removed from these cages and the number of marbles buried for more than ⅔rds of their surface was scored.

Results

Student's t-test analysis between the control group and the vehicle MIA group revealed that the vehicle MIA mice buried more marbles compared to the control group (t(21)=2.751, P=0.011; FIG. 28). ANOVA of the number of marbles buried revealed a significant effect of treatment [F(3,48)=18.39; P<0.001]. Post-hoc comparisons revealed that chronic treatment with MRX006 decreased the number of marbles buried (p<0.01; FIG. 28).

Conclusions

The vehicle MIA group showed significantly more marbled buried than the control group, indicating that the MIA model successfully triggered autistic spectrum disorder-like symptoms in the mice. Chronic treatment with MRX006 reduces repetitive, compulsive and anxious behaviour in MIA mice.

Example 3c—Assessment of Social Behaviours—Social Transmission of Food Preference

Rationale

Social transmission food preference is a relevant test of olfactory memory that is used in mice to assess social behaviour. In this test, observer mice interact with a demonstrator mouse that has recently eaten novel food. When observer mice are presented with a choice between the food eaten by the demonstrator and some other novel food, observer mice should prefer the food eaten by the demonstrator. Reduced food preference would indicate reduced sociability.

Methods

This test was performed as previously described (Desbonnet, Clarke et al. 2015). Briefly, 18 hours prior to testing, mice were deprived of food, whereas water was available ad libitum. Food choices consisted of either 1% ground cinnamon or 2% powdered cocoa made with grounded mouse chow. A demonstrator mouse was randomly selected from each cage and the tail was marked using a blue marker to enable identification during subsequent social interactions. Demonstrator food containers were weighed before and after the 1 hour sampling sessions. A minimum of 0.2 g of consumed food was required for inclusion in the test. Demonstrator mice were placed back into their respective home cages for a 30 minute interaction period with cage-mates. Subsequently, cage-mates were individually tested for preference of cued food or novel food. Containers were weighed immediately before and after each choice session. Observed mice were then placed back into their respective home cages and the choice session was repeated 24 hours later. The test mice should smell the cinnamon or cocoa off the demonstrator mouse as a social cue, and preferentially choose the same food when given a choice between the two.

Results

Student's t-test of percentage food preference revealed no difference between control and vehicle MIA groups at either the 0 hour (t(22)=0.3325, P=0.7427) or 24 hour (t(21)=0.2878, P=0.7763) assessment. ANOVA of demonstrator cued food preference revealed no significant difference when observers were exposed to food choice at the 0 hour [F(3,48)=1.49, P=0.228; FIG. 29A] or 24 hour assessment [F(3,47)=2.66, P=0.059; FIG. 29B]. Treatment with MRX006 did not alter preference for cued food in the social transmission of food preference test.

Conclusions

The vehicle MIA group did not display reduced social transmission food preference (the MIA vehicle displayed no alteration in food preference compared to the control), suggesting the MIA model has not triggered the reduced sociability phenotype. Chronic treatment with MRX006 had not effect on food preference. However, as the MIA model appears not to have caused a reduced sociability phenotype in this test, it is not possible to determine the effects of chronic treatment with MRX006 on sociability in the social transmission of food preference test.

Example 3d—Assessment of Anxiety-Like Behaviour—the Open Field Arena

Rationale

The open field arena is used to assess the response of exposure to a novel stressful environment and locomotor activity. Naïve mice naturally spend most of their time alongside the walls of the arena, as it is less exposed than the centre of the arena. An increase in duration of time spent in the centre represents a decrease in anxiety-like behaviour.

Methods

Mice were individually placed into an open field arena (43×35×25, L×w×h) and allowed to explore for 10 minutes. Experiments occurred under a light intensity of 60 lux. Experiments were videotaped using a ceiling camera to allow for measuring several behavioural parameters using Ethovision software. The distance travelled was scored for locomotor activity.

Results

Student's t-test revealed no significant difference in the total distance moved between control and vehicle MIA groups (t(22)=0.9357, P=0.3596). ANOVA of total distance moved revealed a significant effect of treatment [F(3,47)=4.36, P=0.003, FIG. 30A]. Post hoc comparisons revealed that treatment with MRX006 reduced total distance travelled relative to vehicle treated animals (p<0.05). Student's t-test revealed a significant increase in the time spent in the outer zone of the open field by the vehicle MIA group relative to the control group (t(21)=3.337, P=0.003). ANOVA of time spent in the outer zone of the open field revealed no effect of treatment [F(3,47)=0.093, FIG. 30B]. Student's t-test revealed a significant decrease in the time spent in the inner zone by the vehicle MIA groups relative to the control mice (t(21)=3.337, P=0.003). ANOVA of time spent in the inner zone revealed no effect of treatment [F(3,47)=0.93, P=0.96, FIG. 30C].

Conclusions

Treatment with MRX006 decreases the distance travelled by MIA mice in the open field arena. Therefore, MRX006 may be attenuating stress-induced locomotor activity caused by exposure to the open field arena.

Example 3e—Assessment of Depression-Like Behaviour—the Female Urine Sniffing Test

Rationale

The female urine sniffing test (FUST) is used to assess anhedonic-like behaviour in rodents. A reduction in sniffing time suggests social avoidance/anhedonia while an increase represents an increase in social behaviour/hedonic behaviour.

Methods

Experimental mice are singly housed one week prior to the test. During the test, a cotton tip applicator, dipped in sterile water, is placed into the home cage and mice are allowed to sniff/investigate for three minutes. Following this three minute test, the cotton tip applicator is removed. 45 minutes later, a new cotton tip applicator is dipped into female urine (collected from female mice of the same strain that are in the estrous stage of their cycle), and placed into the cage. Mice are allowed to sniff/investigate this for a further three minutes. The amount of time spent sniffing the water and urine is recorded.

Results

Mann-Whitney U-test revealed that both control [Mann-Whitney U value=7, P=0.0123] and vehicle MIA [Mann Whitney U value=57; P=0.0201] groups spent more time sniffing urine than water (FIG. 31). For time spent sniffing urine, Kruskal-Wallis non-parametric analysis did not reveal an effect of treatment [df=4, P=0.3293].

Conclusions

Chronic treatment with MRX006 does not influence depressive-like behaviour in MIA mice in the female sniffing urine test.

Example 3f—In Vivo Intestinal Motility Assay

Rationale

The MIA model has been reported to lead to changes in gut barrier function. Therefore, it was important to ascertain whether chronic treatment with the biotherapeutic alters intestinal motility.

Methods

Mice are singly housed prior to the commencement of the test. Mice were orally gavaged with a non-absorbable, coloured dye (Carmine Red). The time to excretion of the first coloured faecal bolus was recorded and used as an index of peristaltic motility of the whole intestine.

Results

Student's t-test analysis revealed that vehicle MIA group do not exhibit altered intestinal motility (red pellet detected in less time) when compared to the control group (t(22)=0.006, P=0.9950). ANOVA of motility time revealed no effect of treatment [F(3,50)=0.99; P=0.404, FIG. 32].

Conclusions

In this experiment, the vehicle MIA group did not exhibit altered intestinal motility compared to the control. Chronic treatment with MRX006 did not affect intestinal motility compared to the control or vehicle MIA groups.

Example 3g—Organ Weight and Colon Length

For colon length, student's t-test did not reveal any significant difference between vehicle MIA and control groups (t(21)=1.26, P=0.26). ANOVA of colon of length did not reveal an effect of treatment [F(3,49)=0.69, P=0.57; FIG. 33A]. For caecum weight as a percentage of body weight, student's t-test did not reveal a significant difference between vehicle MIA and control groups (t(22)=0.56, P=0.58). ANOVA revealed no significant effect of treatment upon caecum weight [F(3,48)=0.84, P=0.48, FIG. 33B]. For spleen weight as a percentage of body weight, student's t-test did not reveal a significant difference between vehicle MIA and control groups (t(22)=0.64, P=0.53). ANOVA revealed no significant effect of treatment upon spleen weight [F(3,48)=2.25, P=0.09, FIG. 33C).

Conclusions

Treatment with MRX006 does not influence colon length or organ weight in the MIA mouse model of autism.

Discussion of Results from the MIA Mouse Model

Chronic treatment with MRX006 was able to reverse the phenotype observed in the marble burying test in MIA mice. Chronic treatment with MRX006 was able to reduce the number of marbles buried suggesting a reduction in stereotyped-like behaviour. Furthermore, chronic treatment with MRX006 decreased the distance travelled without having any effect on time spent in the inner and outer zones in the open field arena. Consequently, treatment with MRX006 may attenuate stress-induced locomotor activity caused by exposure to the open field arena. No significant effect of treatment was observed in the social transmission food and the female urine sniffing tests suggesting no directly observable effects in social and depressive-like behaviour in the MIA mouse model. The live biotherapeutic tested did not affect intestinal motility or permeability. Therefore, the MIA model has proven useful for assessing stereotyped-like, repetitive and anxious behaviour, but it did not recreate a number of other symptoms associated with autistic spectrum disorders. Nevertheless, the results display that chronic treatment with MRX006 may have a positive impact on the symptoms of autistic spectrum disorders.

Overall Conclusions Regarding MRX006 in the Treatment of Autistic Spectrum Disorders

MRX006 was shown to be effective in the treatment of stereotyped and anxiety-like behaviours in both the BTBR and MIA mouse model. Therapies that reverse behavioural and biological phenotypes in mouse models of autism are expected to be effective against human disease.

The EMA Guidelines on the clinical development of medicinal products for the treatment of autism spectrum disorder state that, due to the heterogeneity of the diseases, it may not be possible to achieve a significant effect on all core symptoms with a single compound, and so short term efficacy has to be demonstrated on at least one core symptom. The MRX006 live biotherapeutic has shown effective treatment of at least one core symptom of autistic spectrum disorder, so it and related Blautia and B. stercoris strains are expected to be effective against human disease. Similarly, other central nervous system disorders or conditions display complex pathology with multiple different symptoms, and have few approved treatments. Therefore, it is understood that an effective treatment does not need to treat all symptoms of a central nervous system disorder or condition. A treatment would be considered therapeutically useful if it treated one of the symptoms associated with a central nervous system disorder or condition.

Example 4—Measurement of Circulating Cytokines in BTBR Mice

Methods and Results

Blood plasma was collected from trunk blood on the day of the culls from each animal at the end of the experiments. Circulating cytokines were assessed in plasma samples from vehicle and MRX006 groups using a commercially available electrochemiluminescence multiplex system (MSD, Gaithersburg, MSD, USA). The following cytokines were assayed for: IL-1β, IL-4, IL-6, IL-10, IL-17A, IL-21, IL-23, TNF-α and IFN-γ. Multiplex analysis revealed that levels of IL-1β, IL-4, IL-17A, IL-21 and IL-23 were below the limits of detection in both vehicle and MRX006 treated animals. For circulating TNF-α, student's t-test did not reveal a significant effect of treatment with MRX006 (t(21)=0.4264, P=0.6742, FIG. 34A). For circulating IFN-, student's test did not reveal a significant effect of treatment with Mrx006 (t(17)=0.4103, P=0.6867, FIG. 34B). For circulating IL-6, Student's t-test did not reveal a significant effect of treatment with MRX006 (t(11)=0.020, P=0.98, FIG. 34C). For circulating IL-10, chronic treatment with MRX006 causes a non-significant increase in IL-10 levels (t(13)=1.396, P=0.1861, FIG. 34D).

Conclusions

While there was no significant effect of MRX006 in terms of regulating circulating cytokine concentrations, there was a clear non-significant trend for an increase in circulating IL-10 following treatment with the live biotherapeutic. Such results suggest that MRX006 possesses immune-regulatory properties and can increase the production of anti-inflammatory cytokines. While the multiplex assay was performed upon plasma samples that contained basal, unstimulated cytokine concentrations, it will be interesting to assess whether MRX006 is capable of modulating IL-10 and other cytokines under stimulated conditions.

Example 5—Assessing the Effects of Subchronic Treatment with MRX006 Upon Central and Peripheral Oxytocin Levels in C57BL/6 Mice

The bacterial strains were prepared and administered as outlined in the Examples above. The C57BL/6 mice were treated with live biotherapeutic for six days in 7 experimental treatment groups each with 10-12 mice. Subsequently, the hypothalamus was dissected from the mice and the levels of oxytocin in the hypothalamus were detected by radioimmunoassay (RIA), In addition, levels of oxytocin in the plasma were detected by RIA. Furthermore, the levels of oxytocin receptors, interleukins and other inflammatory markers, and vasopressin hormones were detected by RIA and other analytical methods.

Example 6—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 atmosphere 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 7—Administration of Another Live Biotherapeutic in the MIA Mouse Model Example 7a—Materials and Methods for MIA Mouse Model

The mice, live biotherapeutic administration and fecal collection used in this Example are identical to those used in Example 3 above.

Strain

MRX008: Blautia wexlerae, bacteria deposited under accession number NCIMB 42486.

Administration Schedule

The treatment groups for the study are shown below. The vehicle for oral administration is PBS. Daily oral administration occurs via oral gavage.

Group Treatment Number 1 Control (PBS, oral gavage) 11 2 Vehicle MIA (PBS, oral gavage) 10 3 MRX008 MIA (oral gavage in PBS) 11

Experimental Design and Methods

Dosing with MRX008 or vehicle commenced when the mice were 8 weeks old. The behavioural battery occurred in the following order: marble burying test at week 5; social transmission of food preference at week 6 and the forced swimming test at week 8. The carmine red gastrointestinal motility assay and gastrointestinal permeability assay tail bleeds occurred during weeks 7 and 8 respectively. Finally, in week 9, the mice were killed for splenocyte stimulation and ex vivo measurement of FITC in the ileum and colon.

The effects of live biotherapeutic treatment in the MIA model on stereotyped, social and depression-like behaviours, along with gastrointestinal parameters (permeability and motility) are outlined in the following examples.

Group 2, listed in the table above, represents the maternal immune activation mice, the mothers of which were treated with poly (I:C) during pregnancy. These mice would be expected to show phenotypes associated with autistic spectrum disorders compared to the control mice (Group 1)—this control ensures that the poly (I:C) administration did cause the expected behavioural symptoms in the maternal mouse offspring. Any effect of treatment on the behavioural symptoms of autistic spectrum disorders would be identified by differences between Group 2 and Group 3.

Graphical Design and Statistical Analysis

All graphs were generated on graphpad prism software (version 5). Data were analysed using IBM SPSS Statistic 22.0 (EEUU). Data distribution was analysed using the Kolmogorov-Smirnov normality test. Data comparing vehicle group versus the MRX008 group were analysed using one-way ANOVA and Fisher's least significant difference (LSD) post hoc test. If ANOVA did not reveal a significant effect of treatment, a priori pairwise comparisons test against the control group was conducted. Non-normally distributed data were analysed by the Kruskal-Wallis and non-parametric Mann-Whitney U test. P<0.05 was the criterion for statistical significance.

Example 7b—Assessment of Stereotyped Behaviours—the Marble Burying Test

Rationale and Methods

See Example 3b above.

Results

Student's t-test analysis between the control group and the vehicle MIA group revealed that the vehicle MIA mice buried more marbles compared to the control group (t(19)=3.00, P=0.007; FIG. 35). ANOVA of the number of marbles buried revealed an effect of treatment [F(3,42)=6.37, P=0.001]. Chronic treatment with MRX008 showed a reduction in the number of marbles buried relative to the vehicle MIA group.

Conclusions

The vehicle MIA group showed significantly more marbles buried than the control group, indicating that the MIA model successfully triggered autistic spectrum disorder-like symptoms in the mice. There is a trend towards a reduction in repetitive, compulsive and anxious behaviour in MIA mice upon chronic treatment with MRX008.

Example 7c—Assessment of Social Behaviours—Social Transmission of Food Preference

Rationale and Methods

See Example 3c above.

Results

ANOVA of demonstrator cued food preference revealed no significant difference when observers were exposed to food choice immediately after demonstrator interaction (T0) (F(3,34)=0.38, P=0.77; FIG. 36A) or 24 hrs later (F(3,34)=0.85, P=0.48; FIG. 36B), irrespective of vehicle or MRX008 administration.

Conclusions

The vehicle MIA group did not display reduced social transmission food preference (the MIA vehicle displayed no alteration in food preference compared to the control), suggesting the MIA model has not triggered the reduced sociability phenotype. Accordingly, it is not possible to determine the effects of chronic treatment with MRX008 on sociability using the MIA mouse model.

Example 7d—Assessment of Depression-Like Behaviours—the Forced Swimming Test

Rationale

The forced swim test (FST) is the most widely used experimental paradigm to assess antidepressant activity ([57]). In this test, mice are forced to swim for 6 min and the behavioural parameter scored is immobility during the last 4 min of the 6-min test. Naïve animals will display escape behaviour in the form of swimming, climbing and diving before adapting an immobile floating posture. The duration of immobility is indicative of behavioural despair. Antidepressant drugs decrease the time spent immobile in this test.

Methods

Mice are forced to swim for 6 min in a glass cylinder (24×21 cm) filled with 23-25° C. tap water to a depth of 17 cm. The FST was videotaped from a ceiling camera. The behavioural parameter scored is immobility during the last 4 min of the 6-min test.

Results

Student's t test analysis revealed no significant differences on immobility time between the control group and vehicle MIA group (t=0.8968 df=20; 0.3805). ANOVA of immobility time did not reveal an effect of treatment with MRX008, although there appears to be a slight trend towards a reduction in time spent immobile after administration of MRX008 [F(3,42)=1.803; P=0.1625; FIG. 37].

Conclusions

The vehicle MIA group did not display increased immobility time in the forced swim test (the MIA vehicle displayed no alteration in immobility time compared to the control), suggesting the MIA model has not increased depressive-like symptoms. Accordingly, it is not possible to determine the effects of chronic treatment with MRX008 on depressive-like behaviour using the MIA mouse model.

Example 7e—In Vivo Intestinal Permeability Assay

Rationale

The MIA model has been reported to lead to changes in gut barrier function. Therefore, it was important to ascertain whether chronic treatment with the biotherapeutic affects intestinal permeability.

Methods

Test mice were single caged and food was removed overnight. Next day (at around 9 am) mice were administered by oral gavage with FITC dextran (Fluroscein-isothiocynate; MW: 4 kDa, Sigma; concentration: 600 mg/kg per animal of 80 mg/ml FITC in PBS (pH7.4)). Two hours following FITC administration, 100 μl of blood sample, from tail bleeds, was collected in heparin-coated capillary tubes and transferred to amber eppendorf and placed on ice. Samples were centrifuged 3500×g for 15 minutes, plasma was aspirated and samples were stored at −80° D for long storage.

Undiluted plasma was used to quantify FITC concentration. 25 μl of FITC was pipetted in duplicated in 384 well plate (Greiner bio one). FITC was measured with a Victor spectrometer between the ranges of 490 nm-520 nm. For a standard curve, a serial dilution of FITC was prepared in PBS (pH7.4).

In addition, after the cull of the mice in week 9, ex vivo measurements of FITC in the ileum and colon are performed.

Results

Student's t test analysis revealed no differences between the control group and the MIA vehicle group (t(20)=0.56, P=0.58; FIG. 38). ANOVA of FITC concentrations did not reveal a significant effect of treatment [F(3,39)=2.23, P=0.08].

Conclusions

In this experiment, the vehicle MIA group did not display altered intestinal permeability (the MIA vehicle displayed no alteration in permeability compared to the control). Furthermore, chronic treatment with MRX008 did not affect intestinal permeability in MIA mice.

Example 7f—In Vivo Intestinal Motility Assay

Rationale and Methods

See Example 3f above.

Results

Student's t test analysis revealed that the vehicle MIA group exhibited increased intestinal motility (red pellet detected in less time) when compared to the control group (t(19)=3.00, P=0.007). ANOVA of motility time revealed no effect of treatment [F(3,38)=0.74, P=0.54; FIG. 39].

Conclusions

In this experiment, the vehicle MIA group displayed increased intestinal motility compared to the control. Chronic treatment with MRX008 did not affect intestinal motility compared to the control.

Example 8—Administration of Another Live Biotherapeutic in the BTBR Mouse Model Example 8a—Materials and Methods for BTBR Mouse Model

The mice, live biotherapeutic administration and fecal collection used in this Example are identical to those used in Example 2 above.

Strain

MRX008: Blautia wexlerae, bacteria deposited under accession number NCIMB 42486.

Administration Schedule

The treatment groups for the study are shown below. The vehicle for oral administration is PBS. Daily oral administration occurs via oral gavage.

Group Treatment Number 1 Control (PBS, oral gavage) 10 2 MRX008 (oral gavage in PBS) 10

Experimental Design and Methods

As outlined above, dosing with MRX008 commenced when the mice were 8 weeks old. The initial dosing took place for 3 weeks before the behavioural experiments encompassing tests of sociability, anxiety, stereopathy and cognition. The behavioural battery occurred in the following order: marble burying test at week 4; the elevated plus maze at week 5; the open field and novel object recognition tests, and the social transmission of food preference tests at week 6; the female urine sniffing and social interaction tests at week 7, and the forced swimming test at week 9. The carmine red gastrointestinal motility assay and gastrointestinal permeability assay tail bleeds occurred during weeks 8 and 9 respectively. Finally, in weeks 10 to 11, the mice were killed for splenocyte stimulation and ex vivo measurement of FITC in the ileum and colon.

The effects of live biotherapeutic treatment in the BTBR model on stereotyped, social and depression-like behaviours, along with gastrointestinal parameters (permeability and motility) are outlined in the following examples.

Group 1, listed in the table above, represents the control BTBR mice, which would be expected to show phenotypes associated with autistic spectrum disorders. Any effect of treatment on the behavioural symptoms of autistic spectrum disorders would be identified by differences between Group 1 and Group 2.

Graphical Design and Statistical Analysis

All graphs were generated on graphpad prism software (version 5). Data were analysed using IBM SPSS Statistic 22.0 (EEUU). Data distribution was analysed using the Kolmogorov-Smirnov normality test. Data comparing vehicle group versus the MRX008 group were analysed using one-way ANOVA and Fisher's least significant difference (LSD) post hoc test. If ANOVA did not reveal a significant effect of treatment, a priori pairwise comparisons test against the control group was conducted. Non-normally distributed data were analysed by the Kruskal-Wallis and non-parametric Mann-Whitney U test. P<0.05 was the criterion for statistical significance.

Example 8b—Assessment of Social Behaviours—Social Transmission of Food Preference

Rationale

Social transmission food preference is a relevant test of olfactory memory that is used in mice to assess social behaviour. In this test, observer mice interact with a demonstrator mouse that has recently eaten novel food. When observer mice are presented with a choice between the food eaten by the demonstrator and some other novel food, observer mice should prefer the food eaten by the demonstrator. Reduced food preference would indicate reduced sociability.

Methods

This test was performed as previously described (Desbonnet, Clarke et al. 2015). Briefly, 18 hours prior to testing, mice were deprived of food, whereas water was available ad libitum. Food choices consisted of either 1% ground cinnamon or 2% powdered cocoa made with grounded mouse chow. A demonstrator mouse was randomly selected from each cage and the tail was marked using a blue marker to enable identification during subsequent social interactions. Demonstrator food containers were weighed before and after the 1 hour sampling sessions. A minimum of 0.2 g of consumed food was required for inclusion in the test. Demonstrator mice were placed back into their respective home cages for a 30 minute interaction period with cage-mates. Subsequently, cage-mates were individually tested for preference of cued food or novel food. Containers were weighed immediately before and after each choice session. Observed mice were then placed back into their respective home cages and the choice session was repeated 24 hours later. The test mice should smell the cinnamon or cocoa off the demonstrator mouse as a social cue, and preferentially choose the same food when given a choice between the two.

Results

ANOVA of demonstrator cued food preference revealed no significant difference when observers were exposed to food choice immediately after demonstrator interaction (T0) (F(3,36)=1.123; P=0.354; FIG. 40A) or 24 hrs later (F(3,38)=0.138; P=0.936; FIG. 40B).

Conclusions

Treatment with MRX008 did not affect the sociability of BTBR mice in the social transmission food preference test.

Example 8c—Assessment of Social Behaviours—Forced Intruder Test

Rationale and Methods

See Example 2c above.

Results

ANOVA of interaction time did not reveal an effect of treatment [F(3,36)=1.905; P=0.1462; FIG. 41].

Conclusions

Treatment with MRX008 did not influence social behaviour of BTBR mice in the social interaction test.

Example 8d—Assessment of Stereotyped Behaviours—the Marble Burying Test

Rationale and Methods

See Example 2d above.

Results

ANOVA of the number of marbles buried did not reveal a significant effect of treatment [F(3,39)=0.835; P=0.483; FIG. 42], although, chronic treatment with MRX008 does display a trend towards a reduction in number of marbles buried by BTBR mice.

Conclusions

Chronic treatment with MRX008 did not significantly affect repetitive, compulsive and anxious behaviour in BTBR mice, although it does display a trend towards reduced levels of this behaviour.

Example 8e—Assessment of Anxiety-Like Behaviour—the Elevated Plus Maze

Rationale and Methods

See Example 2f above.

Results

ANOVA of percentage time spent in closed arms revealed no effect of treatment [F(3,39)=0.556; P=0.647; FIG. 43A]. Mice treated with MRX008 appear to spent more time in the open arms compared to the vehicle group (FIG. 43B). In line with this, chronic treatment with MRX008 appears to increased the number of entries into open arms compared to the vehicle group (FIG. 43D). ANOVA of the number of entries into the closed arms revealed no effect of treatment [F(3,39)=0.556; P=0.647; FIG. 43C].

Conclusions

Chronic treatment with MRX008 shows a non-significant trends towards anti-anxiety behaviour in BTBR mice in the elevated plus maze.

Example 8f—Assessment of Anxiety-Like Behaviour—the Open Field Arena

Rationale and Methods

See Example 2g above.

Results

ANOVA of distance moved did not reveal a significant effect of treatment upon locomotor activity in the open field arena [F(3,37)=1.325; P=0.282, FIG. 44A], although MRX008 appeared to reduce the distance moved, suggesting a reduction in stress-induced locomotor activity. ANOVA of time spent in the outer zone did not reveal an effect of treatment [F(3,37)=1.598; P=0.208; FIG. 44B]. A priori pairwise comparison revealed that treatment with Mrx008 decreased the time spent in the inner zone [t=2.388 df=17; P=0.0288; FIG. 44C].

Conclusions

Chronic treatment with MRX008 shows a trend towards a reduction in stress-induced locomotor activity, but did show a reduction of time in the inner zone implicating anxiety-like behaviour.

Example 8g—Assessment of Depression-Like Behaviour—the Forced Swimming Test

Rationale and Methods

See Example 2h above.

Results

ANOVA of immobility time did not reveal an effect of treatment on immobility time of BTBR mice in the FST [F(3,38)=1.879; P=0.151; FIG. 45], although chronic treatment with MRX008 does cause a trend towards a reduction in the time spent immobile suggesting anti-depressant behaviour.

Conclusions

Treatment with MRX008 non-significantly reduces immobility time of BTBR mice in the forced swimming test implicating an anti-depressant effect of treatment.

Example 8h—Assessment of Depression-Like Behaviour—the Female Urine Sniffing Test

Rationale and Methods

See Example 2i above.

Results

For the vehicle group, Mann-Whitney U test revealed a significant increase in the time spent sniffing urine relative to the time spent sniffing water [t=2.976 df=18; P=0.0081]. For exposure to water, Kruskal Wallis non-parametric analysis of time spent sniffing did not reveal an effect of treatment in the water group [Chi squared: 6.352; df=3; P=0.096]. For exposure to urine, Kruskal Wallis non-parametric analysis of time spent sniffing did not reveal an effect of treatment [Chi squared: 3.639; df=3; P=0.303, FIG. 46].

Conclusions

Treatment with MRX008 had no effect upon the time spent sniffing urine in BTBR mice.

Example 8i—In Vivo Gastrointestinal Motility Assay

Rationale and Methods

See Example 2m above.

Results

ANOVA of motility time revealed no effect of treatment [F(3,39)=2.072; P=0.121; FIG. 47].

Conclusions

Treatment with MRX008 did not influence intestinal motility.

Example 8j—Organ Weight and Colon Length

ANOVA of organ weight as a percentage of body weight did not reveal an effect of treatment for the adrenals [F(3,37)=0.208; P=0.890; FIG. 48A], spleen F(3,35)=0.629; P=0.601; FIG. 48B] or caecum [F(3,37)=0.883; P=0.460; FIG. 48C]. ANOVA of colon length did not reveal an effect of treatment [F(3,37)=5.635; P=0.003; FIG. 48D].

Overall Conclusions Regarding MRX008 in the Treatment of Autistic Spectrum Disorders

The experiments disclosed herein display evidence that administration of another Blautia species (namely Blautia wexlerae MRX008) may be applicable for the treatment of neurodevelopmental and neuropsychiatric disorders in mice models. In particular, treatment with MRX008 displayed trends towards potential anti-anxiolytic effects as well as anti-depressive effects in the elevated plus maze and forced swim tests, respectively, in the BTBR mouse model, although the open field arena assay suggested MRX008 did not affect anxiety-like behaviour. In addition, the MRX008 may reduce stereotyped, repetitive and anxious behaviour as shown by the marble burying test in both the MIA and BTBR mouse models. Treatment with the MRX008 biotherapeutic did not alter the several physiological parameters measured in these studies.

The EMA Guidelines on the clinical development of medicinal products for the treatment of autism spectrum disorder state that, due to the heterogeneity of the diseases, it may not be possible to achieve a significant effect on all core symptoms with a single compound, and so short term efficacy has to be demonstrated on at least one core symptom. The MRX008 live biotherapeutic has shown effective treatment of at least one core symptom of autistic spectrum disorder, so it and related Blautia and B. wexlerae strains are expected to be effective against human disease. Similarly, other central nervous system disorders or conditions display complex pathology with multiple different symptoms, and have few approved treatments. Therefore, it is understood that an effective treatment does not need to treat all symptoms of a central nervous system disorder or condition. A treatment would be considered therapeutically useful if it treated one of the symptoms associated with a central nervous system disorder or condition.

Example 9—Assessing the Effects of Subchronic Treatment with MRX008 Upon Central and Peripheral Oxytocin Levels in C57BL/6 Mice

The bacterial strains were prepared and administered as outlined in the Examples above. The C57BL/6 mice were treated with live biotherapeutic for six days in 7 experimental treatment groups each with 10-12 mice. Subsequently, the hypothalamus was dissected from the mice and the levels of oxytocin in the hypothalamus were detected by radioimmunoassay (RIA), In addition, levels of oxytocin in the plasma were detected by RIA. Furthermore, the levels of oxytocin receptors, interleukins and other inflammatory markers, and vasopressin hormones were detected by RIA and other analytical methods.

Example 10—Assessing the Effects of Chronic Treatment with MRX006 on Gene Expression Levels of Oxytocin, Vasopressin and their Respective Receptors in the Hypothalamus and the Amygdala of BTBR Mice

Chronic treatment with MRX006 increases the level of gene expression of oxytocin and vasopressin in the hypothalamus of BTBR mice (see FIGS. 49C and D). The effect on levels of oxytocin and vasopressin receptors in this tissue are shown in FIGS. 49A and B.

The effects of chronic treatment with MRX006 on the level of gene expression of oxytocin, vasopressin or their respective receptors in the amygdala of BTBR mice is shown in FIG. 50.

Therefore, chronic treatment with MRX006 increases the expression of vasopressin and oxytocin in the hypothalamus of BTBR mice. This striking result provides a correlation between chemical changes in the brain and behavioural changes upon administration of a live biotherapeutic. This is the first time any study has reported that a live biotherapeutic is capable of altering the central oxytocin/vasopressin system, with a concurrent change in social, anxiety-like and stereotyped behaviour with an improvement in gastrointestinal function.

Example 11—Administration of Blautia hydrogenotrophica in the C57BL/6 and BTBR Mouse Models

In behavioural experiments using BTBR mice as a model for autism spectrum disorder and other neurological disorders, C57BL/6 mice administered both PBS and LYO were used as controls to confirm that the BTBR mice model effectively demonstrated increased anxiety, reduced social aversion, and increased stereotypes behaviours. This allowed an assessment of the effect of bacterial treatment on these ASD related behavioural defects.

Example 11a—Assessment of Anxiety-Like Behaviour—the Open Field Arena

Rationale and Methods

See Example 2g above. The horizontal activity is the distance travelled by the mouse in the open field arena. The vertical activity is the number of occasions on which the mouse reared onto the hind legs. A higher frequency of these behaviours indicates increased locomotion and exploration and/or a lower level of anxiety. An increase frequency of these behaviours in the central area of the arena indicates high exploratory behaviour and low anxiety levels.

PBS is the negative control for the butyrate administration as the butyrate was administered in PBS. LYO is the negative control for the administration of the Blautia hydrogenotrophica. After the first analyses (FIGS. 51 A, C and E), the values for the negative controls of PBS and LYO in both the C57BL/6 and BTBR models are combined and averaged to provide a simplified comparison in the second analysis (FIGS. 51 B, D and F).

Results

Horizontal Activity

As would be expected from an anxiety and/or autism-related model, BTBR mice display significantly reduced horizontal activity compared to C57BL/6 mice. The LYO negative control showed no effect on the horizontal activity in C57BL/6 mice compared to the PBS control. Compared to the PBS control within the first 30 minutes, BTBR mice treated with the LYO control or butyrate alone showed no significant difference in distance travelled, although in the second 30 minutes, the LYO control reduced the distance travelled by BTBR mice. However, mice treated with the bacterial strain showed a significant increase in distance travelled compared to the BTBR control mice (FIG. 51A).

To provide a further comparison between the controls and the experimental values, as outlined above, the values for the PBS and LYO controls were combined in the second analysis. Similarly to the first analysis, the administration of butyrate did not affect the horizontal activity. However, the administration of the bacterial strain significantly increased the horizontal activity compared to the BTBR model control (FIG. 51B).

Vertical Activity

BTBR mice display significantly reduced vertical activity (rearing) compared to C57BL/6 mice. The LYO negative control showed no effect on the vertical activity in C57BL/6 mice compared to the PBS control. Compared to the BTBR PBS control mice, BTBR mice treated with butyrate alone showed no difference in rearing, while the LYO control reduced the vertical activity of BTBR mice. However, mice treated with the bacterial strain showed a significant increase in vertical activity compared to the BTBR LYO control mice (FIG. 51C).

To provide a further comparison between the controls and the experimental values, as outlined above, the values for the PBS and LYO controls were combined in the second analysis. Similarly to the first analysis, the administration of butyrate did not affect the vertical activity of BTBR mice. However, the administration of the bacterial strain significantly increased the vertical activity compared to the BTBR control (FIG. 51D).

% distance travelled in the centre of the open field in the first 5 minutes

As would be expected, in the first five minutes of the analysis BTBR mice showed an increased percentage of their distance travelled in the centre of the open field arena compared to C57BL/6 mice.

This is reflective of the reduced overall distance travelled by the BTBR mice, which display increased anxious behaviour, and the fact that within the first 5 minutes of the assay, the more anxious BTBR mice are more likely to familiarise themselves with their initial environment rather than enter an exploratory phase (FIG. 51E).

To provide a further comparison between the controls and the experimental values, as outlined above, the values for the PBS and LYO controls were combined in the second analysis (FIG. 51F).

% time spent in the centre of the open field arena

When considering the entire time of the analysis, BTBR mice show a reduced percentage time spent in the centre of the arena compared to C57BL/6 mice. This is reflective of the increased anxiety and reduced horizontal activity of the BTBR mice. Neither the LYO control nor butyrate alone affected the time spent in the centre of the arena. However, the administration of the bacterial strain significantly increased the amount of time spent in the centre of the arena compared to the LYO control in BTBR mice (FIG. 51G).

To provide a further comparison between the controls and the experimental values, as outlined above, the values for the PBS and LYO controls were combined in the second analysis. Similarly to the first the administration of butyrate did not affect the time spent in the centre of the field compared to the BTBR control. The administration of the bacterial strain increased the amount of time spent in the centre of the open field.

Conclusions

The chronic treatment with a composition of Blautia hydrogenotrophica increases exploratory activity and reduces anxiety-like behaviour in the BTBR mouse model in the open field arena test. Critically, administration of the bacterial strain to the BTBR mouse model increased horizontal and vertical activity and increased the total amount of time spent in the centre of the arena compared to the BTBR control. Accordingly, this bacterial strain has anxiolytic effects and improves exploratory behaviour in a mouse model representative of central nervous system disorders (e.g. autism spectrum disorders).

Example 11b—Assessment of Stereotyped Behaviours—the Marble Burying Test

Rationale and Methods

See Example 2d above. PBS is the negative control for the administration of butyrate. LYO is the negative control for the administration of the Blautia hydrogenotrophica strain. After the first analyses (FIG. 52 A), the values for the negative controls of PBS and LYO in both the C57BL/6 and BTBR models are combined and averaged to provide a simplified comparison in the second analysis (FIG. 52B).

Results

As would be expected, the BTBR model mice displayed an increase in repetitive behaviour showing significantly more marbles buried compared to the C57BL/6 model control (FIG. 52B). The administration of butyrate and the bacterial strain reduced the number of marbles buried (FIGS. 52A and B).

Conclusion

Administration of butyrate and/or the bacterial strain reduces the number of marbles buried, indicating a reduction in anxious or stereotyped behaviours.

Example 11c—Assessment of Stereotyped Behaviours—the Digging Test

Rationale and Methods

Similar to the rationale of the marble burying test, increased digging behaviour corresponds to an increase in repetitive and stereotyped behaviour.

Results

As would be expected, there was a significant increase in time spent digging in the BTBR model compared to the C57BL/6 control strain (FIG. 53A). However, the number of digging bouts was not significantly different between the C57BL/6 and BTBR strains (FIG. 53B). Therefore, it is not possible to assess the role of the bacterial strain or butyrate in preventing repetitive behaviour in this analysis.

Example 11d—Assessment of Stereotyped Behaviours—the Self-Grooming Test

Rationale and Methods

See Example 2e above. PBS is the negative control for the administration of butyrate. LYO is the negative control for the administration of the Blautia hydrogenotrophica. After the first analyses (FIGS. 54A, C and E), the values for the negative controls of PBS and LYO in both the C57BL/6 and BTBR models were combined and averaged to provide a simplified comparison in the second analysis (FIGS. 54B, D and F).

Results

In line with the BTBR model for stereotyped behaviours, the BTBR mice showed increased time spent grooming as well as increased numbers of grooming bouts compared to the C57BL/6 mouse model, in both the PBS and LYO controls. Administration of butyrate alone showed a reduction in the time spent grooming, the number of grooming bouts, and the time spent grooming per bout, compared to the PBS control. Administration of the bacterial strain reduced the time spent grooming per grooming bout (FIGS. 54A, C and E).

To provide a further comparison between the controls and the experimental values, as outlined above, the values for the PBS and LYO controls were combined in the second analysis. This second analysis provided similar results to those of the first analysis (FIGS. 54B, D and F).

Conclusion

Administration of butyrate or Blautia hydrogenotrophica reduces the amount of time spent grooming per bout of grooming.

Example 11e—Overall Conclusion of the Blautia hydrogenotrophica Experiments

In the open field arena test, Blautia hydrogenotrophica significantly improved the exploratory behaviour of the BTBR mice. Furthermore, this bacterial strain reduced anxiety-like behaviour of these mice. Accordingly, it is clear that administration of this bacteria modulates the behaviour of the BTBR mice which display autism-like characteristics. Therefore, one would expect these bacteria to be useful in the treatment and/or prevention of central nervous system disorders or conditions, including neurodevelopmental and/or a neuropsychiatric disorders or conditions.

Administration of this bacterial strain also appears to reduce the amount of time performing stereotyped behaviour per grooming bout in the self-grooming test.

Example 11f—Overall Conclusion of the Butyrate Experiments

The data from the stereotyped behaviour assays point towards a therapeutic role for butyrate in central nervous system disorders.

The administration of butyrate reduced the number of marbles buried compared to the BTBR control and returned the average number to a level similar to that observed in the C57BL/6 wild-type control mice. In addition, the administration of butyrate alone reduced the overall time spend grooming and number of grooming bouts compared to the BTBR control.

These results provide telling indications regarding a role for butyrate in reducing repetitive and stereotyped behaviours in animal models.

Example 12—Effects of Bacterial Lyophilisate on SCFA Production Healthy Rats

The effects of chronic administration of a lyophilisate of Blautia hydrogenotrophica strain DSM 14294 on SCFA production in healthy HIM rats were studied and the results are reported in FIG. 55. Further details regarding the experiments are provided above in the descriptions of the figure. FIG. 55 shows that administration of BH induces a significant increase in acetate as well as in butyrate production.

Example 13—Efficacy of B. hydrogenotrophica Studied in Human Microbiota Associated Rat (HMA Rat) Model

Summary

Groups of 16 germ-free rats (comprising 8 rats in the control group and 8 rats in the treatment group) were inoculated with the faecal microbiota from a human IBS subject (IBS-HMA rats). Three successive experiments were carried out using faecal samples from 3 different IBS patients. Two other groups of rats (n=10) were inoculated with faecal samples of healthy subject (n=2 subjects; 2 groups of healthy-HMA rats) as visceral sensitivity control. Thus, there were 24 IBS-microbiota associated rats (control), 24 IBS microbiota associated rats treated with Blautix and 20 healthy-microbiota associated rats. Half of the IBS-HMA rats were then administered for 28 days with composition comprising the bacterial strain of B. hydrogenotrophica according to the invention while the other half animals received a control solution.

Strain

Blautia hydrogenotrophica (BH) strain DSM 14294.

Composition and Administration

BH lyophilisate was suspended in sterile mineral solution to a concentration of 1010 bacteria per ml. Two ml of this suspension was administered daily per IBS-HMA rat, by oral gavage, for a 28 days period.

The control solution was the sterile mineral solution that was administered daily (2 ml per rat) by oral gavage to the control group of IBS-HMA rats.

Rats

Germ-Free male Fisher rats (aged 10 weeks) were inoculated with human faecal microbiota from an IBS subject (IBS-HMA rats). Sixteen rats were inoculated with the same human faecal inoculum. Three successive experiments were performed with faecal samples from three different IBS subjects. Two other groups of ten rats were inoculated with faecal sample from 2 healthy subjects (normo-sensitivity control groups).

Study Design

Day −14—Inoculation of Germ-free rats with human faecal microbiota.

Days 0 to 28—Daily dose of BH lyophilisate (assay group), or control solution (control group) by oral gavage

Between days 14 and 22—operation to implant electrode into the abdomen (for distension assay)

Days 22-28—Adaptation of the rats to avoid stress associated with distension test.

Day 28—distension assay and euthanasia of animals to collect the caecal samples for sulphides and short chain fatty acid (SCFA) analysis.

Days 0, 14 and 28—Collection of faecal samples for microbial analysis: qPCR for evaluating BH population and other commensal groups of microorganisms and enumeration of functional groups of microorganisms using selective media and strictly anaerobic method.

Results

FIG. 56 presents the results of qPCR analysis of the B. hydrogenotrophica population in faecal samples from IBS-HMA rats receiving control solution or BH lyophilisate. A significant increase in the BH population was observed at the end of the administration period (D 28) in rats receiving the BH lyophilisate, which confirms successful delivery of BH in the colon.

FIG. 57 reports on the impact of administration of BH on the main fermentative metabolites, short chain fatty acids, in caecal samples of IBS-HMA rats. Administration of BH-resulted in a significant increase in acetate concentration as well as in a significant increase in butyrate concentration (FIG. 57B).

Example 14—Assessment of Social Interaction Behaviour in the Three Chamber Test

Rationale and Methods

See Example 1b above. In this experiment, the data recorded is the exploration time, which is defined by the sniffing time of the cylinders (containing an object, a congener) during the first 5-min period and during the 10-min session.

Read-outs:

    • Test 1, sociability (congener vs. object):
      • Index of sociability: % sniffing time of the congener (if >50%: sociability, i.e. preference for the congener vs. the object)
      • Other read-outs, indices of exploratory behaviour: exploration time of the congener, the object, total
    • Test 2, social novelty preference (new congener vs. familiar congener):
      • Index of social novelty preference (or aversion): % sniffing time of the new congener
      • Other read-outs, indices of exploratory behaviour: exploration time of the new and familiar congeners, total

Results

Test 1: Sociability (FIG. 58A):

In the C57BL/6 mice the sociability is not different in PBS and in LYO treated mice. As would be expected, BTBR mice showed reduced sociability in the PBS control. Unexpectedly BTBR mice displayed improved sociability when treated with LYO. However, the differences between BTBR-PBS vs. C57-PBS, BTBR-LYO vs. C57-LYO and BTBR-PBS vs. BTBR-LYO are not significant. Interestingly, administration of butyrate improved sociability in BTBR mice (significantly different between BTBR-PBS vs. BTBR-BUT. Administration of Blautia hydrogenotrophica increased sociability compared to the bacterial PBS control.

Test 2: Social Novelty (FIG. 58B):

There is a social novelty preference in C57BL/6 mice treated with PBS, but this preference is reduced in C57BL/6 mice administered LYO (these differences are not significant). In the 10 min session, BTBR mice showed reduced social novelty preference compared to C57BL/6 mice when treated with either PBS or LYO. The differences between differences BTBR-PBS vs. C57-PBS, BTBR-LYO vs. C57 LYO and BTBR-PBS vs. BTBR-LYO are not significant. The results shown in FIG. 58B are therefore difficult to interpret.

Overall Conclusions Regarding Blautia hydrogenotrophica in the Treatment of Autistic Spectrum Disorders

The experiments disclosed herein display evidence that administration of another Blautia species (namely Blautia hydrogenotrophica) may be applicable for the treatment of neurodevelopmental and neuropsychiatric disorders in mice models. In particular, treatment with Blautia hydrogenotrophica reduced anxiety-like, stereotyped and repetitive behaviour, and increased sociability in mice.

The EMA Guidelines on the clinical development of medicinal products for the treatment of autism spectrum disorder state that, due to the heterogeneity of the diseases, it may not be possible to achieve a significant effect on all core symptoms with a single compound, and so short term efficacy has to be demonstrated on at least one core symptom. The Blautia hydrogenotrophica live biotherapeutic has shown effective treatment of at least one core symptom of autistic spectrum disorder, so it and related Blautia and B. hydrogenotrophica strains are expected to be effective against human disease. Similarly, other central nervous system disorders or conditions display complex pathology with multiple different symptoms, and have few approved treatments. Therefore, it is understood that an effective treatment does not need to treat all symptoms of a central nervous system disorder or condition. A treatment would be considered therapeutically useful if it treated one of the symptoms associated with a central nervous system disorder or condition.

Example 15—Assessing the Effects of Chronic Treatment with MRX006 on Gene Expression Levels of Oxytocin and its Respective Receptors in the Hypothalamic Cell Lines

Chronic treatment with MRX006 significantly increases the level of mRNA expression of oxytocin and its receptor in hypothalamic cell lines (FIGS. 59A and B).

This striking result provides a correlation between chemical changes in the brain and behavioural changes upon administration of MRX006. This is the first time any study has reported that a live biotherapeutic is capable of altering the central oxytocin system, with a concurrent change in social, anxiety-like and stereotyped behaviour with an improvement in gastrointestinal function.

Example 16—the BALBc Mouse Model Example 16a—Materials and Methods for BALBc Mouse Model

Mice

BALBc (Envigo, UK) adult male mice were group housed under a 12 h light-dark cycle (lights on from 7:00-19:00 hr); standard rodent food and water were available ad libitum. All experiments were conducted in accordance with the European Directive 2010/63/EEC, the requirements of SI. No 543 of 2012, and approved by the Animal Experimentation Ethics Committee of University College Cork. Animals were 8 weeks old at the start of the experiment.

Strain

MRX006: Blautia stercoris bacterium deposited under accession number NCIMB 42381.

The bacteria were provided in glycerol stock and grown in the facility in anaerobic conditions.

MRX006 Administration

Animals were allowed to habituate to their holding room for one week after arrival into the animal unit. Dosing with MRX006 or vehicle commenced when the mice were 8 weeks old. MRX006 (1×107 to 1×109 CFU) was dissolved in PBS prior to administration. The mice received oral gavage (200 μL dose) of MRX006 at a dose of 1×109 CFU for 6 consecutive days between 15:00 and 17:00. On day 7, the animals were decapitated and tissues were harvested for experimentation.

Tissue Collection

Animals were sacrificed in a random fashion regarding treatment and testing condition; sampling occurred between 9.00 a.m. and 2:30 p.m. Trunk blood was collected in potassium EDTA (Ethylene Diamine Tetra Acetic Acid) tubes and spun for 15 min at 4000 g. Plasma was isolated and stored at −80° C. for further analysis. The brain was quickly excised, dissected and each brain region was snap-frozen on dry ice and stored at −80° C. for further analysis. Spleen was removed, collected in 5 mL RPMI media (with L-glutamine and sodium bicarbonate, R8758 Sigma+10% FBS (F7524, Sigma)+1% Pen/Strep (P4333, Sigma)) and processed immediately after culls for ex-vivo immune stimulation. Intestinal tissue (2 cm segments of ileum and colon closest to the caecum were excised, and the furthest 1 cm of tissue from the caecum were used) were mounted into the Ussing chambers for intestinal permeability assay. A further 1 cm of ileum and colon tissue was taken for tight junction gene expression analysis. The caecum was removed, weighed and stored at −80° C. for SCFAs analysis.

Statistical Analysis

Normally distributed data are presented as mean±SEM; Non-parametric datasets are presented as median with inter-quartile range. Unpaired two-tailed t-test were applied to analyse parametric data and Mann-Whitney test was used for non-parametric. Spearman's rank correlation coefficient was employed for the correlation analysis in the pooled datasets. A p value <0.05 was deemed significant in all cases.

Example 16b—Assessing the Effects of Chronic Treatment with MRX006 on Ex Vivo Gastrointestinal Permeability and Tight Junction Expression

Methods

Mice were euthanized by cervical dislocation, and the distal ileum and colon were removed, placed in chilled Krebs solution, opened along the mesenteric line and carefully rinsed. Preparations were then placed in Ussing chambers (Harvard Apparatus, Kent, UK, exposed area of 0.12 cm2) as described previously (Hyland and Cox, 2005 [58]) with oxygenated (95% O2, 5% CO2) Krebs buffer maintained at 37° C. 4 kDa FITC-dextran was added to the mucosal chamber at a final concentration of 2.5 mg/mL; 200 μL samples were collected from the serosal chamber every 30 min for the following 3 h.

Results

Using the passage of FITC from the luminal to the serosal side of the Ussing chamber as an index of gut permeability (as described in Example 21), it was determined that MRX006 had no effect on ileum or colon tissue permeability. FIGS. 60A and 61A demonstrate that chronic treatment with MRX006 does not influence the permeability of the colon or ileum.

MRX006 had no effect on mRNA expression of the tight junction protein (involved in maintaining the integrity of the gut barrier) occludin, the enzyme IDO-1 (Indoleamine-pyrrole 2,3-dioxygenase-1 the first and rate-limiting enzyme in the tryptophan/kynurenine pathway), nor TPH1 (Tryptophan hydroxylase 1, an isoform of the enzyme tryptophan hydroxylase, responsible for the synthesis of serotonin) in ileum or colon tissue (FIGS. 60 and 61 B, C and E). MRX006 did however increase TJP-1 (Tight Junction Protein 1, a tight junction protein) mRNA expression in the ileum, but not the colon (FIGS. 60D and 61D).

Discussion

MRX006 had no effect on ileum or colon permeability, but did increase TJP1 expression. TJP1 is one of a number of tight junction proteins associated with maintaining gut integrity, and while we did see this increase in mRNA expression, this may not necessarily reflect the protein expression of this tight junction nor its incorporation into the endothelium. The finding that the 6 day treatment with MRX006 does not alter permeability suggests that it does not negatively impact on gut permeability and integrity. MRX006 also did not alter IDO-1 nor TPH1 suggesting that it does not alter serotonin production nor the tryptophan/kynurenine pathway in the gut.

These data demonstrate that chronic treatment with MRX006 does not alter the gut permeability and does not affect the integrity of the gut barrier. This shows that the ability of MRX006 attenuate stereotyped and anxiety-related behaviours does not lead to a deficiency in the gut barrier integrity.

Example 16c—Assessing the Effects of Chronic Treatment with MRX006 on Caecal Short Chain Fatty Acid Production

Methods

Caecum content was mixed and vortexed with MilliQ water and incubated at room temperature for 10 min. Supernatants were obtained by centrifugation (10000 g, 5 min, 4° C.) to pellet bacteria and other solids and filtration by 0.2 m. It was transferred to a clear GC vial and 2-Ethylbutyric acid (Sigma) was used as the internal standard. The concentration of SCFA was analyzed using a Varian 3500 GC flame-ionization system, fitted with a with a ZB-FFAP column (30 m×0.32 mm×0.25 mm; Phenomenex). A standard curve was built with different concentrations of a standard mix containing acetate, propionate, iso-butyrate, n-butyrate, isovalerate and valerate (Sigma). Peaks were integrated by using the Varian Star Chromatography Workstation version 6.0 software. All SCFA data are expressed as pmol/g.

Results

Short chain fatty acids (SCFAs) are produced when non-digestible fibres from the diet are fermented by bacteria in the gut. 6 days of MRX006 administration had no effect on acetate (t12=0.959, p=0.357), propionate (t12=1.033, p=0.322), isobutyrate (t12=1.859, p=0.090), butyrate (t12=0.857, p=0.408), isovalearate (t12=1.757, p=0.107) or valearate (t12=0.434, p=0.672), when compared to vehicle PBS administration (FIG. 62).

Discussion

The administration of MRX006 had no effect on caecal SCFA production. This suggests that the 6 day regime of MRX006 did not alter the fermentation, or the bacteria responsible for the fermentation of non-digestible fibres from the diet.

Example 16d—Assessing the Effects of Chronic Treatment with MRX006 on Cytokine Expression from Splenocytes

Rationale Methods

The ex-vivo splenocyte assay involves challenging the splenocytes (cells isolated from the spleen—a main organ involved in immune defence), with a bacterio- or viral-mimetic challenge.

Spleens were collected immediately in 5 mL RPMI media following sacrifice and cultured immediately. Spleen cells were first homogenised in the RPMI media. The homogenate step was followed by RBC lysis step where the cells were incubated for 5 mins in 1 ml of RBC lysis buffer (11814389001 ROCHE, Sigma). 10 ml of the media was added to stop the lysis and followed by 200 g centrifugation for 5 mins. This was followed by final step where the cells were passed through 40 um strainer. The homogenate was then filtered over a 40 um strainer, centrifuged at 200 g for 5 min and resuspended in media. Cells were counted and seeded (4,000,000/mL media). After 2.5 h of adaptation, cells were stimulated with lipopolysaccharide (LPS-2 μg/ml) or concanavalin A (ConA-2.5 μg/ml) for 24 h. Following stimulation, the supernatants were harvested to assess the cytokine release using Proinflammatory Panel 1 (mouse) V-PLEX Kit (Meso Scale Discovery, Maryland, USA) for TNFα, IL-10, IL-1β, Interferon γ, CXCL2 and IL6. The analyses were performed using MESO QuickPlex SQ 120, SECTOR Imager 2400, SECTOR Imager 6000, SECTOR S 600.

Results

MRX006 had no effect on splenocyte release of proinflammatory (IFNγ, TNFα, IL-1β) nor anti-inflammatory (IL-10, IL-6) or CXCL1 (marker of immune response activation) in response to LPS (mimicking a bacterial infection) or concavalin A (mimicking a viral infection) stimulation (FIG. 63).

Discussion

MRX006 also had no effect on cytokine expression from splenocytes following a challenge with LPS or Concavalin A. This demonstrates that the 6-day MRX006 administration did not negatively influence the innate peripheral immune response. This shows that MRX006 treatment does not activate the systemic immune activation.

Example 16e—Assessing the Effects of Chronic Treatment with MRX006 on Plasma Levels of Amino Acids

Rationale and Methods

At the end of the experiment trunk blood was collected for amino acid analysis in the plasma. This would give an index of the biosynthesis and catabolism of essential amino acids by changes in microbiota.

Animals were sacrificed in a random fashion regarding treatment and testing condition; sampling occurred between 9.00 a.m. and 2:30 p.m. Trunk blood was collected in potassium EDTA (Ethylene Diamine Tetra Acetic Acid) tubes and spun for 15 min at 4000 g. Plasma was isolated and stored at −80° C. for further analysis. Plasma was diluted with 0.2 mol/L sodium citrate buffer, pH 2.2 to yield 250 nmol of each amino acid residue. Samples were diluted with the internal standard norleucine, to give a final concentration of 125 nm/mL. Amino acids were quantified using a Jeol JLC-500/V amino acid analyser (Jeol Ltd, Garden City, Herts, UK) fitted with a Jeol Na+ high performance cation exchange column.

Results

MRX006 decreased proline and phenylalanine levels in the plasma.

Discussion

Plasma levels of amino acids were largely unaltered following 6 day administration of MRX006. There are nine essential amino acids that cannot be synthesised de novo and must be supplied directly in the diet or by breakdown of the diet. These include valine, phenylalanine, threonine, tryptophan, methionine, leucine, isoleucine, lysine, and histidine. Six other amino acids are considered conditionally essential in the human diet, meaning their synthesis can be limited under special pathophysiological conditions, such as prematurity in the infant or individuals in severe catabolic distress. These six amino acids include arginine, cysteine, glycine, glutamine, proline, and tyrosine. Five amino acids are dispensable in humans, meaning they can be synthesized in sufficient quantities in the body. These five are alanine, aspartic acid, asparagine, glutamic acid and serine.

In this study the essential amino acid phenylalanine, and proline another important amino acid were decreased following MRX006 administration, suggesting that this probiotic may play a role in metabolism of key amino acids from the diet.

Example 16f—Assessing the Effects of Chronic Treatment with MRX006 on Neurotransmitter Levels in the Brainstem

Methods

Neurotransmitter concentration was analysed by HPLC on samples from the brainstem. Briefly, brainstem tissue was sonicated in 500 μl of chilled mobile phase spiked with 4 ng/40 μl of N-Methyl 5-HT (Sigma Chemical Co., UK) as internal standard. The mobile phase contained 0.1 M citric acid, 5.6 mM octane-1-sulphonic acid (Sigma), 0.1 M sodium dihydrogen phosphate, 0.01 mM EDTA (Alkem/Reagecon, Cork) and 9% (v/v) methanol (Alkem/Reagecon), and was adjusted to pH 2.8 using 4 N sodium hydroxide (Alkem/Reagecon). Homogenates were then centrifuged for 15 min at 22,000×g at 4° C. and 40 μl of the supernatant injected onto the HPLC system which consisted of a SCL 10-Avp system controller, LECD 6A electrochemical detector (Shimadzu), a LC-10AS pump, a CTO-10A oven, a SIL-10A autoinjector (with sample cooler maintained at 40 C) and an online Gastorr Degasser (ISS, UK). A reverse-phase column (Kinetex 2.6 u C18 100×4.6 mm, Phenomenex) maintained at 30° C. was employed in the separation (Flow rate 0.9 ml/min). The glassy carbon working electrode combined with an Ag/AgCl reference electrode (Shimdazu) operated a +0.8 V and the chromatograms generated were analyzed using Class-VP 5 software (Shimadzu). The neurotransmitters were identified by their characteristic retention times as determined by standard injections, which run at regular intervals during the sample analysis. The ratios of peak heights of analyte versus internal standard were measured and compared with standard injection. Results were expressed as ng of neurotransmitter per g fresh weight of tissue.

Results

6 days administration of MRX006 had no effect on levels of noradrenaline, dopamine, serotonin, 5-HIAA (5-hydroxy-indole-acetic acid; a metabolite of 5-HT (5-hydroxy-tryptamine (serotonin)), or serotonin turnover (the ratio of 5-HIAA:5-HT) as determined by unpaired 2-tailed t-test (FIG. 65). Noradrenaline (t12=0.307, p=0.764), dopamine (t12=0.957, p=0.357), serotonin (t12=0.745, p=0.074), 5-HIAA (t12=0.379, p=0.711) levels or serotonin turnover in brainstem (t12=0.683, p=0.507).

Discussion

Neurotransmitter levels in the brainstem were unaltered following 6-day MRX006 administration. These data suggest that MRX006 does not negatively impact on behaviours that are governed by monoamine levels at the level of the brainstem.

Example 16g—Assessing the Effects of Chronic Treatment with MRX006 on Central and Gastrointestinal Gene Expression

Rationale

Expression of genes for neurotransmitter receptors [serotonin receptor 1a(5-HT1a), dopamine D1 receptor, GABAB receptor subunit B1, GABAA receptor, NMDA2A and NMDA2B receptor], inflammatory markers [IL-1, IL6, CD11b, TNFα and TLR4], and endocrine markers [corticosterone releasing factor (CRF), corticosterone releasing factor receptors 1 and 2 (CRFR1, CRFR2), brain-derived neurotrophin factor (BDNF), vasopressin receptor, oxytocin receptor, glucocorticoid receptor and mineralocorticoid receptor] were analysed in brain tissue from the amygdala, prefrontal cortex and hippocampus.

Methods

Total RNA was extracted using the mirVana™ miRNA Isolation kit (Ambion/Llife technologies, Paisley, UK) and DNase treated (Turbo DNA-free, Ambion/life technologies) according to the manufacturers recommendations. RNA was quantified using NanoDrop™ spectrophotometer (Thermo Fisher Scientific Inc., Wilmington, Del., USA) according to the manufacturer's instructions. RNA quality was assessed using the Agilent Bioanalyzer (Agilent, Stockport, UK) according to the manufacturer's procedure and an RNA integrity number (RIN) was calculated. RNA with RIN value >7 was used for subsequent experiments. RNA was reverse transcribed to cDNA using the Applied Biosystems High Capacity cDNA kit (Applied Biosystems, Warrington, UK) according to manufacturer's instructions. Briefly, Multiscribe Reverse Transcriptase (50 U/μL) (1)(2)(1)(10) was added as part of RT master mix, incubated for 25° C. for 10 min, 37° C. for 2 h, 85° C. for 5 min and stored at 4° C. Quantitative PCR was carried out using probes (6 carboxy fluorescein—FAM) designed by Applied Biosystems to mouse specific targeted genes, while using β-actin as an endogenous control. Amplification reactions contained 1 μl cDNA, 5 μl of the 2×PCR Master mix (Roche), 900 nM of each primer and were brought to a total of 10 μl by the addition of RNase-free water. All reactions were performed in triplicate using 96-well plates on the LightCycler®480 System. Thermal cycling conditions were as recommended by the manufacturer (Roche) for 55 cycles. To check for amplicon contamination, each run contained no template controls in triplicate for each probe used. Cycle threshold (Ct) values were recorded. Data was normalized using R-actin and transformed using the 2-ΔΔCT method and presented as a fold change vs. control group.

Results

FIG. 66 shows that MRX006 had no effect on hippocampal gene expression of the neurotransmitter receptors serotonin 1a (5-HT1a) (t11=0.742, p=0.474), dopamine D1 receptor (t10=1.426, p=0.184), GABAB receptor B1 subunit (t12=1.871, p=0.086), GABAA receptor (t12=0.017, p=0.987), NMDA receptor subunit 2A (t11=1.275, p=0.229), NMDA receptor subunit 2B (t11=1.39, p=0.192).

FIG. 67 shows that MRX006 had no effect on amygdalar gene expression of the neurotransmitter receptors dopamine D1 receptor (t11=0.429, p=0.677), GABAB receptor B1 subunit (t11=0.998, p=0.344), GABAA receptor (t11=1.145, p=0.277), NMDA receptor subunit 2A (t12=0.852, p=0.411), NMDA receptor subunit 2B (t12=0.395, p=0.707).

FIG. 68 shows that MRX006 had no effect on prefrontal cortex gene expression of the neurotransmitter receptors dopamine D1 receptor (t11=0.583, p=0.571), GABAB receptor B1 subunit (t12=1.304, p=0.217), GABAA receptor (t10=2.043, p=0.068), NMDA receptor subunit 2A (t11=0.177, p=0.104), NMDA receptor subunit 2B (t11=1.235, p=0.243).

There was no effect of MRX006 on mRNA expression of neurotransmitter receptors in any of the brain regions investigated (FIGS. 66-68).

In the hippocampus and amygdala (FIGS. 69 and 70) there was no effect on mRNA expression of the various inflammatory markers. MRX006 had no effect on Hippocampal gene expression of the inflammatory markers IL-13 (t10=1.346, p=0.208), IL-6 (t12=1.041, p=0.308), CD11b (t12=1.195, p=0.255), TNFα (t11=0.816, p=0.342), TLR4 (t12=0.521, p=0.612). MRX006 had no effect on amygdalar gene expression of the inflammatory markers IL-1β (t11=1.53, p=0.988), IL-6 (t11=1.145, p=0.217), CD11b (t11=1.143, p=0.275), TLR4 (t11=0.971, p=0.532).

In the prefrontal cortex, MRX006 decreased mRNA expression for TLR4 without any changes in other inflammatory markers (FIG. 71). MRX006 significantly decreased mRNA expression of TLR4 (t12=2.639, p=0.0216) in the prefrontal cortex, but had no further effect on the prefrontal cortex gene expression for IL-6 (t11=1.145, p=0.217) or CD11b (t11=2.175, p=0.523).

MRX006 significantly decreased vasopressin receptor mRNA expression in hippocampus (t12=2.389, p=0.0342), but had no further effect on mRNA expression for endocrine markers CRF (t12=0.767, p=0.458), CRFR1 (t12=0.174, p=0.865), CRFR2 (t11=0.238, p=0.816), BDNF (t12=1.548, p=0.148), oxytocin receptor (t12=0.762, p=0.461), glucocorticoid receptor (t12=0.607, p=0.556), mineralocorticoid receptor (t12=0.67, p=0.516) (FIG. 72).

MRX006 had no effect on mRNA expression of amygdalar endocrine markers CRFR1 (t12=0.226, p=0.825), CRFR2 (t11=0.78, p=0.451), BDNF (t12=0.201, p=0.844), vasopressin receptor (t12=0.756, p=0.465), oxytocin receptor (t11=0.167, p=0.87), glucocorticoid receptor (t11=1.027, p=0.327), mineralocorticoid receptor (t11=1.448, p=0.175) (FIG. 73).

MRX006 had no effect on mRNA expression of the prefrontal cortex endocrine markers CRFR1 (t12=1.666, p=0.122), CRFR2 (t11=1.179, p=0.261), BDNF (t11=1.065, p=0.310), oxytocin receptor (t11=1.037, p=0.322), glucocorticoid receptor (t12=1.185, p=0.259), mineralocorticoid receptor (t11=1.910, p=0.083) (FIG. 74).

In the amygdala and the prefrontal cortex (FIGS. 73 and 74), there were no changes in mRNA expression of any endocrine markers, while in the hippocampus (FIG. 72) there was a decrease in mRNA expression of vasopressin receptor without any effect on the other endocrine markers analysed.

Discussion

The central gene expression for inflammatory, endocrine and neurotransmitter receptors were mostly unaltered following 6-day MRX006 administration.

Overall Conclusions Regarding MRX006 Administration on Physiological Parameters

Overall these data confirm that MRX006 administration does not negatively impact on systemic and central physiological events. These data suggest that MRX006 may have a high tolerability profile with minimal non-desirable side-effects.

Example 17—the Maternal Immune Activation (MIA) Mouse Model

The MIA mice used are the same as described in Example 3a.

Example 17a—Assessing the Effects of Chronic Treatment with MRX006 on In Vivo Gastrointestinal Permeability in MIA Model

The permeability of the ileum and colon was assessed in vivo using Ussing chambers as described in Example 2k. FIG. 75 demonstrates that chronic treatment with MRX006 does not influence the permeability of the colon or ileum in MIA model.

This confirms that chronic treatment with MRX006 does not alter the gut permeability, which shows that the beneficial social behaviour, reduce anxiety-like behaviour and stereotype behaviour effects of MRX006 do not lead to a deficit in the integrity of the gut.

Example 17b—Assessment of Social Behaviours—the Three Chamber Social Interaction Test

The 3-Chamber Social Interaction Test (3-CSIT) was conducted as described on example 1a, however this data was manually scored by a researcher that was blinded to treatment. The data in example 1a was automatically generated by computer tracking software which cannot distinguish between interaction with the mouse and just being in the same chamber as the mouse.

In the social novelty test (FIG. 76), there was no MIA-induced deficit in social discrimination and MRX006 had no further effect on social novelty.

FIG. 77 shows that in the sociability test, MRX006 was able to reverse MIA-induced deficits in social behaviour. This is similar to data seen in the BTBR model where MRX006 could reverse deficits in sociability.

Example 17c—Assessment of Social Behaviours—the Grooming Test

The grooming test was conducted as described in example 2e. Chronic treatment with MRX006 did not lead to a change in repetitive behaviours in MIA mice in the grooming test (FIG. 78).

Example 17d—Assessment of Social Behaviours—the Elevated Plus Maze

The elevated plus maze test was conducted as described in example 2f. Treatment with MRX006 has no effect on anxiety-like behaviour in MIA mice in the elevated plus maze (FIG. 79).

Example 17e—Assessment of Social Behaviours—the Forced Swim Test

The forced swim test was conducted as described in example 2h. Chronic treatment with MRX006 did reduce the immobility time of MIA mice in the forced swimming test (FIG. 80).

Example 17f—Stress-Induced Circulating Corticosterone Determination

The levels of corticosterone were measured as described in example 2n. Chronic treatment with MRX006 does not influence stress-induced corticosterone levels in MIA mice exposed to the forced swimming test (FIG. 81).

Conclusions

Treatment with MRX006 reversed MIA-induced deficits in social behaviour and reduced the immobility time in the forced swimming test. This demonstrates the ability of MRX006 to improve the sociability and antidepressant activity.

In addition to the results described above in example 3, MRX006 has been demonstrated to have a positive impact on the symptoms of autistic spectrum disorders.

Sequences (Blautia stercoris strain GAM6-1 16S ribosomal RNA gene, partial sequence- HM626177) SEQ ID NO: 1    1 tgcaagtcga gcgaagcgct tacgacagaa ccttcggggg aagatgtaag ggactgagcg   61 gcggacgggt gagtaacgcg tgggtaacct gcctcataca gggggataac agttggaaac  121 ggctgctaat accgcataag cgcacggtat cgcatgatac agtgtgaaaa actccggtgg  181 tatgagatgg acccgcgtct gattagctag ttggaggggt aacggcccac caaggcgacg  241 atcagtagcc ggcctgagag ggtgaacggc cacattggga ctgagacacg gcccagactc  301 ctacgggagg cagcagtggg gaatattgca caatggggga aaccctgatg cagcgacgcc  361 gcgtgaagga agaagtatct cggtatgtaa acttctatca gcagggaaga aaatgacggt  421 acctgactaa gaagccccgg ctaactacgt gccagcagcc gcggtaatac gtagggggca  481 agcgttatcc ggatttactg ggtgtaaagg gagcgtagac ggaagagcaa gtctgatgtg  541 aaaggctggg gcttaacccc aggactgcat tggaaactgt ttttcttgag tgccggagag  601 gtaagcggaa ttcctagtgt agcggtgaaa tgcgtagata ttaggaggaa caccagtggc  661 gaaggcggct tactggacgg taactgacgt tgaggctcga aagcgtgggg agcaaacagg  721 attagatacc ctggtagtcc acgccgtaaa cgatgaatac taggtgttgg ggagcaaagc  781 tcttcggtgc cgcagcaaac gcaataagta ttccacctgg ggagtacgtt cgcaagaatg  841 aaactcaaag gaattgacgg ggacccgcac aagcggtgga gcatgtggtt taattcgaag  901 caacgcgaag aaccttacca agtcttgaca tcgatctgac cggttcgtaa tggaaccttt  961 ccttcgggac agagaagaca ggtggtgcat ggttgtcgtc agctcgtgtc gtgagatgtt 1021 gggttaagtc ccgcaacgag cgcaacccct atcctcagta gccagcaggt gaagctgggc 1081 actctgtgga gactgccagg gataacctgg aggaaggcgg ggacgacgtc aaatcatcat 1141 gccccttatg atttgggcta cacacgtgct acaatggcgt aaacaaaggg aagcgagccc 1201 gcgaggggga gcaaatccca aaaataacgt cccagttcgg actgcagtct gcaactcgac 1261 tgcacgaagc tggaatcgct agtaatcgcg aatcagaatg tcgcggtgaa tacgttcccg 1321 ggtcttgtac acaccgcccg tcacaccatg ggagtcagta acgcccgaag tc (consensus 16S rRNA sequence for Blautia stercoris MRX006 (strain 830)) SEQ ID NO: 2 TTTKGTCTGGCTCAGGATGAACGCTGGCGGCGTGCTTAACACATGCAAGTCGAGCGAAGCGCTTACGACAGAACCTT CGGGGGAAGATGTAAGGGACTGAGCGGCGGACGGGTGAGTAACGCGTGGGTAACCTGCCTCATACAGGGGGATAACA GTTGGAAACGGCTGCTAATACCGCATAAGCGCACAGTATCGCATGATACAGTGTGAAAAACTCCGGTGGTATGAGAT GGACCCGCGTCTGATTAGCTAGTTGGAGGGGTAACGGCCCACCAAGGCGACGATCAGTAGCCGGCCTGAGAGGGTGA ACGGCCACATTGGGACTGAGACACGGCCCAGACTCCTACGGGAGGCAGCAGTGGGGAATATTGCACAATGGGGGAAA CCCTGATGCAGCGACGCCGCGTGAAGGAAGAAGTATCTCGGTATGTAAACTTCTATCAGCAGGGAAGAAAATGACGG TACCTGACTAAGAAGCCCCGGCTAACTACGTGCCAGCAGCCGCGGTAATACGTAGGGGGCAAGCGTTATCCGGATTT ACTGGGTGTAAAGGGAGCGTAGACGGAAGAGCAAGTCTGATGTGAAAGGCTGGGGCTTAACCCCAGGACTGCATTGG AAACTGTTTTTCTTGAGTGCCGGAGAGGTAAGCGGAATTCCTAGTGTAGCGGTGAAATGCGTAGATATTAGGAGGAA CACCAGTGGCGAAGGCGGCTTACTGGACGGTAACTGACGTTGAGGCTCGAAAGCGTGGGGAGCAAACAGGATTAGAT ACCCTGGTAGTCCACGCCGTAAACGATGAATACTAGGTGTTGGGGAGCAAAGCTCTTCGGTGCCGCAGCAAACGCAA TAAGTATTCCACCTGGGGAGTACGTTCGCAAGAATGAAACTCAAAGGAATTGACGGGGACCCGCACAAGCGGTGGAG CATGTGGTTTATTCGAAGCAACGCGAAGAACCTTACCAAGTCTTGACATCGATCTGACCGGTTCGTAATGGAACCTT TCCTTCGGGACAGAGAAGACAGGTGGTGCATGGTTGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAA CGAGCGCAACCCCTATCGTCAGTAGCCAGCAGGTAAAGCTGGGCACTCTGAGGAGACTGCCAGGGATAACCTGGAGG AAGGCGGGGACGACGTCAAATCATCATGCCCCTTATGATTTGGGCTACACACGTGCTACAATGGCGTAAACAAAGGG AAGCGAGCCCGCGAGGGGGAGCAAATCCCAAAAATAACGTCCCAGTTCGGACTGCAGTCTGCAACTCGACTGCACGA AGCTGGAATCGCTAGTAATCGCGAATCAGAATGTCGCGGTGAATACGTTCCCGGGTCTTGTACACACCGCCCGTCAC ACCATGGGAGTCAGTAACGCCCGAAGTCAGTGACCCAACCTTAGGGAGGGAGCTGCCGAAGGCGGGATTGATAACTG GGGTGAAGTCTAGGGGGT (Blautia wexlerae strain WAL 14507 16S ribosomal RNA gene, partial sequence- EF036467) SEQ ID NO: 3    1 caagtcgaac gggaattant ttattgaaac ttcggtcgat ttaatttaat tctagtggcg   61 gacgggtgag taacgcgtgg gtaacctgcc ttatacaggg ggataacagt cagaaatggc  121 tgctaatacc gcataagcgc acagagctgc atggctcagt gtgaaaaact ccggtggtat  181 aagatggacc cgcgttggat tagcttgttg gtggggtaac ggcccaccaa ggcgacgatc  241 catagccggc ctgagagggt gaacggccac attgggactg agacacggcc cagactccta  301 cgggaggcag cagtggggaa tattgcacaa tgggggaaac cctgatgcag cgacgccgcg  361 tgaaggaaga agtatctcgg tatgtaaact tctatcagca gggaagatag tgacggtacc  421 tgactaagaa gccccggcta actacgtgcc agcagccgcg gtaatacgta gggggcaagc  481 gttatccgga tttactgggt gtaaagggag cgtagacggt gtggcaagtc tgatgtgaaa  541 ggcatgggct caacctgtgg actgcattgg aaactgtcat acttgagtgc cggaggggta  601 agcggaattc ctagtgtagc ggtgaaatgc gtagatatta ggaggaacac cagtggcgaa  661 ggcggcttac tggacggtaa ctgacgttga ggctcgaaag cgtggggagc aaacaggatt  721 agataccctg gtagtccacg ccgtaaacga tgaataacta ggtgtcgggt ggcaaagcca  781 ttcggtgccg tcgcaaacgc agtaagtatt ccacctgggg agtacgttcg caagaatgaa  841 actcaaagga attgacgggg acccgcacaa gcggtggagc atgtggttta attcgaagca  901 acgcgaagaa ccttaccaag tcttgacatc cgcctgaccg atccttaacc ggatctttcc  961 ttcgggacag gcgagacagg tggtgcatgg ttgtcgtcag ctcgtgtcgt gagatgttgg 1021 gttaagtccc gcaacgagcg caacccctat cctcagtagc cagcatttaa ggtgggcact 1081 ctggggagac tgccagggat aacctggagg aaggcgggga tgacgtcaaa tcatcatgcc 1141 ccttatgatt tgggctacac acgtgctaca atggcgtaaa caaagggaag cgagattgtg 1201 agatggagca aatcccaaaa ataacgtccc agttcggact gtagtctgca acccgactac 1261 acgaagctgg aatcgctagt aatcgcggat cagaatgccg cggtgaatac gttcccgggt 1321 cttgtacaca ccgcccgtca caccatggga gtcagtaacg cccgaagtca gtgacctaac 1381 tgcaaagaag gagctgccga aggcgggacc gatgactggg gtgaagtcgt aacaaggt (consensus 16S rRNA sequence for Blautia wexlerae strain MRX008) SEQ ID NO: 4 TTCATTGAGACTTCGGTGGATTTAGATTCTATTTCTAGTGGCGGACGGGTGAGTAACGCGTGGGTAACCTGCCTTAT ACAGGGGGATAACAGTCAGAAATGGCTGCTAATACCGCATAAGCGCACAGAGCTGCATGGCTCAGTGTGAAAAACTC CGGTGGTATAAGATGGACCCGCGTTGGATTAGCTTGTTGGTGGGGTAACGGCCCACCAAGGCGACGATCCATAGCCG GCCTGAGAGGGTGAACGGCCACATTGGGACTGAGACACGGCCCAGACTCCTACGGGAGGCAGCAGTGGGGAATATTG CACAATGGGGGAAACCCTGATGCAGCGACGCCGCGTGAAGGAAGAAGTATCTCGGTATGTAAACTTCTATCAGCAGG GAAGATAGTGACGGTACCTGACTAAGAAGCCCCGGCTAACTACGTGCCAGCAGCCGCGGTAATACGTAGGGGGCAAG CGTTATCCGGATTTACTGGGTGTAAAGGGAGCGTAGACGGTGTGGCAAGTCTGATGTGAAAGGCATGGGCTCAACCT GTGGACTGCATTGGAAACTGTCATACTTGAGTGCCGGAGGGGTAAGCGGAATTCCTAGTGTAGCGGTGAAATGCGTA GATATTAGGAGGAACACCAGTGGCGAAGGCGGCTTACTGGACGGTAACTGACGTTGAGGCTCGAAAGCGTGGGGAGC AAACAGGATTAGATACCCTGGTAGTCCACGCCGTAAACGATGAATACTAGGTGTCNGGGGAGCATGGCTCTTCGGTG CCGTCGCAAACGCAGTAAGTATTCCACCTGGGGAGTACGTTCGCAAGAATGAAACTCAAAGGAATTGACGGGGACCC GCACAAGCGGTGGAGCATGTGGTTTAATTCGAAGCAACGCGAAGAACCTTACCAAGTCTTGACATCCGCCTGACCGA TCCTTAACCGGATCTTTCCTTCGGGACAGGCGAGACAGGTGGTGCATGGTTGTCGTCAGCTCGTGTCGTGAGATGTT GGGTTAAGTCCCGCAACGAGCGCAACCCCTATCCTCAGTAGCCAGCATTTAAGGTGGGCACTCTGGGGAGACTGCCA GGGATAACCTGGAGGAAGGCGGGGATGACGTCAAATCATCATGCCCCTTATGATTTGGGCTACACACGTGCTACAAT GGCGTAAACAAAGGGAAGCGAGATCGTGAGATGGAGCAAATCCCAAAAATAACGTCCCAGTTCGGACTGTAGTCTGC AACCCGACTACACGAAGCTGGAATCGCTAGTAATCGCGGATCAGAATGCCGCGGTGAATACGTTCCCGGGTCTTGTA CACACCGCCCGTCACACCATGGGAGTCAGTAACGCCCGAAGTCAGTGACCTAACTGCAAAGAAGGAGCTGCCGAA (MRX006 (strain 830) chromosome sequence)- SEQ ID NO: 5 see electronic sequence listing. (MRX006 (strain 830) plasmid sequence)- SEQ ID NO: 6 see electronic sequence listing. (Blautia hydrogenotrophica strain S5a36 16S ribosomal RNA gene, partial sequence-X95624.1) SEQ ID NO: 7    1 gatgaacgct ggcggcgtgc ttaacacatg caagtcgaac gaagcgatag agaacggaga   61 tttcggttga agttttctat tgactgagtg gcggacgggt gagtaacgcg tgggtaacct  121 gccctataca gggggataac agttagaaat gactgctaat accgcataag cgcacagctt  181 cgcatgaagc ggtgtgaaaa actgaggtgg tataggatgg acccgcgttg gattagctag  241 ttggtgaggt aacggcccac caaggcgacg atccatagcc ggcctgagag ggtgaacggc  301 cacattggga ctgagacacg gcccaaactc ctacgggagg cagcagtggg gaatattgca  361 caatggggga aaccctgatg cagcgacgcc gcgtgaagga agaagtatct cggtatgtaa  421 acttctatca gcagggaaga aagtgacggt acctgactaa gaagccccgg ctaattacgt  481 gccagcagcc gcggtaatac gtaaggggca agcgttatcc ggatttactg ggtgtaaagg  541 gagcgtagac ggtttggcaa gtctgatgtg aaaggcatgg gctcaacctg tggactgcat  601 tggaaactgt cagacttgag tgccggagag gcaagcggaa ttcctagtgt agcggtgaaa  661 tgcgtagata ttaggaggaa caccagtggc gaaggcggcc tgctggacgg taactgacgt  721 tgaggctcga aagcgtgggg agcaaacagg attagatacc ctggtagtcc acgctgtaaa  781 cgatgaatac taggtgtcgg gtggcaaagc cattcggtgc cgcagcaaac gcaataagta  841 ttcccacctg gggagtacgt tcgcaagaat gaaactcaaa ggaattgacg gggacccgca  901 caagcggtgg agcatgtggt ttaattcgaa gcaacgcgaa gaaccttacc aaatcttgac  961 atccctctga ccgggaagta atgttccctt ttcttcggaa cagaggagac aggtggtgca 1021 tggttgtcgt cagctcgtgt cgtgagatgt tgggttaagt cccgcaacga gcgcaaccct 1081 tattcttagt agccagcagg tagagctggg cactctaggg agactgccag ggataacctg 1141 gaggaaggtg gggatgacgt caaatcatca tgccccttat gatttgggct acacacgtgc 1201 tacaatggcg taaacaaagg gaagcgaagg ggtgacctgg agcaaatctc aaaaataacg 1261 tctcagttcg gattgtagtc tgcaactcga ctacatgaag ctggaatcgc tagtaatcgc 1321 gaatcagaat gtcgcggtga atacgttccc gggtcttgta cacaccgccc gtcacaccat 1381 gggagtcagt aacgcccgaa gtcagtgacc caaccnaaag gagggagctg ccgaaggtgg 1441 gactgataac tggggtga

REFERENCES

  • [1] Spor et al. (2011) Nat Rev Microbiol. 9(4):279-90.
  • [2] Eckburg et al. (2005) Science. 10; 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] Kang et al. (2010) Inflamm Bowel Dis. 16(12):2034-42.
  • [9] Machiels et al. (2013) Gut. 63(8):1275-83.[10] Mayer et al (2014) The Journal of Neuroscience 34(46):15490-15496
  • [11] Cryan and Dinan (2015) Neuropsychopharmacology, 40: 241-2.
  • [12] Zhou and Foster (2015) Neuropsychiatric Disease and Treatment 11: 715-723.
  • [13] Wang and Kasper (2014) Brain Behav Immun. 38: 1-12.
  • [14] WO 2013/050792
  • [15] WO 03/046580
  • [16] WO 2013/008039
  • [17] WO 2014/167338
  • [18] Goldin and Gorbach (2008) Clin Infect Dis. 46 Suppl 2:S96-100.
  • [19] Azad et al. (2013) BMJ. 347:f6471.
  • [20] Bravo et al. (2011) Proc Natl Acad Sci USA, 108: 16050-5.
  • [21] Kantak et al. (2014) Behav Pharmacol. 25: 71-9.
  • [22] Savignac et al. (2014) Neurogastroenterol Motil. 26: 1615-27.
  • [23] de Theije et al. (2014) Brain Behav Immun. 37: 197-206.
  • [24] Hsiao et al. (2013) Cell, 155: 1451-63.
  • [25] Meyza and Blanchard (2017) Neurosci Biobehav Rev.
  • [26] Liu et al. (2008) Int J Syst Evol Microbiol 58, 1896-1902.
  • [27] Park et al. (2012) Int J Syst Evol Microbiol. 62(Pt 4):776-9.
  • [28] Liu et al. (2008) Int J Syst Evol Microbiol. 58(Pt 8):1896-902.
  • [29] Masco et al. (2003) Systematic and Applied Microbiology, 26:557-563.
  • [30] Srůtková et al. (2011) J Microbiol. Methods, 87(1):10-6.
  • [31] Bernalier et al. (1996) Arch. Microbiol. 166 (3), 176-183.
  • [32] Wang et al. (2016) J Neurogastroenterol Motil 22: 589-605.
  • [33] Li and Zhou (2016) Neuroscience 324: 131-139.
  • [34] Hyland and Stanton (2016) The Gut-Brain Axis: Dietary, Probiotic and Prebiotic Interventions on the Microbiota (Academic Press).
  • [35] Bourassa et al. (2016) Neuroscience Letters 625, 56-63
  • [36] Miyamoto-Shinohara et al. (2008) J. Gen. Appl. Microbiol., 54, 9-24.
  • [37] Cryopreservation and Freeze-Drying Protocols, ed. by Day and McLellan, Humana Press.
  • [38] Leslie et al. (1995) Appl. Environ. Microbiol. 61, 3592-3597.
  • [39] Mitropoulou et al. (2013) J Nutr Metab. (2013) 716861.
  • [40] Kailasapathy et al. (2002) Curr Issues Intest Microbiol. 3(2):39-48.
  • [41] Handbook of Pharmaceutical Excipients, 2nd Edition, (1994), Edited by A Wade and P J Weller
  • [42] Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985)
  • [43] US 2016/0067188
  • [44] Handbook of Microbiological Media, Fourth Edition (2010) Ronald Atlas, CRC Press.
  • [45] Maintaining Cultures for Biotechnology and Industry (1996) Jennie C. Hunter-Cevera, Academic Press
  • [46] Strobel (2009) Methods Mol Biol. 581:247-61.
  • [47] Gennaro (2000) Remington: The Science and Practice of Pharmacy. 20th edition, ISBN: 0683306472.
  • [48] Molecular Biology Techniques: An Intensive Laboratory Course, (Ream et al., eds., 1998, Academic Press).
  • [49] Methods In Enzymology (S. Colowick and N. Kaplan, eds., Academic Press, Inc.)
  • [50] Handbook of Experimental Immunology, Vols. I-IV (D. M. Weir and C. C. Blackwell, eds, 1986, Blackwell
  • Scientific Publications) [51] Sambrook et al. (2001) Molecular Cloning: A Laboratory Manual, 3rd edition (Cold Spring Harbor Laboratory Press).
  • [52] Handbook of Surface and Colloidal Chemistry (Birdi, K. S. ed., CRC Press, 1997)
  • [53] Ausubel et al. (eds) (2002) Short protocols in molecular biology, 5th edition (Current Protocols).
  • [54] PCR (Introduction to Biotechniques Series), 2nd ed. (Newton & Graham eds., 1997, Springer Verlag)
  • [55] Current Protocols in Molecular Biology (F. M. Ausubel et al., eds., 1987) Supplement 30
  • [56] Smith & Waterman (1981) Adv. Appl. Math. 2: 482-489.
  • [57] Cryan and Mombereau (2004) Mol Psychiatry 9: 326-57.
  • [58] Hyland and Cox (2006) Br J Pharmacol. 146(5): 712-722

Claims

1.-81. (canceled)

82. A method of treating a disease or a condition associated with dysfunction of a microbiota-gut-brain axis in a subject in need thereof, comprising administering to the subject a pharmaceutical composition that comprises a therapeutically effective amount of a bacterial strain of the genus Blautia comprising a 16S rRNA gene sequence that has at least 95% sequence identity to the polynucleotide sequence of SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:7, wherein the administering is effective to treat the disease or the condition associated with dysfunction of the microbiota-gut-brain axis in the subject.

83. The method of claim 82, wherein the disease or the condition associated with the microbiota-gut-brain axis is a disease or a condition of the central nervous system (CNS).

84. The method of claim 82, wherein the disease or the condition associated with the microbiota-gut-brain axis comprises a neurodevelopmental disorder or a neuropsychiatric condition.

85. The method of claim 82, wherein the disease or the condition associated with the microbiota-gut-brain axis comprises autism spectrum disorders (ASDs), child developmental disorder, obsessive compulsive disorder (OCD), major depressive disorder, depression, seasonal affective disorder, anxiety disorders, schizophrenia spectrum disorders, schizophrenia, bipolar disorder, psychosis, mood disorder, chronic fatigue syndrome (myalgic encephalomyelitis), stress disorder, post-traumatic stress disorder, dementia, Alzheimer's disease, Parkinson's disease, chronic pain, motor neuron disease, Huntington's disease, Guillain-Barre syndrome, or meningitis.

86. The method of claim 82, wherein the treating comprises treating one or more symptoms, tissue damages, or behaviors associated with the disease or the condition.

87. The method of claim 82, wherein the bacterial strain modulates a level of commensal metabolites in the subject.

88. The method of claim 82, wherein the bacterial strain increases the level of butyrate in the subject.

89. The method of claim 82, wherein the pharmaceutical composition is formulated for delivery to an intestine of the subject.

90. The method of claim 82, wherein the bacterial strain is live and capable of at least partially colonizing an intestine of the subject.

91. The method of claim 82, wherein the pharmaceutical composition is formulated for oral administration.

92. The method of claim 82, wherein the bacterial strain is lyophilized.

93. The method of claim 82, wherein the therapeutically effective amount comprises from about 1×103 to about 1×1011 colony forming units (CFU).

94. The method of claim 82, wherein the at least 95% sequence identity is determined by a Smith-Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2.

95. The method of claim 82, wherein the pharmaceutical composition comprises one or more pharmaceutically acceptable excipients or carriers.

96. The method of claim 82, wherein the bacterial strain is of the species Blautia stercoris, Blautia wexlerae, or Blautia hydrogenotrophica.

97. The method of claim 82, wherein the bacterial strain comprises a 16S rRNA gene sequence that has at least 98% sequence identity to the polynucleotide sequence of SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:7, as determined by a Smith-Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2.

98. The method of claim 82, wherein the bacterial strain comprises a 16S rRNA gene sequence of SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:7.

99. The method of claim 82, wherein the bacterial strain is the bacterial strain deposited under accession number NCIMB 42381, the bacterial strain deposited under accession number NCIMB 42486, or the bacterial strain deposited under accession number DSM 14294.

100. A method of modulating a microbiota-gut-brain axis in a subject in need thereof, comprising administering to the subject a pharmaceutical composition that comprises a therapeutically effective amount of a bacterial strain of the genus Blautia comprising a 16S rRNA gene sequence that has at least 95% sequence identity to the polynucleotide sequence of SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:7, as determined by a Smith-Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2, wherein the administering is effective to modulate the microbiota-gut-brain axis in the subject.

101. The method of claim 100, wherein the bacterial strain comprises a 16S rRNA gene sequence that has at least 98% sequence identity to the polynucleotide sequence of SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:7, as determined by a Smith-Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2.

Patent History
Publication number: 20220184145
Type: Application
Filed: Nov 19, 2021
Publication Date: Jun 16, 2022
Inventors: Helene SAVIGNAC (Aberdeen), Imke Elisabeth MULDER (Aberdeen), Alexander James STEVENSON (Aberdeen), Ted DINAN (County Cork), John CRYAN (County Cork)
Application Number: 17/530,517
Classifications
International Classification: A61K 35/74 (20060101); A61P 25/28 (20060101); A61P 25/22 (20060101); A61P 25/18 (20060101); A61P 25/24 (20060101); A61P 25/16 (20060101); A61P 25/00 (20060101); A61K 9/00 (20060101); A61K 35/741 (20060101); A61K 9/48 (20060101);