WO2011092261A1 - Probiotic composition for use in the treatment of bowel inflammation - Google Patents

Probiotic composition for use in the treatment of bowel inflammation Download PDF

Info

Publication number
WO2011092261A1
WO2011092261A1 PCT/EP2011/051170 EP2011051170W WO2011092261A1 WO 2011092261 A1 WO2011092261 A1 WO 2011092261A1 EP 2011051170 W EP2011051170 W EP 2011051170W WO 2011092261 A1 WO2011092261 A1 WO 2011092261A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
treatment
strains
cect
strain
Prior art date
Application number
PCT/EP2011/051170
Other languages
French (fr)
Inventor
Jordi Espadaler Mazo
Jordi CUÑÉ CASTELLANA
Original Assignee
Ab-Biotics S.A.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to MX2012008817A priority Critical patent/MX2012008817A/en
Priority to CN201180007399.0A priority patent/CN102905712B/en
Application filed by Ab-Biotics S.A. filed Critical Ab-Biotics S.A.
Priority to UAA201210207A priority patent/UA109123C2/en
Priority to BR112012018813-5A priority patent/BR112012018813B1/en
Priority to ES11702426.5T priority patent/ES2437940T3/en
Priority to US13/575,865 priority patent/US20130101566A1/en
Priority to RU2012136615/10A priority patent/RU2563525C2/en
Priority to JP2012550449A priority patent/JP5777640B2/en
Priority to KR1020127021713A priority patent/KR101840239B1/en
Priority to CA2787544A priority patent/CA2787544C/en
Priority to AU2011209407A priority patent/AU2011209407B2/en
Priority to PL11702426T priority patent/PL2528610T3/en
Priority to EP11702426.5A priority patent/EP2528610B1/en
Publication of WO2011092261A1 publication Critical patent/WO2011092261A1/en
Priority to ZA2012/05683A priority patent/ZA201205683B/en
Priority to US15/134,558 priority patent/US10155015B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • A61K35/747Lactobacilli, e.g. L. acidophilus or L. brevis
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/135Bacteria or derivatives thereof, e.g. probiotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/06Anti-spasmodics, e.g. drugs for colics, esophagic dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/02Nutrients, e.g. vitamins, minerals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/32Foods, ingredients or supplements having a functional effect on health having an effect on the health of the digestive tract
    • A23V2200/3204Probiotics, living bacteria to be ingested for action in the digestive tract
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2400/00Lactic or propionic acid bacteria
    • A23V2400/11Lactobacillus
    • A23V2400/169Plantarum
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2400/00Lactic or propionic acid bacteria
    • A23V2400/41Pediococcus
    • A23V2400/413Acidilactici
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K2035/11Medicinal preparations comprising living procariotic cells
    • A61K2035/115Probiotics

Definitions

  • Probiotic composition for use in the treatment of bowel inflammation
  • the present invention relates to the fields of medicine, microbiology and nutrition and, particularly, to a novel probiotic composition.
  • new strains of Lactobacillus plantarum and Pediococcus acidilactici have been isolated and combined in a formulation useful in the treatment of
  • gastrointestinal diseases such as bowel inflammation (e.g., inflammatory bowel disease) and irritable bowel syndrome.
  • Ulcerative colitis UC
  • Pouchitis and Crohn's Disease are examples of Inflammatory Bowel Diseases (IBD) characterized by chronic inflammation in the intestine.
  • the clinical symptoms are diarrhea, abdominal pain, occasional rectal bleeding, weight loss, tiredness and sometimes fever. Although occurring at any age, IBD is most common in teenagers and young adults, which consequently may suffer from delayed development and stunted growth. The frequency of the disease is similar to type 1 diabetes in Europe and the USA.
  • the clinical course of IBD varies considerably. Patients with mild to moderate symptoms may be treated without hospitalization. However, 10-15 % of patients experience a severe course of the disease, which in many cases is followed by surgery.
  • IBD is treated medically by reducing the inflammation and thereby controlling the gastrointestinal symptoms.
  • Coloectomy may eliminate UC but reduces life quality and increases the risk of complications.
  • the available medical treatments include the use of 5- aminosalicylic acid (5-ASA), corticosteroids and immunomodulatory
  • Prolonged treatment of mild to moderate IBD symptoms is usually carried out using 5-ASA while corticosteroids and immunomodulatory medicaments are used to treat severe symptoms.
  • Diarrhea or abdominal pain appear as side effects of 5-ASA whereas long term use of corticosteroids frequently shows serious side effects including reduction in bone mass, infection, diabetes, muscle wasting and psychiatric disturbances.
  • Immunomodulatory medicaments suppress the immune system, which controls the IBD symptoms. However, the resulting immuno-compromised state leaves the patient susceptible to many diseases.
  • IBS Irritable Bowel Syndrome
  • Probiotics are defined as "living microorganisms, which upon ingestion in certain numbers, exert health benefits beyond inherent basic nutrition" (Araya M. et al., 2002; Guarner F. et al.,, 1998).
  • probiotics Several lactic acid bacteria and species from the genus Bifidobacterium are probiotic, which implies that they have been shown to promote a specific health effect.
  • Probiotic bacteria must fulfil several requirements related to lack of toxicity, viability, adhesion and beneficial effects. These probiotic features are strain-dependent, even among bacteria of the same species. Therefore, it is important to find those strains that have a better performance in all probiotic requirements.
  • WO 96/29083 and EP 554418 disclose two intestine colonizing Lactobacillus strains including Lactobacillus plantarum 299v (DSM 6595) and Lactobacillus casei ssp. rhamnosus 271 (DSM 6594).
  • EP 415941 discloses methods for preparing nutrient composition comprising treatment of oat gruel with enzymes before mixing with lactobacilli.
  • US Patent 7195906 discloses a strain of
  • gastrointestinal inflammatory activity such as IBD
  • IBS gastrointestinal inflammatory activity
  • the inventors of the present invention have found that a composition comprising Lactobacillus and Pediococcus strains is effective in the treatment of bowel inflammation.
  • a composition comprising Lactobacillus and Pediococcus strains is effective in the treatment of bowel inflammation.
  • three new probiotic strains belonging to Lactobacillus plantarum genus and Pediococcus acidilactici genus have been isolated, said strains allowing the efficient treatment of the bowel inflammation when combined into the form of a single formulation.
  • the present invention provides a composition comprising an effective amount of Lactobacillus plantarum CECT 7484, Lactobacillus plantarum CECT 7485, and Pediococcus acidilactici CECT 7483, or mutants or variants thereof.
  • Lactobacillus plantarum strains CECT 7485 and CECT 7484, and
  • Pediococcus acidilactici strain CECT 7483 were deposited in the Spanish Type Culture Collection (Valencia, Spain) on 04.02.2009. All three deposited strains are viable and keep all their features related to their deposit.
  • an effective amount means an amount of an active agent high enough to deliver the desired benefit, but low enough to avoid serious side effects within the scope of medical judgment. It is clear that by using the deposited strains as starting material, the skilled person in the art can routinely, by conventional mutagenesis or re-isolation techniques, obtain further mutants or derivatives thereof that retain or enhance the herein described relevant features and advantages of the strains forming the composition of the invention. The skilled person in the art will decide upon the adequate method to be employed for determining the antiinflammatory, immunomodulatory or anti-IBS or anti-abdominal bloating activity of the strains. Examples of possible methods to measure this activity are shown in the examples below.
  • the mutant is a genetically modified mutant.
  • the variant is a naturally occurring variant.
  • strains forming part of the composition of the first aspect of the invention may be in the form of viable cells.
  • the strain may be in the form of non-viable cells.
  • plantarum CECT 7484 and CECT 7485 are in the form of viable cells.
  • non-viable cells such as killed cultures or compositions containing beneficial factors produced by P. acidilactici CECT 7483, as well as of L. plantarum CECT 7484 and CECT 7485.
  • This could include thermally killed micro-organisms or micro-organisms killed by exposure to altered pH, sonication, radiation or subjection to pressure.
  • product preparation is simpler, cells may be incorporated easily into pharmaceuticals and storage requirements are much less limited than viable cells.
  • the strains are, preferably, in a concentration ratio of 1 : 1 : 1 .
  • Strains CECT 7483, CECT 7484, and CECT 7485 display a significant inhibitory activity against several pathogenic and potentially pathogenic bacterial strains, while displaying minimal antagonism against common commensal strains of the human gastrointestinal flora. Moreover, these three strains show a lack of significant inhibitory activity between them, thus allowing their combined use in a single formula. This is of relevance because this means that the composition as defined in the first aspect of the invention exerts a beneficial effect in the intestine owing to the "intact" effect of each one of the three strains. The combination of these strains into a single formula (i.e.
  • composition of the invention displays the ability to improve clinical symptoms (such as weight loss and diarrhoea) in different animal models of bowel inflammation.
  • clinical symptoms such as weight loss and diarrhoea
  • the composition of the invention shows the unique ability to significantly reduce acute (IL-6) and chronic (IFNy) cytokines.
  • CECT 7484 and CECT 7485 belong to bacterial species that have QPS status (Andreoletti O. et al., 2008).
  • the strains of the present invention have the advantage that they are particularly useful as probiotics.
  • probiotic bacteria must fulfil several requirements related to lack of toxicity, viability, adhesion and beneficial effects. These probiotic features are strain-dependent, even among bacteria of the same species. Therefore, it is important to find those strains that have a better performance in all probiotic requirements.
  • the examples bellow provide (by way of example) protocols to determine each one of the probiotic features and it is also demonstrated that said strains have excellent probiotic features.
  • Intrinsic resistance When resistance to an antimicrobial is inherent to a bacterial species, it is generally referred to as 'intrinsic resistance' (sometimes called 'natural resistance'). Intrinsic resistance is presumed to present a minimal potential for horizontal spread, whereas acquired resistance mediated by added genes is considered as having a high potential for lateral spread.
  • the inventors of the present invention have found that the strains forming the composition of the invention do not display any significant resistance to antibiotics of human and/or veterinary importance (ampicillin, gentamicin, streptomycin,
  • the inventors of the present invention have found that the strains CECT 7483, CECT 7484, and CECT 7485 can be co-administered with other medicaments used for the treatment of IBD (such as mesalazine). As it is shown below, the growth of said strains is not completely inhibited even using saturated concentrations of mesalazine. In other words, even using high concentrations of mesalazine the efficacy of composition of the invention comprising the probiotic strains is not compromise and, therefore, it can exert both the probiotic and anti-inflammation functions.
  • strains of the invention have demonstrated to be highly resistant to the conditions of the gastrointestinal environment of mammals (acidic
  • GIT gastrointestinal tract
  • the present strains have several beneficial effects in the host. In addition to the anti-inflammatory activity in bowel, they benefit the intestinal microbiota balance due to their antagonistic activity.
  • antagonistic activity refers to the inhibition of growth of gastrointestinal non-beneficial bacteria by the activity of probiotic bacteria.
  • SCFA short chain fatty acids
  • SCFAs especially butyric acid
  • SCFAs are readily absorbed by intestinal mucosa, stimulate sodium and water absorption in the colon, and are trophic to the intestinal mucosa (D'Argenio G. et al., 1999; Tedelind S. et al., 2007).
  • butyric acid is used as fuel by colonocytes.
  • Each strain in the formula is a strong producer of a different SCFA, either acetic, propionic or butyric, which are the three major SCFAs found in the intestine.
  • the better understanding of how short chain fatty acids act on inflammation process can help in improving the efficacy of current bowel inflammation treatments. In this regard, it has been reported the relation between SCFAs and the regulation of inflammatory conditions through G protein-coupled receptors (Maslowski K. M. et al., 2009).
  • strains CECT 7483, CECT 7484 and CECT 7485 promote immunomodulatory effects in the host, since they induce an improved cytokine pattern from the intestinal mucosa. These immunomodulatory effects are beneficial to the host because they help to achieve an improved disease resistance and diminished risk of allergies.
  • Gram-negative bacteria in the GIT display the molecule lipopolisaccharide (LPS) in their surface, which induces the production of pro-inflammatory signals by the intestinal mucosa cells. Probiotic supplementation can change this situation to favour one greater presence of Gram positive bacteria in the GIT (grouped in the lactic acid bacteria group), with better aesthetic fitness or with antagonistic properties against some Gram negative microorganisms.
  • LPS lipopolisaccharide
  • cytokines are messenger molecules that regulate the inflammatory and immune responses in the body.
  • some probiotic bacteria induce a better balanced pattern between pro/anti-inflammatory signalling in the intestinal mucosa (regardless of the effect on Gram negative bacteria).
  • the composition's strains of the invention promote a reduction of inflammatory cytokines (IFN-y and IL-6) levels, thus inducing an improved cytokine pattern from the intestinal mucosa.
  • This immunomodulatory effect is complemented by the strain's antagonistic properties in reducing the presence of pathogenic Gram negative bacteria in the GIT.
  • non-viable bacteria as well as bacterial components can have immunomodulatory effects per se. For instance, cell components of
  • Lactobacilli species have been reported to induce anti-inflammatory cytokines (Pathmakanthan S. et al., 2004) or to reduce pro-inflammatory cytokines (Zhang L. et al., 2005). Upon isolation of these components, pharmaceutical grade manipulation is anticipated.
  • strains CECT 7483, CECT 7484 and CECT 7485 are characterized by specific traits such as: survival to
  • composition and isolated strains of the present invention are not obviously derived from the prior art because they are the result of a complex investigation and the results which have been obtained regarding the bowel inflammation activity are surprising. Protocols for determining each one of said traits are included below. From the content of the present application, the skilled in the art could find other strains belonging to Lactobacillus and Pediococcus genus, and more particularly to
  • Lactobacillus plantarum and Pediococcus acidilactici species which, when administered separately or combined into a single composition, show the same probiotic and therapeutic effects than those described in the present application.
  • the invention provides a composition comprising an effective amount of the strains of the invention, or mutant strains thereof, for use as a medicament.
  • the composition comprising the strains CECT 7483, CECT 7484 and CECT 7485 has an anti-inflammatory activity in bowel in IBD models.
  • bowel inflammation is one of the main characteristics of IBD.
  • the composition of the first aspect of the invention is useful in the prevention or treatment of said disease.
  • the invention provides the composition as defined in the first aspect of the invention for use in the prevention or treatment of bowel inflammation in an animal, including a human.
  • This aspect can be alternatively formulated as the use of a composition as defined in the first aspect of the invention for the manufacture of a medicament for the prevention and/or treatment of bowel inflammation.
  • This may be alternatively formulated as a method for the prevention and/or treatment of bowel inflammation in an animal, including a human, comprising administering to said animal in need thereof an effective amount of the composition as defined in the first aspect of the invention.
  • the composition is used for the treatment or prevention of Inflammatory Bowel Disease.
  • the administration of the strains of the invention which are effective in the treatment of conditions characterized by bowel inflammation and diarrhoea, can also be useful to treat other conditions characterised by inflammation of the bowel mucosa or submucosa and where diarrhoea is prevalent, such as enteritis caused by radiotherapy or
  • enteritis is a common side effect of abdominal and pelvic radiotherapy, affecting 60-70% of patients. Enteritis can force schedule changes in the radiotherapy regime to decrease side-effects, potentially leading to sub-optimal anti-tumoral efficacy of the treatment.
  • some probiotics have shown to be promising in randomized clinical trials (RCTs).
  • a probiotic composition such as the one of the present invention, combining the health-promoting effects of SCFAs production, the ability to withstand reactive oxygen and nitrogen species found in the inflamed mucosa, and the antimicrobial activity against opportunistic pathogens can be useful to treat radiotherapy and chemotherapy-induced enteritis.
  • the present inventors have found that the strains of the present invention are efficient in the treatment of IBS.
  • the composition of the first aspect of the invention is useful in treating IBS, as assessed by a randomized double-blind placebo-controlled intervention trial.
  • the present invention provides the composition of the first aspect of the invention for use in the prevention and/or treatment of IBS.
  • This aspect can be alternatively formulated as the use of a composition as defined in the first aspect of the invention for the manufacture of a medicament for the prevention and/or treatment of IBS.
  • This may be alternatively formulated as a method for the prevention and/or treatment of IBS in an animal, including a human, comprising administering to said animal in need thereof an effective amount of the composition as defined in the first aspect of the invention.
  • the present inventors have found that due to the features of the strains, the composition of the first aspect of the invention is useful in the treatment of abdominal bloating and distension. As it is shown below, when the composition of the invention is administered to people suffering from abdominal bloating and distension, it is observed a surprising improvement. Therefore, in a fifth aspect the present invention provides the composition of the first aspect of the invention for use in the treatment of abdominal bloating and distension. This aspect can be alternatively formulated as the use of a composition as defined in the first aspect of the invention for the manufacture of a medicament for the treatment of abdominal bloating and distension.
  • This may be alternatively formulated as a method for the treatment of abdominal bloating and distension in an animal, including a human, comprising administering to said animal in need thereof an effective amount of the composition as defined in the first aspect of the invention.
  • the surprising beneficial effects observed in people suffering from IBS, and/or abdominal bloating and distension may be due to the fact that the strains of the invention CECT 7483, CECT 7484 and CECT 7485 have the ability of producing the SCFAs listed in Table 6 and the antagonistic activity shown in Table 3.
  • SCFAs are known to stimulate serotonin (5-HT) release in rat colon (Fukumoto S. et al., 2003; Tazoe H. et al., 2008) which plays a pivotal role in the regulation of both gut motility and sensation.
  • butyric acid has been described to decrease visceral sensitivity of the intestine in human volunteers (Vanhoutvin S. A. et al., 2009). From this, it can be concluded that the strains forming the composition of the invention can be useful to treat, not only IBS or abdominal pain, but also other conditions related to
  • composition and isolated strains of the present invention are not obviously derived from the prior art because they are the result of a complex investigation and the results which have been obtained regarding the efficiency in the treatment of IBS and abdominal bloating and distension are surprising.
  • composition according to the invention that comprises an effective amount of the strains of the invention, or of their mutants, can be formulated as edible, pharmaceutical or veterinary products, in which said strains are the only active agents or are mixed with one or more other active agents and/or are mixed with pharmaceutically or veterinary acceptable excipients (in the case of a pharmaceutical or veterinary product) or adequate additives (in the case of an edible product).
  • the products additionally contain one or more further active agents.
  • the additional active agent or agents are other probiotic bacteria which are not antagonic to the strains forming the composition of the invention.
  • the strains may be added as purified bacteria, as a bacterial culture, as part of a bacterial culture, as a bacterial culture which has been post-treated, and alone or together with suitable carriers or ingredients. Prebiotics could be also added.
  • the invention provides a pharmaceutical and veterinary products that contain an effective amount of the composition of the invention together with adequate amounts of pharmaceutically or veterinary acceptable excipients.
  • the pharmaceutical product may be prepared in any suitable form which does not negatively affect to the bioavailability of the strains forming the composition of the invention.
  • composition of the invention can be formulated to be administered orally in the form, for instance, of freeze-dried power, capsules, liquid preparations, etc. Selection of the excipients and the most appropriate methods for formulation in view of the particular purpose of the composition is within the scope of ordinary persons skilled in the art of pharmaceutical technology. Although oral administration is preferred, other forms are possible, such as injectable, rectal or topical.
  • Suitable carriers, excipients, etc. can be found in standard pharmaceutical texts.
  • veterinary acceptable means suitable for use in contact with the tissues of a non-human animal.
  • the strains of the invention can be also included in a variety of edible products, such as a milk products, a yogurt, a curd, a cheese (e.g. quark, cream, processed, soft and hard), a fermented milk, a milk powder, a milk based fermented product, an ice-cream, a fermented cereal based product, a milk based powder, a beverage, a dressing, and a pet food.
  • edible product is used herein in its broadest meaning, including any type of product, in any form of presentation, which can be ingested by an animal, but excluding pharmaceutical and veterinary products.
  • meat products e.g.
  • liver paste, frankfurter and salami sausages or meat spreads can be chocolate spreads, fillings (e.g. truffle, cream) and frostings, chocolate, confectionery (e.g. caramel, fondants or toffee), baked goods (cakes, pastries), sauces and soups, fruit juices and coffee whiteners.
  • dietary supplements also include nutraceuticals, which are known to be extracts of foods that have a medicinal effect on human health.
  • Fodders for animal food are also included in the scope of the invention.
  • the compositions of the invention could be also used as an ingredient in other food products.
  • an edible product which contains the composition of the invention together with appropriate amounts of edible ingredients.
  • the composition of the invention is a dietary supplement.
  • colony forming units for each strain in the composition will be determined by the skilled in the art and will depend upon the final formulation. For instance, in edible products, the strain or strains are present in an amount from about 10 5 cfu/g to about 10 12 cfu/g, preferably in an amount from about 10 7 cfu/g to about 10 12 cfu/g, according to the current legislation.
  • colony forming unit (“cfu”) is defined as number of bacterial cells as revealed by microbiological counts on agar plates.
  • Dietary supplements usually contain probiotic strains in an amount ranging from 10 7 and 10 12 cfu/g.
  • the composition of the invention is a dietary supplement comprising between 10 9 -10 11 cfu/g.
  • the strains of the invention are produced by cultivating the bacteria in a suitable medium and under suitable conditions.
  • the strains can be cultivated alone to form a pure culture, or as a mixed culture together with other microorganisms, or by cultivating bacteria of different types separately and then combining them in the desired proportions.
  • the cell suspension is recovered and used as such or treated in the desired manner, for instance, by concentrating or freeze-drying, to be further employed in the preparation of pharmaceutical or edible products.
  • the probiotic preparation is subjected to an immobilisation or encapsulation process in order to improve the shelf life.
  • immobilisation or encapsulation of bacteria are known in the art.
  • composition according to the invention can be administered as such, can be mixed with a suitable drinkable liquid, such as water, yoghurt, milk or fruit juice, or can be mixed with solid or liquid food.
  • a suitable drinkable liquid such as water, yoghurt, milk or fruit juice
  • the dietary supplement can be in the form of tablets, pills, capsules, granules, powders, suspensions, sachets, pastilles, sweets, bars, syrups and corresponding administration forms, usually in the form of a unit dose.
  • the composition of the invention is administered in the form of tablets, capsules or powders, manufactured in conventional processes of preparing pharmaceutical products.
  • FIG. 1 represents the pulsed field gel electrophoresis patterns of Not-I or Sfi-I (left) and Sma-I (right) restricted genomic DNA of: 1 , Pediococcus acidilactici CECT 7483; 2, Lactobacillus plantarum CECT 7484; 3, Lactobacillus plantarum CECT 7485. As controls: 4, commercial strain P. acidilactici
  • FIG. 2 represents the Disease Activity Index (Y-axis) in a group of mice suffering from a DSS-induced intestinal inflammation.
  • X-axis represents the administration of: a, probiotic formulation of the invention in a group of mice suffering from a DSS-induced intestinal inflammation; b, a commercial probiotic formulation (VSL#3) in a group of mice suffering from a DSS-induced intestinal inflammation; c, vehicle in a group of mice suffering from a DSS- induced intestinal inflammation; and d, vehicle in a healthy control group.
  • FIG. 3 represents the levels of IL-6 (Y-axis) in a group of mice suffering from a DSS-induced intestinal inflammation.
  • X-axis represents: a, probiotic
  • formulation of the invention administered to a group of mice suffering from a DSS-induced intestinal inflannnnation; b, a commercial probiotic formulation (VSL#3) administered to a group of mice suffering from a DSS-induced intestinal inflammation; c, vehicle administered to a group of mice suffering from a DSS-induced intestinal inflammation; and d, vehicle in a healthy control group.
  • VSL#3 a commercial probiotic formulation administered to a group of mice suffering from a DSS-induced intestinal inflammation
  • vehicle administered to a group of mice suffering from a DSS-induced intestinal inflammation
  • d vehicle in a healthy control group.
  • FIG. 4 represents the number of Symptom-free weeks (i.e. number of weeks before the onset of the first symptom, thus being the Disease Activity Index equal to zero) (Y-axis) in a IL-10 knock-out mouse model.
  • X-axis represents the administration of: a, the probiotic composition of the invention to a IL-10 knock-out mice group; b, the commercial probiotic formulation VSL#3 to a IL- 10 knock-out mice group; c, PBS to a IL-10 knock-out mice group; and d, vehicle in a healthy control group.
  • This figure refers to the "In Vivo Effect on Spontaneous Gut Inflammation" section.
  • FIG. 5 represents the levels of IFN- ⁇ (Y-axis) in a IL-10 knock-out mouse model.
  • X-axis represents: a, probiotic formulation of the invention to a IL-10 knock-out mice group; b, a commercial probiotic formulation (VSL#3) to a IL- 10 knock-out mice group; c, vehicle to a IL-10 knock-out mice group; and d, vehicle to a healthy control group.
  • VSL#3 commercial probiotic formulation
  • FIG. 6 represents the levels of IL-6 (Y-axis) in a IL-10 knock-out mouse model.
  • X-axis represents: a, probiotic formulation of the invention to a IL-10 knock-out mice group; b, a commercial probiotic formulation (VSL#3) , to a IL- 10 knock-out mice group; c, vehicle administered to a IL-10 knock-out mice group; and d, vehicle to a healthy control group.
  • VSL#3 commercial probiotic formulation
  • FIG. 7 represents the variation of the IBSQoL score compared to baseline (Y- axis) of volunteers treated with capsules including the composition (black bars) or placebo (white bars).
  • X-axis represents the variation of the score 21 days and after 42 days of treatment. This figure refers to the "Improvement of Health-Related Quality of Life” in the "In Vivo Efficacy on IBS Subjects” section.
  • FIG. 8 represents the variation of the VSI score compared to baseline (Y-axis) of volunteers treated with capsules including the composition (black bars) or placebo (white bars).
  • X-axis represents the variation of the score after three weeks and after six weeks of treatment. This figure refers to the "Improvement of the Visceral Sensitivity" in the "In Vivo Efficacy on IBS Subjects” section.
  • strain F1033 corresponds to Pediococcus acidilactici CETC 7483, strain F2064 to
  • Lactobacillus plantarum CECT 7484 and strain F2076 to Lactobacillus plantarum CECT 7485.
  • Novel strains F2064, F2076 and F1033 were grown on MRS agar
  • strains F2064 and F2076 are Gram-positive bacilli, while strain F1033 is a Gram-positive with coccal morphology.
  • Genomic DNA was extracted using Wizard genomic DNA purification kit (Promega). For each isolated strain, the 16S gene was amplified by PCR, using the universal primers 27f, 357f, 907r and 1492r (Weisburg W. G. et al., 1991 ), which generate a nearly full-length 16S rRNA fragment (1465 bp). DNA was washed using Quiaquick kit (Quiagene, GmbH, Hilden, Germany) and four sequencing reactions were performed per sample, using BigDye v.3.1 kit, on a Genetic Analyzer 3130 (Applied Biosystems).
  • Strains F2064 and F2076 were identified as members of the Lactobacillus plantarum group. Strain F1033 was identified as Pediococcus acidilactici.
  • Strains also posses a marked resistance to bile salts, with less than a 50% reduction in the number of viable cells after 3 h incubation in MRS supplemented with 0.5% of bile salts.
  • Porcine intestine was washed with PBS pH 7.4, containing 0.01 % gelatin and a cocktail of protease inhibitors (Complete®, Sigma).
  • the mucosa was scrapped and dissolved in HEPES-Hank ' s buffer (10 mM HEPES, pH 7.4) (Collado M. et al., 2007) containing the aforementioned inhibitors. Then, mucus was centrifuged at 13000 rpm for 10 min using the same buffer.
  • Each strain to be tested was grown overnight in MRS medium supplemented with tritium-labeled thymidine (5 ⁇ in 3 ml of MRS). Cultures were centrifuged and adjusted to 10 8 cfu/ml in PBS by counting on a Neubauer chamber and samples of each culture were taken to determine the amount of tritium-labeled thymidine incorporated by means of a scintillation reader. Then, 0.5 ml were added to the mucus-containing wells of the 24-well plate and incubated at 37 °C for 60 min. Supernatant of each well was removed, and wells were washed twice with MEM Alpha medium (Gibco) to remove loosely adherent bacteria.
  • MRS medium MEM Alpha medium
  • Caco-2 cells were obtained from ATCC (ECACC N°: 86010202). Cells were seeded in 24-well plates and allowed to grow in DMEM until confluence (37 °C, 5% CO 2 ). The experimental procedure to obtain the number of bacteria that adhere per unit of caco-2 cells area is essentially the same as the one explained above for adhesion to mucus.
  • Adhesion capacity of strains F1033, F2064 and F2076 was measured from scintillation of tritium-labeled thymidine and compared to those of the commercial strain L. rhamnosus GG.
  • Adhesion to epithelial cells using the Caco-2 model is a common assay for probiotic strains. Compared to L rhamnosus GG, strains F2064 and F2076 show an affinity for epithelial cells 60% lower. However, considering the high affinity of L. rhamnosus GG for epithelial cells, these values are comparable to other well known probiotics such as L. plantarum 299v, and superior to many other probiotic strains (Jacobsen C. N. et al., 1999).
  • strain F1033 adhesion of strain F1033 to epithelial cells is 2.5 times higher than L. rhamnosus GG.
  • strains F2064 and F2076 displayed a much higher affinity for intestinal mucus than for epithelial cells, while strain F1033 showed the opposite behavior. Results are shown in the following table.
  • the following indicator strains were used: P. mirabilis CECT 4557, K. oxytoca CIP 103434, C. perfrinqens ATCC 13124.
  • C. albicans. S. enterica thyphimurium, S. enterica cholerasuis, C. jejuni. E. coli and P_. aeruginosa were lab isolates.
  • Indicator strains were swabbed uniformly in plates containing the appropriate medium (Oxoid) and grown to confluence at the appropriate temperatures in microaerophilic conditions (5% CO 2 ). Then, 6 mm (diameter) cylinder sections of confluent F1033, F2064 or F2076 agar plates were placed upside-down on the indicator strain plate and incubated overnight at 37 °C. The next day, inhibition zones were measured by placing the agar plate over a flat rule. Growth inhibitory activity (Gl) was calculated as follows:
  • Strains F2064, F2076 and F1033 displayed significant inhibitory activity against Candida albicans and several potentially pathogenic bacteria. On the other hand, the strains displayed minimal activity against commensal strains commonly found in the indigenous gastrointestinal flora of the Bacteroides genus. Also, strains F2064, F2076 and F1033 did not display significant inhibitory activity among them. It is noteworthy that strain F1033 is the only strain displaying high inhibitory activity against Campylobacter jejuni, while strain F2076 outstands in inhibiting Escherichia coli and strain F2064 in inhibiting both Candida albicans and Proteus mirabilis. 6. Antioxidant Capacity
  • strain F1033, F2064 and F2076 showed a capacity to survive under strong oxidizing conditions comparable to the well-known probiotic strain L. rhamnosus GG, as well as to the L. plantarum strain isolated from the VSL#3 formula. It is worth noting that strain F2076 displayed the highest resistance both to paraquat (superoxide anion donor) and sodium nitroprusside (nitric oxide donor). Resistance to oxidative stress is a desirable trait for probiotic strains that are expected to survive in the environment of an inflamed mucosa. TABLE 4: Percent of growth in medium containing 10 mM of paraquat or sodium nitroprusside, compared to standard MRS medium.
  • FIG. 1 shows the pulse-field electrophoresis profiles obtained.
  • Strain F1033 shows a similar genomic restriction profile to P. acidilactici R1001 after digestion with Sma-I.
  • the genomic profile obtained after digestion with enzyme Not-I is clearly different.
  • the genomic restriction profiles of strains F2064 and F2076 are clearly different among them and also compared both to L. plantarum 299v and to the L. plantarum strain contained in the VSL#3 formula.
  • each one inulin, pectin and FOS in a specific amount, under microaerophilic conditions (5% CO 2 ) at 37 °C.
  • cells were removed by centrifugation at 12.000 rpm for 10 min and
  • SCFAs Short chain fatty acids
  • SCFAs are the end products of anaerobic bacteria break down of carbohydrates in the large bowel. SCFAs, mainly acetate, propionate and butyrate account for approximately 80% of the colonic anion concentration and are produced in nearly constant molar ratio 62:22:15.
  • SCFAs especially butyric acid, but also acetic and propionic acid
  • SCFAs are readily absorbed by intestinal mucosa, are relatively high in caloric content, are metabolized by colonocytes and hepatocytes, stimulate sodium and water absorption in the colon and are trophic to the intestinal mucosa (D'Argenio G. et al., 1999).
  • high amounts of acetic acid have long been known to be irritant to the intestinal mucosa (Yamada Y. et al., 1992).
  • Strains F1033, F2064 and F2076 are strong producers of either acetic, propionic or butyric acid.
  • DSS dextran sodium sulfate
  • Probiotics were administered by oral gavage for ten days before (day -10) starting DSS administration (day 0). Each mouse received daily 2.5 x 10 8 cfus of probiotic in 0.1 ml_ of sterilized water (vehicle) by gavage. Non- probiotic treated mice received the same volume of vehicle (distilled water with 15% skim milk and 4% sucrose).
  • mice were fed with 3% (w/v) DSS (mol. Wt 40kD, Applichem Lifescience, VWR, Barcelona) in their drinking water for 5 days (days 0 to 4, followed by three days without DSS) according to a previously described method with minor modifications (Okayasu I, et al. Gastroenterol 1990). Healthy controls never received DSS.
  • DSS mol. Wt 40kD, Applichem Lifescience, VWR, Barcelona
  • the Disease Activity Index score used hereby was first described by Cooper et al. and combines several clinical symptoms into one normalized score (Cooper H. S. et al., 1993). Maximum score is 12 points. This score has been widely used to evaluate the efficacy of experimental treatments - probiotics among them - in animal models of IBD (Fitzpatrick L. R. et al., 2007; Grabig A. et al., 2006; Sasaki M. et al., 2005). After being sacrificed by anesthetic overdose of inhaled Halothane
  • Luminex® Platform (Luminex® Co, Austin, USA). Fluorescent microparticle beads, pre-spotted with cytokine-specific antibodies, were incubated with 50 ⁇ 1 :5 diluted supernatant. Specific-biotinylated secondary antibodies and streptavidin-phycoerythrin (S-PE) were sequentially added. Data were expressed as pg of cytokine per mg of protein (Quick Start Bradford Protein Assay, BIO-RAD, CA, USA). All measurements were done in duplicate. B) Results
  • the group receiving the probiotic formula of the invention displayed a significant improvement of the clinical symptoms when compared to DSS-treated controls, as assessed by the Disease Activity Index (p ⁇ 0.05, two-tail ANOVA with Tukey-Kramer post-hoc test). Healthy controls also displayed a lower Disease Activity Index (p ⁇ 0.05).
  • probiotic formula of the invention significantly decreased IL-6 when compared to DSS- treated controls (p ⁇ 0.01 , two-tail ANOVA with Tukey-Kramer post-hoc test), while the effect of commercial probiotic formula VSL#3 failed to achieve significance (p > 0.05).
  • IL-6 is a marker of acute inflammation (FIG. 3).
  • levels of IL-6 in healthy controls were also significantly lower than DSS-treated controls (p ⁇ 0.05).
  • a statistically significant correlation was found between clinical symptoms (DAI score) and IL-6 levels in the intestinal mucosa (p ⁇ 0.05, Spearman ranks test) (data not shown).
  • correlation between clinical symptoms and IL-10, IL-23, TNFa or IFNy was not statistically significant, and the probiotic formula of the invention did not significantly affect the levels of these cytokines.
  • Interleukin 10 is an important regulatory cytokine that supresses effector functions of macrophage/monocytes, T helper 1 (Th1 ) cells, and natural killer cells.
  • Th1 T helper 1
  • IL-10 augments proliferation and differentiation of B cells.
  • the IL-10 knock-out mouse has been widely used to evaluate new therapeutic options for IBD.
  • mice were kept under specific pathogen-free (SPF) conditions in an isolator (Harlan Iberica, Barcelona, Spain) at constant temperature (22°C) in a 12-hour of light/dark cycle.
  • Mice had free access to sterilized diets (diet based in AIN-93 for maintenance of mice was composed by 12% of water, 14,5% of protein, 4% of fat, 4,5% of fibre and 4,7% of ash; Harlan Interfauna Iberica S.A., Barcelona, Spain) and to drinking fluid.
  • mice Sixteen-weeks-old mice were sacrificed by anaesthetic overdose of inhaled Halothane (Fluotane®, Zeneca Ltd, UK). Colon samples of the animals were harvested and washed in cold PBS. Blood samples were also collected by cardiac puncture to analyze hematocrit and hemoglobin concentration (Coulter MaxM Analyzer with autoloader, Izasa, Spain). Colon weight/length ratio was recorded. Then, colons were frozen in liquid nitrogen and cytokines IL-6, and IFNy were measured using the same protocol as in the model of DSS-induced gut inflammation (see above). B) Results
  • probiotic composition of the invention significantly decreased IFNy levels in knockout mice when compared both to vehicle-treated knock-outs (p ⁇ 0.01 , two-tail nonparametric ANOVA with Dunn post-hoc test) and to commercial formula VLS#3 (p ⁇ 0.05).
  • the levels of IFNy attained the same levels as those of wild-type healthy controls.
  • FIG. 6 there was also a clear tendency of probiotic formulas to reduce the levels of IL-6, although results did not reach significance due to the large standard deviation among vehicle knockout mice.
  • Hydroxymethyl propyl cellulose capsules were filled with: (1 ) 150 mg of maltodextrin, (2) 5 mg of magnesium stearate, (3) 5 mg of silicon dioxide and (4) 200 mg of a 1 :1 :1 mixture of the three strains of the invention (at a concentration 5-10 10 cfus/capsule).
  • a placebo was made with the same list of excipients and amounts but without including the composition of the invention. Content of the capsules throughout the study ranged from 5-10 10 to 1 -10 10 cfus.
  • HRQOL health-related quality of life
  • the composition of the invention significantly improved health-related quality of life compared to placebo when assessed both after 21 days and 42 days of treatment (p ⁇ 0.05, T-test). Therefore, it is demonstrated that the composition of the present invention significantly reduces morbility and improves the quality of life of IBS subjects well above the placebo effect.
  • the positive effects of the composition include the food-related distress, anxiety, interference in daily activities and sleep disturbance domains of the HRQOL questionnaire.
  • the improvements in these scales suggest a reduction in abdominal pain, discomfort and altered bowel habits. To our knowledge, this is the first time that it is shown a probiotic composition displaying a significant effect on the global health-related quality of life of IBS patients. Improvement in the Visceral Sensitivity Index (FIG. 8)
  • composition of the present invention significantly reduced the
  • TABLE 10 shows the numbers of subjects reporting a significant improvement related to bloating and distension (as defined by an increase of at least two points compared to baseline in the 6-point scale of the VSI questionnaire that measures bloating and distension-related anxiety) at the end of the treatment. The difference between the two groups is statistically significant (p ⁇ 0.05, Fisher's exact test).
  • composition of the invention is effective in treating abdominal distension and bloating.
  • This example further supports the use of the composition of the invention to treat abdominal bloating and altered intestinal motility in subjects which are not classified as having Irritable Bowel Syndrome.
  • Vanhoutvin, S.A., et al. "The effects of butyrate enemas on visceral perception in healthy volunteers", Neurogastroenterology & Motility, 2009, vol. 21 , p. 952- e976.
  • Yamada, Y., et al. "A comparative analysis of two models of colitis in rats", Gastroenterology, 1992, vol. 102, p. 1524-1534.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Mycology (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Microbiology (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nutrition Science (AREA)
  • Polymers & Plastics (AREA)
  • Food Science & Technology (AREA)
  • Immunology (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Coloring Foods And Improving Nutritive Qualities (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The composition comprises an effective amount of Lactobacillus plantarum CECT 7484, Lactobacillus plantarum CECT 7485, and Pediococcus acidilactici CECT 7483. This composition is useful in the treatment of gastrointestinal diseases or conditions such as Inflammatory Bowel Disease, Irritable Bowel Syndrome or abdominal distension and bloating.

Description

Probiotic composition for use in the treatment of bowel inflammation
This application claims the benefit of the U.S. provisional application No. 61/299,1 16, and of the European patent application EP10151998, both filed on 28th January 2010, and which are incorporated herein by reference.
The present invention relates to the fields of medicine, microbiology and nutrition and, particularly, to a novel probiotic composition. Particularly, new strains of Lactobacillus plantarum and Pediococcus acidilactici have been isolated and combined in a formulation useful in the treatment of
gastrointestinal diseases, such as bowel inflammation (e.g., inflammatory bowel disease) and irritable bowel syndrome.
BACKGROUND OF THE INVENTION
Ulcerative colitis (UC), Pouchitis, and Crohn's Disease are examples of Inflammatory Bowel Diseases (IBD) characterized by chronic inflammation in the intestine. The clinical symptoms are diarrhea, abdominal pain, occasional rectal bleeding, weight loss, tiredness and sometimes fever. Although occurring at any age, IBD is most common in teenagers and young adults, which consequently may suffer from delayed development and stunted growth. The frequency of the disease is similar to type 1 diabetes in Europe and the USA. The clinical course of IBD varies considerably. Patients with mild to moderate symptoms may be treated without hospitalization. However, 10-15 % of patients experience a severe course of the disease, which in many cases is followed by surgery.
IBD is treated medically by reducing the inflammation and thereby controlling the gastrointestinal symptoms. However, there is currently no medical cure for IBD. Coloectomy may eliminate UC but reduces life quality and increases the risk of complications. The available medical treatments include the use of 5- aminosalicylic acid (5-ASA), corticosteroids and immunomodulatory
medicaments. Prolonged treatment of mild to moderate IBD symptoms is usually carried out using 5-ASA while corticosteroids and immunomodulatory medicaments are used to treat severe symptoms. Diarrhea or abdominal pain appear as side effects of 5-ASA whereas long term use of corticosteroids frequently shows serious side effects including reduction in bone mass, infection, diabetes, muscle wasting and psychiatric disturbances.
Immunomodulatory medicaments suppress the immune system, which controls the IBD symptoms. However, the resulting immuno-compromised state leaves the patient susceptible to many diseases.
Irritable Bowel Syndrome (IBS) is a condition characterized by abdominal pain and/or discomfort which is associated to altered bowel habit or defecation, where symptoms are not explained by structural or biochemical abnormalities. Urgency, bloating and feeling of incomplete bowel movements are also common in IBS. Therefore, it is classified among functional gastrointestinal disorders which include diseases such as functional bloating, noncardiac chest pain, non-ulcer dyspepsia, and chronic constipation or diarrhea
(Longstreth G. H. et al., 2006). Noteworthy, IBS has a substantial impact on morbidity and quality of life beyond abdominal pain and discomfort, as the associated symptoms affect both the sufferer's sense of well-being and the ability to function normally (Dean B. B. et al., 2005).
There is a tremendous activity in the field of drug development for the treatment of IBS. In this regard, various antidepressants have gained popularity although their efficacy in clinical trials has been modest and their clinical utility is limited by untoward side effects. Serotonergic agents have demonstrated efficacy on the global symptoms of IBS. However, recent concerns about safety have severely limited their use. Therefore, the development of novel therapies for IBS is of great interest.
Probiotics are defined as "living microorganisms, which upon ingestion in certain numbers, exert health benefits beyond inherent basic nutrition" (Araya M. et al., 2002; Guarner F. et al.,, 1998). Several lactic acid bacteria and species from the genus Bifidobacterium are probiotic, which implies that they have been shown to promote a specific health effect. Probiotic bacteria must fulfil several requirements related to lack of toxicity, viability, adhesion and beneficial effects. These probiotic features are strain-dependent, even among bacteria of the same species. Therefore, it is important to find those strains that have a better performance in all probiotic requirements. Human clinical trials using probiotics alone or in combination with antibiotics have been performed to identify strains and/or formulations for the treatment of patients with IBD or IBS, or for keeping already treated IBD patients in remission. WO 96/29083 and EP 554418 disclose two intestine colonizing Lactobacillus strains including Lactobacillus plantarum 299v (DSM 6595) and Lactobacillus casei ssp. rhamnosus 271 (DSM 6594). EP 415941 discloses methods for preparing nutrient composition comprising treatment of oat gruel with enzymes before mixing with lactobacilli. US Patent 7195906 discloses a strain of
Bifidobacterium isolated from resected and washed human gastrointestinal tract for the treatment of inflammatory diseases, especially gastrointestinal inflammatory activity, such as IBD, and IBS. In spite of a promising potential, a considerably improvement of the effect of probiotics for use in the treatment of inflammation bowel diseases (such as IBD) as well as of other gastrointestinal diseases (such as IBS) is needed.
SUMMARY OF THE INVENTION
The inventors of the present invention have found that a composition comprising Lactobacillus and Pediococcus strains is effective in the treatment of bowel inflammation. Particularly, three new probiotic strains belonging to Lactobacillus plantarum genus and Pediococcus acidilactici genus have been isolated, said strains allowing the efficient treatment of the bowel inflammation when combined into the form of a single formulation.
Thus, in a first aspect the present invention provides a composition comprising an effective amount of Lactobacillus plantarum CECT 7484, Lactobacillus plantarum CECT 7485, and Pediococcus acidilactici CECT 7483, or mutants or variants thereof.
Lactobacillus plantarum strains CECT 7485 and CECT 7484, and
Pediococcus acidilactici strain CECT 7483 were deposited in the Spanish Type Culture Collection (Valencia, Spain) on 04.02.2009. All three deposited strains are viable and keep all their features related to their deposit.
The term "effective amount" as used herein, means an amount of an active agent high enough to deliver the desired benefit, but low enough to avoid serious side effects within the scope of medical judgment. It is clear that by using the deposited strains as starting material, the skilled person in the art can routinely, by conventional mutagenesis or re-isolation techniques, obtain further mutants or derivatives thereof that retain or enhance the herein described relevant features and advantages of the strains forming the composition of the invention. The skilled person in the art will decide upon the adequate method to be employed for determining the antiinflammatory, immunomodulatory or anti-IBS or anti-abdominal bloating activity of the strains. Examples of possible methods to measure this activity are shown in the examples below.
In one embodiment, the mutant is a genetically modified mutant.
In another embodiment of the first aspect of the invention, the variant is a naturally occurring variant.
The strains forming part of the composition of the first aspect of the invention may be in the form of viable cells. Alternatively, the strain may be in the form of non-viable cells. The general use of strains P. acidilactici CECT 7483, as well as of L
plantarum CECT 7484 and CECT 7485 are in the form of viable cells.
However, it could also be extended to non-viable cells such as killed cultures or compositions containing beneficial factors produced by P. acidilactici CECT 7483, as well as of L. plantarum CECT 7484 and CECT 7485. This could include thermally killed micro-organisms or micro-organisms killed by exposure to altered pH, sonication, radiation or subjection to pressure. With non-viable cells product preparation is simpler, cells may be incorporated easily into pharmaceuticals and storage requirements are much less limited than viable cells.
When used in the form of the composition of the invention, the strains are, preferably, in a concentration ratio of 1 : 1 : 1 .
Strains CECT 7483, CECT 7484, and CECT 7485 display a significant inhibitory activity against several pathogenic and potentially pathogenic bacterial strains, while displaying minimal antagonism against common commensal strains of the human gastrointestinal flora. Moreover, these three strains show a lack of significant inhibitory activity between them, thus allowing their combined use in a single formula. This is of relevance because this means that the composition as defined in the first aspect of the invention exerts a beneficial effect in the intestine owing to the "intact" effect of each one of the three strains. The combination of these strains into a single formula (i.e. the composition of the invention) displays the ability to improve clinical symptoms (such as weight loss and diarrhoea) in different animal models of bowel inflammation. In line with these results, the composition of the invention shows the unique ability to significantly reduce acute (IL-6) and chronic (IFNy) cytokines.
A wide variety of lactic acid bacterial species have a long history of apparent safe use. The European Food Safety Authority has developed a system granting the "Qualified Presumption of Safety" (QPS) status to taxonomical units with a proven long history of apparent safe use. Strains CECT 7483,
CECT 7484 and CECT 7485 belong to bacterial species that have QPS status (Andreoletti O. et al., 2008).
The strains of the present invention have the advantage that they are particularly useful as probiotics. As mentioned above, probiotic bacteria must fulfil several requirements related to lack of toxicity, viability, adhesion and beneficial effects. These probiotic features are strain-dependent, even among bacteria of the same species. Therefore, it is important to find those strains that have a better performance in all probiotic requirements. The examples bellow provide (by way of example) protocols to determine each one of the probiotic features and it is also demonstrated that said strains have excellent probiotic features.
The emergence and spread of resistance to antimicrobials in bacteria pose a threat to human and animal health and present a major financial and societal cost. When resistance to an antimicrobial is inherent to a bacterial species, it is generally referred to as 'intrinsic resistance' (sometimes called 'natural resistance'). Intrinsic resistance is presumed to present a minimal potential for horizontal spread, whereas acquired resistance mediated by added genes is considered as having a high potential for lateral spread. The inventors of the present invention have found that the strains forming the composition of the invention do not display any significant resistance to antibiotics of human and/or veterinary importance (ampicillin, gentamicin, streptomycin,
erythromycin, tetracycline, clindamycin, and chloramphenicol) according to the guidelines of the European Food Safety Authority (Anadon A. et al., 2005; Bories G. et al., 2008), thus precluding the risk of a potential transfer of antibiotic resistance to pathogenic species.
Additionally, the inventors of the present invention have found that the strains CECT 7483, CECT 7484, and CECT 7485 can be co-administered with other medicaments used for the treatment of IBD (such as mesalazine). As it is shown below, the growth of said strains is not completely inhibited even using saturated concentrations of mesalazine. In other words, even using high concentrations of mesalazine the efficacy of composition of the invention comprising the probiotic strains is not compromise and, therefore, it can exert both the probiotic and anti-inflammation functions.
The strains of the invention have demonstrated to be highly resistant to the conditions of the gastrointestinal environment of mammals (acidic
environment, bile salts, and high lysozyme, and oxygen peroxide
concentrations), thus being able to survive passage through the
gastrointestinal tract (hereinafter also referred as "GIT"). The strains also have good adhesion to the intestinal epithelium, which allows them to remain in the intestinal tract and to exert their probiotic effects.
Further, the present strains have several beneficial effects in the host. In addition to the anti-inflammatory activity in bowel, they benefit the intestinal microbiota balance due to their antagonistic activity. The term "antagonistic activity" refers to the inhibition of growth of gastrointestinal non-beneficial bacteria by the activity of probiotic bacteria. The condition of having
inadequate gastrointestinal microbial balance is known as disbiosis and has multiple negative consequences for human well-being. It will be shown below that the strains have a high capacity to inhibit the growth of pathogenic strains when compared to other commercial strains. Additionally, as mentioned above, the inventors have found that the new strains of the invention do not display significant inhibitory activity among them.
Additionally, it has been found that the strains forming the composition of the first aspect of the invention produce large quantities of short chain fatty acids (SCFA). Production of SCFA from non-digestible fibres is an interesting probiotic trait. This trait is desirable in a probiotic because the produced SCFA shows several beneficial properties in the host. Among their various
properties, SCFAs, especially butyric acid, are readily absorbed by intestinal mucosa, stimulate sodium and water absorption in the colon, and are trophic to the intestinal mucosa (D'Argenio G. et al., 1999; Tedelind S. et al., 2007). Moreover, butyric acid is used as fuel by colonocytes. Each strain in the formula is a strong producer of a different SCFA, either acetic, propionic or butyric, which are the three major SCFAs found in the intestine. The better understanding of how short chain fatty acids act on inflammation process can help in improving the efficacy of current bowel inflammation treatments. In this regard, it has been reported the relation between SCFAs and the regulation of inflammatory conditions through G protein-coupled receptors (Maslowski K. M. et al., 2009).
Furthermore, the strains CECT 7483, CECT 7484 and CECT 7485 promote immunomodulatory effects in the host, since they induce an improved cytokine pattern from the intestinal mucosa. These immunomodulatory effects are beneficial to the host because they help to achieve an improved disease resistance and diminished risk of allergies. It is known that Gram-negative bacteria in the GIT display the molecule lipopolisaccharide (LPS) in their surface, which induces the production of pro-inflammatory signals by the intestinal mucosa cells. Probiotic supplementation can change this situation to favour one greater presence of Gram positive bacteria in the GIT (grouped in the lactic acid bacteria group), with better ecologic fitness or with antagonistic properties against some Gram negative microorganisms. Nevertheless, some probiotic microorganisms show the ability to modulate per se the production of cytokines, which are messenger molecules that regulate the inflammatory and immune responses in the body. Particularly, some probiotic bacteria induce a better balanced pattern between pro/anti-inflammatory signalling in the intestinal mucosa (regardless of the effect on Gram negative bacteria). As will be illustrated below, it was found that the composition's strains of the invention promote a reduction of inflammatory cytokines (IFN-y and IL-6) levels, thus inducing an improved cytokine pattern from the intestinal mucosa. This immunomodulatory effect is complemented by the strain's antagonistic properties in reducing the presence of pathogenic Gram negative bacteria in the GIT. It is known that non-viable bacteria as well as bacterial components can have immunomodulatory effects per se. For instance, cell components of
Lactobacilli species have been reported to induce anti-inflammatory cytokines (Pathmakanthan S. et al., 2004) or to reduce pro-inflammatory cytokines (Zhang L. et al., 2005). Upon isolation of these components, pharmaceutical grade manipulation is anticipated.
Considering the results shown below, it is clear that the strains CECT 7483, CECT 7484 and CECT 7485, forming the composition of the first aspect of the invention, are characterized by specific traits such as: survival to
gastrointestinal passage, adherence to intestinal mucosa, resistance to oxidative stress, production of metabolites with anti-inflammatory activity (either short chain fatty acids or other products with said activity) and absence of antagonism between them. The composition and isolated strains of the present invention are not obviously derived from the prior art because they are the result of a complex investigation and the results which have been obtained regarding the bowel inflammation activity are surprising. Protocols for determining each one of said traits are included below. From the content of the present application, the skilled in the art could find other strains belonging to Lactobacillus and Pediococcus genus, and more particularly to
Lactobacillus plantarum and Pediococcus acidilactici species which, when administered separately or combined into a single composition, show the same probiotic and therapeutic effects than those described in the present application.
In a second aspect, the invention provides a composition comprising an effective amount of the strains of the invention, or mutant strains thereof, for use as a medicament. Particularly, it has been found that the composition comprising the strains CECT 7483, CECT 7484 and CECT 7485 has an anti-inflammatory activity in bowel in IBD models. As explained above, bowel inflammation is one of the main characteristics of IBD. Thus, the composition of the first aspect of the invention is useful in the prevention or treatment of said disease.
Therefore, in a third aspect, the invention provides the composition as defined in the first aspect of the invention for use in the prevention or treatment of bowel inflammation in an animal, including a human. This aspect can be alternatively formulated as the use of a composition as defined in the first aspect of the invention for the manufacture of a medicament for the prevention and/or treatment of bowel inflammation. This may be alternatively formulated as a method for the prevention and/or treatment of bowel inflammation in an animal, including a human, comprising administering to said animal in need thereof an effective amount of the composition as defined in the first aspect of the invention. In one embodiment of the third aspect of the invention, the composition is used for the treatment or prevention of Inflammatory Bowel Disease.
From the data obtained using the IBD models reported in the examples (see below), it is derived that the administration of the strains of the invention, which are effective in the treatment of conditions characterized by bowel inflammation and diarrhoea, can also be useful to treat other conditions characterised by inflammation of the bowel mucosa or submucosa and where diarrhoea is prevalent, such as enteritis caused by radiotherapy or
chemotherapy. Enteritis is a common side effect of abdominal and pelvic radiotherapy, affecting 60-70% of patients. Enteritis can force schedule changes in the radiotherapy regime to decrease side-effects, potentially leading to sub-optimal anti-tumoral efficacy of the treatment. There are currently no preventive strategies for radiotherapy-induced enteritis. However, some probiotics have shown to be promising in randomized clinical trials (RCTs). A probiotic composition, such as the one of the present invention, combining the health-promoting effects of SCFAs production, the ability to withstand reactive oxygen and nitrogen species found in the inflamed mucosa, and the antimicrobial activity against opportunistic pathogens can be useful to treat radiotherapy and chemotherapy-induced enteritis.
On the other hand, the present inventors have found that the strains of the present invention are efficient in the treatment of IBS. As it is shown below, the composition of the first aspect of the invention is useful in treating IBS, as assessed by a randomized double-blind placebo-controlled intervention trial.
Therefore, in a fourth aspect the present invention provides the composition of the first aspect of the invention for use in the prevention and/or treatment of IBS. This aspect can be alternatively formulated as the use of a composition as defined in the first aspect of the invention for the manufacture of a medicament for the prevention and/or treatment of IBS. This may be alternatively formulated as a method for the prevention and/or treatment of IBS in an animal, including a human, comprising administering to said animal in need thereof an effective amount of the composition as defined in the first aspect of the invention.
Furthermore, the present inventors have found that due to the features of the strains, the composition of the first aspect of the invention is useful in the treatment of abdominal bloating and distension. As it is shown below, when the composition of the invention is administered to people suffering from abdominal bloating and distension, it is observed a surprising improvement. Therefore, in a fifth aspect the present invention provides the composition of the first aspect of the invention for use in the treatment of abdominal bloating and distension. This aspect can be alternatively formulated as the use of a composition as defined in the first aspect of the invention for the manufacture of a medicament for the treatment of abdominal bloating and distension. This may be alternatively formulated as a method for the treatment of abdominal bloating and distension in an animal, including a human, comprising administering to said animal in need thereof an effective amount of the composition as defined in the first aspect of the invention. The surprising beneficial effects observed in people suffering from IBS, and/or abdominal bloating and distension may be due to the fact that the strains of the invention CECT 7483, CECT 7484 and CECT 7485 have the ability of producing the SCFAs listed in Table 6 and the antagonistic activity shown in Table 3.
It is well-known in the state of the art that SCFAs modulate gut motility.
Particularly, SCFAs are known to stimulate serotonin (5-HT) release in rat colon (Fukumoto S. et al., 2003; Tazoe H. et al., 2008) which plays a pivotal role in the regulation of both gut motility and sensation. Similarly, butyric acid has been described to decrease visceral sensitivity of the intestine in human volunteers (Vanhoutvin S. A. et al., 2009). From this, it can be concluded that the strains forming the composition of the invention can be useful to treat, not only IBS or abdominal pain, but also other conditions related to
gastrointestinal motility and/or gastrointestinal pain, such as functional constipation or functional diarrhoea. The composition and isolated strains of the present invention are not obviously derived from the prior art because they are the result of a complex investigation and the results which have been obtained regarding the efficiency in the treatment of IBS and abdominal bloating and distension are surprising.
Surprisingly, the present inventors have found, for the first time, a
Pediococcus acidilactici strain with the ability of treating IBD and IBS. Said ability, without being bound the theory, is believed to be due to the specific properties, pointed out throughout the specification, of the isolated
Pediococcus strain. In the light of the teachings and protocols provided in the present specification, the skilled person in the art will be able to find further P_. acidilactici strains with the same probiotic and therapeutic features than the one object of the present application. The composition according to the invention that comprises an effective amount of the strains of the invention, or of their mutants, can be formulated as edible, pharmaceutical or veterinary products, in which said strains are the only active agents or are mixed with one or more other active agents and/or are mixed with pharmaceutically or veterinary acceptable excipients (in the case of a pharmaceutical or veterinary product) or adequate additives (in the case of an edible product). In a particular embodiment of the invention, the products additionally contain one or more further active agents. Preferably, the additional active agent or agents are other probiotic bacteria which are not antagonic to the strains forming the composition of the invention. Depending on the formulation, the strains may be added as purified bacteria, as a bacterial culture, as part of a bacterial culture, as a bacterial culture which has been post-treated, and alone or together with suitable carriers or ingredients. Prebiotics could be also added. In other aspects the invention provides a pharmaceutical and veterinary products that contain an effective amount of the composition of the invention together with adequate amounts of pharmaceutically or veterinary acceptable excipients. In this regard, the pharmaceutical product may be prepared in any suitable form which does not negatively affect to the bioavailability of the strains forming the composition of the invention. Thus, the composition of the invention can be formulated to be administered orally in the form, for instance, of freeze-dried power, capsules, liquid preparations, etc. Selection of the excipients and the most appropriate methods for formulation in view of the particular purpose of the composition is within the scope of ordinary persons skilled in the art of pharmaceutical technology. Although oral administration is preferred, other forms are possible, such as injectable, rectal or topical.
The term "pharmaceutically acceptable" as used herein pertains to
compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of a subject (e.g. human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. Each carrier, excipient, etc. must also be "acceptable" in the sense of being compatible with the other ingredients of the formulation.
Suitable carriers, excipients, etc. can be found in standard pharmaceutical texts. Likewise, the term "veterinary acceptable" means suitable for use in contact with the tissues of a non-human animal.
The strains of the invention can be also included in a variety of edible products, such as a milk products, a yogurt, a curd, a cheese (e.g. quark, cream, processed, soft and hard), a fermented milk, a milk powder, a milk based fermented product, an ice-cream, a fermented cereal based product, a milk based powder, a beverage, a dressing, and a pet food. The term "edible product" is used herein in its broadest meaning, including any type of product, in any form of presentation, which can be ingested by an animal, but excluding pharmaceutical and veterinary products. Examples of other edible products are meat products (e.g. liver paste, frankfurter and salami sausages or meat spreads), chocolate spreads, fillings (e.g. truffle, cream) and frostings, chocolate, confectionery (e.g. caramel, fondants or toffee), baked goods (cakes, pastries), sauces and soups, fruit juices and coffee whiteners.
Particularly interesting edible products are dietary supplements and infant formulas. In the sense of the present invention, dietary supplements also include nutraceuticals, which are known to be extracts of foods that have a medicinal effect on human health. Fodders for animal food are also included in the scope of the invention. The compositions of the invention could be also used as an ingredient in other food products.
Accordingly, in another aspect of the invention, an edible product is provided which contains the composition of the invention together with appropriate amounts of edible ingredients. Preferably, the composition of the invention is a dietary supplement.
The effective amount of colony forming units (cfu) for each strain in the composition will be determined by the skilled in the art and will depend upon the final formulation. For instance, in edible products, the strain or strains are present in an amount from about 105 cfu/g to about 1012 cfu/g, preferably in an amount from about 107 cfu/g to about 1012 cfu/g, according to the current legislation. The term "colony forming unit" ("cfu") is defined as number of bacterial cells as revealed by microbiological counts on agar plates.
Dietary supplements usually contain probiotic strains in an amount ranging from 107 and 1012 cfu/g. In a particular embodiment, the composition of the invention is a dietary supplement comprising between 109-1011 cfu/g.
The strains of the invention are produced by cultivating the bacteria in a suitable medium and under suitable conditions. The strains can be cultivated alone to form a pure culture, or as a mixed culture together with other microorganisms, or by cultivating bacteria of different types separately and then combining them in the desired proportions. After cultivation, the cell suspension is recovered and used as such or treated in the desired manner, for instance, by concentrating or freeze-drying, to be further employed in the preparation of pharmaceutical or edible products. Sometimes the probiotic preparation is subjected to an immobilisation or encapsulation process in order to improve the shelf life. Several techniques for immobilisation or encapsulation of bacteria are known in the art.
If the composition according to the invention is used as a dietary supplement, it can be administered as such, can be mixed with a suitable drinkable liquid, such as water, yoghurt, milk or fruit juice, or can be mixed with solid or liquid food. In this context the dietary supplement can be in the form of tablets, pills, capsules, granules, powders, suspensions, sachets, pastilles, sweets, bars, syrups and corresponding administration forms, usually in the form of a unit dose. Preferably, the composition of the invention is administered in the form of tablets, capsules or powders, manufactured in conventional processes of preparing pharmaceutical products.
Throughout the description and claims the word "comprise" and its variations are not intended to exclude other technical features, additives, components, or steps. Additional objects, advantages and features of the invention will become apparent to those skilled in the art upon examination of the
description or may be learned by practice of the invention. Furthermore, the present invention covers all possible combinations of particular and preferred embodiments described herein.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 represents the pulsed field gel electrophoresis patterns of Not-I or Sfi-I (left) and Sma-I (right) restricted genomic DNA of: 1 , Pediococcus acidilactici CECT 7483; 2, Lactobacillus plantarum CECT 7484; 3, Lactobacillus plantarum CECT 7485. As controls: 4, commercial strain P. acidilactici
RosselM 001 (Institut Rossell, Canada); 5, L. plantarum 299v (Probi AB,
Sweden); and 6, L plantarum strain isolated from commercial product VSL#3 (VSL Pharmaceuticals, USA). This Figure refers to the "strain genotyping" section. FIG. 2 represents the Disease Activity Index (Y-axis) in a group of mice suffering from a DSS-induced intestinal inflammation. X-axis represents the administration of: a, probiotic formulation of the invention in a group of mice suffering from a DSS-induced intestinal inflammation; b, a commercial probiotic formulation (VSL#3) in a group of mice suffering from a DSS-induced intestinal inflammation; c, vehicle in a group of mice suffering from a DSS- induced intestinal inflammation; and d, vehicle in a healthy control group. This figure refers to the "In Vivo Effect on Chemically-induced Gut Inflammation" section. FIG. 3 represents the levels of IL-6 (Y-axis) in a group of mice suffering from a DSS-induced intestinal inflammation. X-axis represents: a, probiotic
formulation of the invention administered to a group of mice suffering from a DSS-induced intestinal inflannnnation; b, a commercial probiotic formulation (VSL#3) administered to a group of mice suffering from a DSS-induced intestinal inflammation; c, vehicle administered to a group of mice suffering from a DSS-induced intestinal inflammation; and d, vehicle in a healthy control group. This figure refers to the "In Vivo Effect on Chemically-induced Gut Inflammation" section.
FIG. 4 represents the number of Symptom-free weeks (i.e. number of weeks before the onset of the first symptom, thus being the Disease Activity Index equal to zero) (Y-axis) in a IL-10 knock-out mouse model. X-axis represents the administration of: a, the probiotic composition of the invention to a IL-10 knock-out mice group; b, the commercial probiotic formulation VSL#3 to a IL- 10 knock-out mice group; c, PBS to a IL-10 knock-out mice group; and d, vehicle in a healthy control group. This figure refers to the "In Vivo Effect on Spontaneous Gut Inflammation" section.
FIG. 5 represents the levels of IFN-γ (Y-axis) in a IL-10 knock-out mouse model. X-axis represents: a, probiotic formulation of the invention to a IL-10 knock-out mice group; b, a commercial probiotic formulation (VSL#3) to a IL- 10 knock-out mice group; c, vehicle to a IL-10 knock-out mice group; and d, vehicle to a healthy control group. This figure refers to the "In Vivo Effect on Chemically-induced Gut Inflammation" section.
FIG. 6 represents the levels of IL-6 (Y-axis) in a IL-10 knock-out mouse model. X-axis represents: a, probiotic formulation of the invention to a IL-10 knock-out mice group; b, a commercial probiotic formulation (VSL#3) , to a IL- 10 knock-out mice group; c, vehicle administered to a IL-10 knock-out mice group; and d, vehicle to a healthy control group. This figure refers to the "In Vivo Effect on Chemically-induced Gut Inflammation" section.
FIG. 7 represents the variation of the IBSQoL score compared to baseline (Y- axis) of volunteers treated with capsules including the composition (black bars) or placebo (white bars). X-axis represents the variation of the score 21 days and after 42 days of treatment. This figure refers to the "Improvement of Health-Related Quality of Life" in the "In Vivo Efficacy on IBS Subjects" section. FIG. 8 represents the variation of the VSI score compared to baseline (Y-axis) of volunteers treated with capsules including the composition (black bars) or placebo (white bars). X-axis represents the variation of the score after three weeks and after six weeks of treatment. This figure refers to the "Improvement of the Visceral Sensitivity" in the "In Vivo Efficacy on IBS Subjects" section.
EXAMPLES
The following sections describe the characterization of the strains of the invention, their specific probiotic features and their physiological effects on the gastrointestinal and immune systems. As used hereinafter, strain F1033 corresponds to Pediococcus acidilactici CETC 7483, strain F2064 to
Lactobacillus plantarum CECT 7484, and strain F2076 to Lactobacillus plantarum CECT 7485.
1 . Isolation of microorganisms
A) Methods For isolation of microorganisms, fresh stools and saliva (Daniel C. et al., 2006) were collected from 0-5 year-old children and dissolved in PBS buffer (pH 7.4), aliquoted and plated on MRS supplemented with various antibiotic combinations. Strains were cultured under microaerophilic conditions (5% CO2) at 37 °C. Incubation time depended on the growth rate, but run normally from 24 h to 3 days. Gram staining was carried out in order to get a first identification. Once grown, isolated strains were stored by lyophilisation in PBS 0.1X with 15% skim milk powder.
B) Results
Novel strains F2064, F2076 and F1033 were grown on MRS agar
supplemented with 10 g/ml vancomycin. Microscopic examination revealed that strains F2064 and F2076 are Gram-positive bacilli, while strain F1033 is a Gram-positive with coccal morphology. 2. Identification
A) Methods Genomic DNA was extracted using Wizard genomic DNA purification kit (Promega). For each isolated strain, the 16S gene was amplified by PCR, using the universal primers 27f, 357f, 907r and 1492r (Weisburg W. G. et al., 1991 ), which generate a nearly full-length 16S rRNA fragment (1465 bp). DNA was washed using Quiaquick kit (Quiagene, GmbH, Hilden, Germany) and four sequencing reactions were performed per sample, using BigDye v.3.1 kit, on a Genetic Analyzer 3130 (Applied Biosystems). Selected sequencing primers DNA Sequence Analysis v.5.2 (Applied Biosystems) software was used to collect data and to build chromatograms, which were analyzed through Chromas (Technelysium Pty Ltd.) and BioEdit (Ibis Biosciencies) software. Genus and species identification was performed by comparison of the obtained sequence with 16S sequences of known organisms from both RefSeq data base (http://www.ncbi.nlm.nih.gov/RefSeq/) by means of a BLASTN search (Altschul S. F. et al., 1990), and from the Ribosomal
Database Project (Wang Q. et al., 2007).
TABLE 1 : Primers used for amplifying and sequencing the 16S gene.
Step Primer Orientation 5' -> 3' Sequence
27f forward AGAGTTTGATCCTGGCTCAG (SEQ ID NO:
Amplification 1)
1492r reverse GGTTACCTTGTTACGACTT (SEQ ID NO: 2)
27f forward AGAGTTTGATCCTGGCTCAG (SEQ ID NO:
1)
357f forward CGCCCGCCGCGCCCCGCGCCCGGCCCGC
CGCCCCCGCCCCCCTACGGGAGGCAGCA
Sequencing
G (SEQ ID NO: 3)
907r reverse CCGTCAATTCCTTTGAGTTT (SEQ ID NO:
4)
1492r reverse GGTTACCTTGTTACGACTT (SEQ ID NO: 2) B) Results
Strains F2064 and F2076 were identified as members of the Lactobacillus plantarum group. Strain F1033 was identified as Pediococcus acidilactici.
3. Survival to Gl Tract
A) Methods To assess tolerance to acidic environment, 20-μΙ aliquots of each bacterial strain culture were placed in 96-well plates, together with 200-μΙ aliquots of MRS medium adjusted with HCI to pH 2 and 3 (Panreac). Plates were kept at 37 °C for 1 h and optical density at 620 nm was measured. Finally, viable cells were determined by plate counting and compared to the number of viable cells in the inoculum.
To assess tolerance to bile salts, 20-μΙ aliquots of each bacterial strain culture were placed in a 96-well plate together with 200 μΙ of MRS medium
supplemented with 0.5% Oxgall (Sigma). Plates were incubated at 37 °C and 5% CO2 for 3 hours, and then optical density was measured. Finally, viable cells were determined by plate counting and compared to the number of viable cells in the inoculum.
B) Results
All three strains displayed a good ability to survive acidic environments, with less than one log reduction in the number of viable cells after 1 h incubation in MRS at pH= 2 or pH= 3. Strains also posses a marked resistance to bile salts, with less than a 50% reduction in the number of viable cells after 3 h incubation in MRS supplemented with 0.5% of bile salts.
4. Adherence A) Methods
Porcine intestine was washed with PBS pH 7.4, containing 0.01 % gelatin and a cocktail of protease inhibitors (Complete®, Sigma). The mucosa was scrapped and dissolved in HEPES-Hank's buffer (10 mM HEPES, pH 7.4) (Collado M. et al., 2007) containing the aforementioned inhibitors. Then, mucus was centrifuged at 13000 rpm for 10 min using the same buffer.
Supernatants were recovered and protein content was determined by
Bradford protocol. 24 h before the assay, 1 ml of mucus solution 0.5 mg/ml was incubated in wells of a 24-well ELISA plate.
Each strain to be tested was grown overnight in MRS medium supplemented with tritium-labeled thymidine (5 μΙ in 3 ml of MRS). Cultures were centrifuged and adjusted to 108 cfu/ml in PBS by counting on a Neubauer chamber and samples of each culture were taken to determine the amount of tritium-labeled thymidine incorporated by means of a scintillation reader. Then, 0.5 ml were added to the mucus-containing wells of the 24-well plate and incubated at 37 °C for 60 min. Supernatant of each well was removed, and wells were washed twice with MEM Alpha medium (Gibco) to remove loosely adherent bacteria. Finally, wells were scrapped to retrieve the mucus together with the adhering bacteria, and radioactivity was measured. Specific activity (cpm/CFU) of each culture was calculated from the total radioactivity incorporated in the PBS suspension adjusted to 108 cfu/ml. Lactobacillus rhamnosus GG (Valio Ltd, Finland) was used as a positive control, because of its remarkable high adherence to the intestinal epithelium (Jacobsen C. N. et al., 1999).
Caco-2 cells were obtained from ATCC (ECACC N°: 86010202). Cells were seeded in 24-well plates and allowed to grow in DMEM until confluence (37 °C, 5% CO2). The experimental procedure to obtain the number of bacteria that adhere per unit of caco-2 cells area is essentially the same as the one explained above for adhesion to mucus.
B) Results
Adhesion capacity of strains F1033, F2064 and F2076 was measured from scintillation of tritium-labeled thymidine and compared to those of the commercial strain L. rhamnosus GG. Adhesion to epithelial cells using the Caco-2 model is a common assay for probiotic strains. Compared to L rhamnosus GG, strains F2064 and F2076 show an affinity for epithelial cells 60% lower. However, considering the high affinity of L. rhamnosus GG for epithelial cells, these values are comparable to other well known probiotics such as L. plantarum 299v, and superior to many other probiotic strains (Jacobsen C. N. et al., 1999). On the other hand, adhesion of strain F1033 to epithelial cells is 2.5 times higher than L. rhamnosus GG. Besides, strains F2064 and F2076 displayed a much higher affinity for intestinal mucus than for epithelial cells, while strain F1033 showed the opposite behavior. Results are shown in the following table.
TABLE 2: Mucus adhesion of probiotic bacterial strains. [* From a total bacteria concentration of 108 cfu].
Figure imgf000021_0001
5. Antagonism Capacity
A) Methods
The following indicator strains were used: P. mirabilis CECT 4557, K. oxytoca CIP 103434, C. perfrinqens ATCC 13124. C. ramosum ATCC 25582, E. faecal is CETC 795, Y. pseudotuberculosis ATCC29833, B. vulqatus ATCC 8482 and B. thetaiotaomicron ATCC2079 were collection strains. C. albicans. S. enterica thyphimurium, S. enterica cholerasuis, C. jejuni. E. coli and P_. aeruginosa were lab isolates. Indicator strains were swabbed uniformly in plates containing the appropriate medium (Oxoid) and grown to confluence at the appropriate temperatures in microaerophilic conditions (5% CO2). Then, 6 mm (diameter) cylinder sections of confluent F1033, F2064 or F2076 agar plates were placed upside-down on the indicator strain plate and incubated overnight at 37 °C. The next day, inhibition zones were measured by placing the agar plate over a flat rule. Growth inhibitory activity (Gl) was calculated as follows:
(12 D - Clf) where IZD is the Inhibition Zone Diameter and CD is the cylinder diameter, measured in millimeters.
B) Results
TABLE 3: Growth inhibitory activity (Gl) of probiotic strains against 12 pathogenic or potentially pathogenic strains, and against 2 common commensal strains of the gastrointestinal flora.
Figure imgf000022_0001
Strains F2064, F2076 and F1033 displayed significant inhibitory activity against Candida albicans and several potentially pathogenic bacteria. On the other hand, the strains displayed minimal activity against commensal strains commonly found in the indigenous gastrointestinal flora of the Bacteroides genus. Also, strains F2064, F2076 and F1033 did not display significant inhibitory activity among them. It is noteworthy that strain F1033 is the only strain displaying high inhibitory activity against Campylobacter jejuni, while strain F2076 outstands in inhibiting Escherichia coli and strain F2064 in inhibiting both Candida albicans and Proteus mirabilis. 6. Antioxidant Capacity
A) Methods 20 μΙ aliquots of overnight cultures of each strain (109 cfu/ml aprox) were placed in a 96-well plate. 200 μΙ of MRS supplemented with 10 mM of paraquat (C12HMCI2N2, a superoxide anion donor) or 10 mM of sodium nitroprusside (Na2[Fe(CN)5NO], a nitric oxide donor) were added to wells and plates incubated at 37 °C and 5%CO2. Optical densities at 620 nm were read after 6 h. Results are expressed as percent of growth compared to growth in standard MRS medium. The same protocol was followed with the L
rhamnosus GG strain and the L. plantarum strain isolated from the
commercial formulation VSL#3 (the isolation was performed using standard procedures).
B) Results
Oxidative stress is defined as an imbalance between generation of reactive oxygen species (ROS) and decreased antioxidant defence systems. Oxidative stress develops particularly in inflammatory reactions because the
inflammatory cells, neutrophils, and macrophages produce large amounts of ROS (Rezaie A. et al., 2007; Roessner A. et al., 2008). Strains F1033, F2064 and F2076 showed a capacity to survive under strong oxidizing conditions comparable to the well-known probiotic strain L. rhamnosus GG, as well as to the L. plantarum strain isolated from the VSL#3 formula. It is worth noting that strain F2076 displayed the highest resistance both to paraquat (superoxide anion donor) and sodium nitroprusside (nitric oxide donor). Resistance to oxidative stress is a desirable trait for probiotic strains that are expected to survive in the environment of an inflamed mucosa. TABLE 4: Percent of growth in medium containing 10 mM of paraquat or sodium nitroprusside, compared to standard MRS medium.
Figure imgf000024_0001
7. Strain Genotypinq
A) Methods Strains F1033, F2064 and F2076 were subjected to a previously described protocol (Rodas A. M. et al., 2005) with minor modifications. Strains were grown on MRS agar plates and incubated at 37 °C 5% CO2 for 18 h. Cells were harvested and washed 3 times in 8 ml PET (10 mM Tris pH 7.6, 1 M NaCI) then centrifuged at 6000 rpm 10 min. Pellets were resuspended in 700 ml lysis buffer (6 mM Tris, 1 M NaCI, 0.1 M EDTA, 0.5% SLS, 0.2 %
deoxycholic acid; 1 mg/ml lysozyme; 40 U/ml mutanolysin; 20 mg/ml RNase). An equal volume of 1 .6% low melting point agarose (FMC BioProducts, Rockland, ME, USA) was added to the resuspended cells and solidification was allowed at 4 °C for 1 h. Inserts were transferred to 2 ml lysis buffer II (0.5 M EDTA pH 9.2, 1 % N-lauryl sarcosine and 1 mg/ml pronase) and incubated at 50 °C for 48 h. Then inserts were washed at room temperature with TE buffer (10 mM Tris, 1 mM EDTA pH 8.0). Total DNA digestion was performed separately by Sfi-I and Sma-I restriction enzymes (Roche Diagnostics). Pulse-field electrophoresis was carried out using CHEF DRIII apparatus
(BioRad Laboratories). Inserts were loaded in a 1 % agarose gel (SeaKem ME agarose, FMC BioProducts, ME, USA). DNA MW markers were Lambda ladder PFG Marker and Low Range PFG Marker (New England Biolabs). After electrophoresis, gels were stained with ethidium bromide and UV using GelDoc System (BioRad). B) Results
FIG. 1 shows the pulse-field electrophoresis profiles obtained. Strain F1033 shows a similar genomic restriction profile to P. acidilactici R1001 after digestion with Sma-I. However, the genomic profile obtained after digestion with enzyme Not-I is clearly different. On the other hand, the genomic restriction profiles of strains F2064 and F2076 are clearly different among them and also compared both to L. plantarum 299v and to the L. plantarum strain contained in the VSL#3 formula.
8. Production of Short Chain Fatty Acids A) Methods Strains were incubated overnight in a basal medium (see TABLE 5)
supplemented with different fibers, each one (inulin, pectin and FOS) in a specific amount, under microaerophilic conditions (5% CO2) at 37 °C. Next, cells were removed by centrifugation at 12.000 rpm for 10 min and
supernatants were filtered and frozen in liquid nitrogen and kept at -80 °C until analyzed by gas chromatography, focusing on the amount of acetic, propionic and butyric acids.
TABLE 5
Figure imgf000025_0001
B) Results
Short chain fatty acids (SCFAs) are the end products of anaerobic bacteria break down of carbohydrates in the large bowel. SCFAs, mainly acetate, propionate and butyrate account for approximately 80% of the colonic anion concentration and are produced in nearly constant molar ratio 62:22:15.
Among their various properties, SCFAs, especially butyric acid, but also acetic and propionic acid, are readily absorbed by intestinal mucosa, are relatively high in caloric content, are metabolized by colonocytes and hepatocytes, stimulate sodium and water absorption in the colon and are trophic to the intestinal mucosa (D'Argenio G. et al., 1999). On the other hand, high amounts of acetic acid have long been known to be irritant to the intestinal mucosa (Yamada Y. et al., 1992). Strains F1033, F2064 and F2076 are strong producers of either acetic, propionic or butyric acid.
TABLE 6: Acetic, propionic and butyric acid production by strains grown on basal medium enriched with inulin, pectin and FOS.
Figure imgf000026_0001
(n.d.= non-detected)
9. Compatibility with IBP treatments A) Method Supplemented broth was prepared by dissolving 5-aminosalycilic acid
(Pentasa®, Ferring Pharmaceuticals) at the maximal soluble concentration (0.84 gr/L) and half this concentration (0.42 gr/L) in MRS liquid broth. The strains of the invention were grown in standard MRS broth or 5-aminosalicylic acid-supplemented broth for 4h at 37°C in microaerophilic conditions (5% CO2), and growth was assessed by measuring optical density at 620 nm. Results are expressed as percent of growth in standard MRS medium. B) Results
Prolonged treatment of mild to moderate IBD symptoms is usually carried out using oral aminosalycilates (5-ASA derivatives) (Katz J. A., 2007). Therefore it is of interest to evaluate if the probiotic strains of the invention can be coadministered with 5-ASA derivatives. Considering that growth of none of said strains is completely inhibited despite the high stringency of the conditions, we can conclude that co-administration of mesalazine is not likely to compromise the efficacy of the probiotic, even using saturated concentrations of
mesalazine (0.84 g/L) as shown in TABLE 7:
TABLE 7
Figure imgf000027_0001
10. In Vivo Effect on Chemically-induced Gut Inflammation
A) Methods
The therapeutic effect of the composition of the invention on mild gut inflammation was investigated with a 5-day repetitive oral administration of dextran sodium sulfate (DSS) in the mouse (Okayasu I. et al., 1990). When used in a low dose (2.5-3%) for a short time (5 days), DSS produces mild colitis, with intestinal inflammation at the histological level but without significant macroscopic changes (e.g. colon shortening, mesenteric
adherences).
External symptoms include weight loss and diarrhea, with rare occurrence of blood in feces. Therefore this model is representative of low-grade ulcerative colitis. Strains F1033, F2064 and F2076 were lyophilised in sterile water with 15% skim milk and 4% sucrose as cryoprotectants and mixed in equal amounts (ratio in concentration 1 :1 :1 ). Eight-week-old Balb/c mice (Charles River, Barcelona, Spain), weighing 20-25 g, were kept under specific pathogen-free (SPF) conditions in an isolator (Harlan Iberica, Barcelona, Spain) at constant temperature (22 °C) in a 12- hour of light/dark cycle. Two mice acted as littermates. Mice had free access to sterilized diets (laboratory's standard diet; Harlan Iberica, Barcelona, Spain) and to drinking fluid. Mice were kept for 7 days in the facility before the beginning of the experiments (quarantine). Mice were allocated to one of four groups: a) probiotic composition of the invention + DSS (n=8); b) VSL#3 (VSL Pharmaceuticals, USA) + DSS (n=8); c) vehicle + DSS (n=8); and d) vehicle + healthy controls (n=6).
Probiotics (or vehicle) were administered by oral gavage for ten days before (day -10) starting DSS administration (day 0). Each mouse received daily 2.5 x 108 cfus of probiotic in 0.1 ml_ of sterilized water (vehicle) by gavage. Non- probiotic treated mice received the same volume of vehicle (distilled water with 15% skim milk and 4% sucrose).
Mice were fed with 3% (w/v) DSS (mol. Wt 40kD, Applichem Lifescience, VWR, Barcelona) in their drinking water for 5 days (days 0 to 4, followed by three days without DSS) according to a previously described method with minor modifications (Okayasu I, et al. Gastroenterol 1990). Healthy controls never received DSS.
Clinical signs were daily monitored. Disease Activity Index was calculated according to the following formula and interpretation table:
DAI = SCOreweight Loss + SCOrestool Blood + SCOrestool Consistency
Results are shown in TABLE 8: TABLE 8
Figure imgf000029_0001
The Disease Activity Index score used hereby was first described by Cooper et al. and combines several clinical symptoms into one normalized score (Cooper H. S. et al., 1993). Maximum score is 12 points. This score has been widely used to evaluate the efficacy of experimental treatments - probiotics among them - in animal models of IBD (Fitzpatrick L. R. et al., 2007; Grabig A. et al., 2006; Sasaki M. et al., 2005). After being sacrificed by anesthetic overdose of inhaled Halothane
(Fluotane®, Zeneca Ltd, UK), colon samples of the animals were harvested and washed in cold PBS. Colon weight/length ratio was recorded. Samples for cytokine measurements were frozen in liquid nitrogen and homogenized in 1 mL of cold PBS with inhibitor protein cocktail (Sigma-Aldrich Chem., Spain) and centrifuged (15000 x g, 10 min). IL-6, IL-10, IL23p19, IFN-γ and TNF-cc concentrations were measured in colonic supernatants using Cytokine 6-Plex Assay (Procarta™ Cytokine Profiling Kit, PANOMICS, Spain) for the
Luminex® Platform (Luminex® Co, Austin, USA). Fluorescent microparticle beads, pre-spotted with cytokine-specific antibodies, were incubated with 50 μί 1 :5 diluted supernatant. Specific-biotinylated secondary antibodies and streptavidin-phycoerythrin (S-PE) were sequentially added. Data were expressed as pg of cytokine per mg of protein (Quick Start Bradford Protein Assay, BIO-RAD, CA, USA). All measurements were done in duplicate. B) Results
Disease Activity Index As shown in FIG. 2, the group receiving the probiotic formula of the invention displayed a significant improvement of the clinical symptoms when compared to DSS-treated controls, as assessed by the Disease Activity Index (p < 0.05, two-tail ANOVA with Tukey-Kramer post-hoc test). Healthy controls also displayed a lower Disease Activity Index (p < 0.05).
Cytokine Levels
Analysis of various cytokines in the intestinal mucosa revealed that probiotic formula of the invention significantly decreased IL-6 when compared to DSS- treated controls (p < 0.01 , two-tail ANOVA with Tukey-Kramer post-hoc test), while the effect of commercial probiotic formula VSL#3 failed to achieve significance (p > 0.05). IL-6 is a marker of acute inflammation (FIG. 3). As expected, levels of IL-6 in healthy controls were also significantly lower than DSS-treated controls (p < 0.05). A statistically significant correlation was found between clinical symptoms (DAI score) and IL-6 levels in the intestinal mucosa (p < 0.05, Spearman ranks test) (data not shown). On the other hand, correlation between clinical symptoms and IL-10, IL-23, TNFa or IFNy was not statistically significant, and the probiotic formula of the invention did not significantly affect the levels of these cytokines.
1 1 . In Vivo Effect on Spontaneous Gut Inflammation A) Methods
The therapeutic effect of probiotic formula of the invention was also
investigated in the IL-10 knock-out mouse model. This model spontaneously develops bowel inflammation at 8 to12 weeks of age, with a penetrance of 80- 90% (Scheinin T. et al., 2003). Interleukin 10 (IL-10) is an important regulatory cytokine that supresses effector functions of macrophage/monocytes, T helper 1 (Th1 ) cells, and natural killer cells. In addition, IL-10 augments proliferation and differentiation of B cells. Murine models lacking the IL-10 gene
spontaneously develop inflammatory bowel disease and gastrointestinal tumors. The gastrointestinal flora has been implicated in the pathogenesis of these disease states as germ free animals do not develop disease. The IL-10 knock-out mouse has been widely used to evaluate new therapeutic options for IBD.
Six-week-old C57B6J IL-10-deficient or wild type mice (Charles River,
Barcelona, Spain) were kept under specific pathogen-free (SPF) conditions in an isolator (Harlan Iberica, Barcelona, Spain) at constant temperature (22°C) in a 12-hour of light/dark cycle. Mice had free access to sterilized diets (diet based in AIN-93 for maintenance of mice was composed by 12% of water, 14,5% of protein, 4% of fat, 4,5% of fibre and 4,7% of ash; Harlan Interfauna Iberica S.A., Barcelona, Spain) and to drinking fluid.
Mice were allocated to one of three groups: a) probiotic formula 1.3.1 (n=12 IL- 10-/-; n=5 wild type); b) VSL#3 (n=12 IL-10-/-; n=5 wild type); and c) vehicle (n=12 IL-10-/-; n=5 wild type). Each mouse in groups "a" and "b" received daily 109 CFU of probiotic in sterilized drinking water (vehicle). Non-probiotic treated mice (Placebo group) received vehicle alone. Probiotics (or vehicle) were administered during ten weeks. Clinical signs were daily monitored. Disease Activity Index (Cooper H. S. et al., 1993) was calculated as in the model of DSS-induced gut inflammation (see above).
Sixteen-weeks-old mice were sacrificed by anaesthetic overdose of inhaled Halothane (Fluotane®, Zeneca Ltd, UK). Colon samples of the animals were harvested and washed in cold PBS. Blood samples were also collected by cardiac puncture to analyze hematocrit and hemoglobin concentration (Coulter MaxM Analyzer with autoloader, Izasa, Spain). Colon weight/length ratio was recorded. Then, colons were frozen in liquid nitrogen and cytokines IL-6, and IFNy were measured using the same protocol as in the model of DSS-induced gut inflammation (see above). B) Results
Disease Activity Index As shown in FIG. 4, a significant delay on the onset of the clinical symptoms was observed both in the group treated with the composition of the invention and the VSL#3 commercial formula when compared to vehicle-treated controls (p < 0.01 , two-tail ANOVA with Tukey-Kramer post-hoc test).
Additionally, treated groups tended to display lower Disease Activity Index scores, although the difference did not reach significance (data not shown).
Cytokine Levels
Analysis of various cytokines revealed that probiotic composition of the invention significantly decreased IFNy levels in knockout mice when compared both to vehicle-treated knock-outs (p < 0.01 , two-tail nonparametric ANOVA with Dunn post-hoc test) and to commercial formula VLS#3 (p< 0.05). In fact, as it is shown in FIG. 5, the levels of IFNy attained the same levels as those of wild-type healthy controls. Additionally, as it is derived from FIG. 6, there was also a clear tendency of probiotic formulas to reduce the levels of IL-6, although results did not reach significance due to the large standard deviation among vehicle knockout mice.
A significant correlation was found between the severity of clinical symptoms (Disease Activity Index) and the levels of IFNy at the end of the study in colonic mucosa measured after the sacrifice (p < 0.05, Spearman rank's test) (data not shown).
Safety of the Probiotic Formula
Clinical signs (weight loss, altered behavior, fur aspect, diarrhoea and stool blood) were daily monitored in wild-type mice receiving daily doses of the probiotic formula of the invention, the VSL#3 formula or vehicle during 10 weeks. No morbidity signs were detected during the study. Upon sacrifice, animals were subjected to gross necropsy. Analysis of all major cavities and organs did not reveal any pathological alteration (data not shown). 12. In Vivo Efficacy on IBS Subjects A) Methods Study Design
A multicenter randomized, double-blind, placebo-controlled clinical trial to study the effect of the composition of the invention on IBS patients was conducted.
Hydroxymethyl propyl cellulose capsules were filled with: (1 ) 150 mg of maltodextrin, (2) 5 mg of magnesium stearate, (3) 5 mg of silicon dioxide and (4) 200 mg of a 1 :1 :1 mixture of the three strains of the invention (at a concentration 5-1010 cfus/capsule). In addition a placebo was made with the same list of excipients and amounts but without including the composition of the invention. Content of the capsules throughout the study ranged from 5-1010 to 1 -1010 cfus.
33 eligible adult patients of both sexes meeting Rome III criteria for irritable bowel syndrome (Longstreth G. F. et al., 2006) were enrolled and randomly allocated to one of the following treatments for 6 weeks: a) the capsule including the composition of the invention once daily (n = 18); and b) the placebo capsule once daily (n = 15). The study was conducted according to the Helsinki Declaration for Clinical Trials and approved by the appropriate Ethical Committee.
Efficacy Assessment
The primary endpoint of this study was the global effect on health-related quality of life (hereinafter also referred as "HRQOL"), as assessed using a specific questionnaire for IBS: the validated Spanish version of the IBSQOL questionnaire (Badia X. et al., 2000). Following the guidelines from the
Spanish Gastroenterology Association, scores were standardized to a 0-100 scale. The secondary endpoint was the assessment of anxiety related to gastrointestinal sensations and symptoms by means of the validated Visceral Sensitivity Index questionnaire (hereinafter also referred as "VSI") (Labus J. S. et al., 2004). Volunteers were asked to fill these questionnaires at baseline (day 1 ), on day 21 and on day 42. Data was assessed per intent to treat analysis. The results are shown in FIG.'s 7 and 8.
B) Results
Baseline characteristics
No significant differences were evident between the groups in terms of baseline characteristics, as can be seen in Table 9, indicating that subjects in both groups were comparable in terms of the variables assessed. Groups were also comparable in terms of baseline standard blood biochemical parameters, anthropometric parameters, age and sex.
TABLE 9: Baseline scores for the two treatment groups
Figure imgf000034_0001
Improvement of Health-related Quality of Life (FIG. 7) The composition of the invention significantly improved health-related quality of life compared to placebo when assessed both after 21 days and 42 days of treatment (p < 0.05, T-test). Therefore, it is demonstrated that the composition of the present invention significantly reduces morbility and improves the quality of life of IBS subjects well above the placebo effect. The positive effects of the composition include the food-related distress, anxiety, interference in daily activities and sleep disturbance domains of the HRQOL questionnaire. The improvements in these scales suggest a reduction in abdominal pain, discomfort and altered bowel habits. To our knowledge, this is the first time that it is shown a probiotic composition displaying a significant effect on the global health-related quality of life of IBS patients. Improvement in the Visceral Sensitivity Index (FIG. 8)
The composition of the present invention significantly reduced the
gastrointestinal symptom-specific visceral sensitivity of IBS subjects compared to placebo. The effect was close to significant after 21 days of treatment, and clearly significant after 42 days of treatment (p < 0.01 , T-test), further confirming the usefulness of the composition of the present invention in treating IBS. The most pronounced improvement was observed in abdominal discomfort and bloating-related items of the questionnaire. Particularly, TABLE 10 shows the numbers of subjects reporting a significant improvement related to bloating and distension (as defined by an increase of at least two points compared to baseline in the 6-point scale of the VSI questionnaire that measures bloating and distension-related anxiety) at the end of the treatment. The difference between the two groups is statistically significant (p < 0.05, Fisher's exact test).
TABLE 10. Effect on abdominal bloating and distension-related anxiety, according to the VSI questionnaire, after 42 days of treatment
Figure imgf000035_0001
From the results obtained, therefore it is concluded that the composition of the invention is effective in treating abdominal distension and bloating.
13. Effect on Abdominal Bloating and Reduced Bowel Movements A 25 years old woman was suffering from chronic abdominal bloating and altered intestinal motility, reporting sometimes as few as on bowel movement per week. Diagnostic revealed a hypotonic and hypokinetic stomach, without evidence of other structural alterations in the gastrointestinal tract. The patient undertook a treatment of one capsule per day (as those described in example 12). After one week of treatment the patient reported a significant reduction of abdominal bloating and distension and a normalization of bowel habits. Symptoms reappeared after stopping the treatment for a few days. After restarting of the treatment in the form of one capsule every two days, the patient reported again a noticeable and long-lasting positive effect both on bloating and bowel habits.
This example further supports the use of the composition of the invention to treat abdominal bloating and altered intestinal motility in subjects which are not classified as having Irritable Bowel Syndrome.
BIBLIOGRAPHIC REFERENCES
Altschul, S.F., et al. "Basic local alignment search tool", J. Mol. Biol., 1990, vol. 215, p. 403-410.
Anadon, A., et al. Opinion of the Scientific Panel on Additives and Products or Substances used in Animal Feed on the updating of the criteria used in the assessment of bacteria for resistance to antibiotics of human veterinary importance", The EFSA Journal, 2005, vol. 233, p. 1 -12. Andreoletti, O., et al. "The maintenance of the list of QPS microorganisms intentionally added to food or feed. Question no: EFSA-Q-2008-006", The EFSA Journal, 2008, vol. 923, p. 1 -48.
Araya, M., et al. (2002) Guidelines for the Evaluation of Probiotics in Food - Joint FAO/WHO Working Group. FAO/WHO, Ontario, Canada. Badia, X., et al. "Adaptacion al espahol del cuestionario IBSQoL para la medicion de la calidad de vida en pacientes con sindrome de intestino irritable.", Rev Esp Enferm Dig, 2000, vol. 92, p. 637-643.
Bories, G., et al. "Update on the criteria used in the assessment of bacterial resistance to antibiotics of human or veterinary importance", The EFSA
Journal, 2008, vol . 732, p. 1 -15. Collado, M., et al. "Probiotic Strains and Their Combination Inhibit In Vitro Adhesion of Pathogens to Pig Intestinal Mucosa", Current Microbiology, 2007, vol. 55, p. 260-265.
Cooper, H.S., et al. "Clinicopathologic study of dextran sulfate sodium experimental murine colitis", Lab Invest.. 1993, vol. 69, p. 238-249.
D'Argenio, G. and Mazzacca, G. "Short-chain fatty acid in the human colon. Relation to inflammatory bowel diseases and colon cancer", Adv Exp Med Biol, 1999, vol. 472, p. 149-158.
Daniel, C, et al. "Selecting Lactic Acid Bacteria for Their Safety and
Functionality by Use of a Mouse Colitis Model", Appl. Environ. Microbiol., 2006, vol. 72, p. 5799-5805.
Dean, B.B., et al. "Impairment in work productivity and health-related quality of life in patients with IBS", Am J Manag Care., 2005, vol. 1 1 , p. S17-26.
Fitzpatrick, L.R., et al. "Effects of the probiotic formulation VSL#3 on colitis in weanling rats", J Pediatr Gastroenterol Nutr., 2007, vol. 44, p. 561 -570.
Fukumoto, S., et al. "Short-chain fatty acids stimulate colonic transit via intraluminal 5-HT release in rats", Am J Physiol Regul Integr Comp Physiol, 2003, vol. 284, p. R1269-1276.
Grabig, A., et al. "Escherichia coli Strain Nissle 1917 Ameliorates
Experimental Colitis via Toll-Like Receptor 2- and Toll-Like Receptor 4- Dependent Pathways", Infect. Immun., 2006, vol. 74, p. 4075-4082.
Guarner, F. and Schaafsma, G.J. "Probiotics", Int J Food Microbiol. , 1998, vol. 39, p. 237-238.
Jacobsen, C.N., et al. "Screening of Probiotic Activities of Forty-Seven Strains of Lactobacillus spp. by In Vitro Techniques and Evaluation of the
Colonization Ability of Five Selected Strains in Humans", Appl. Environ.
Microbiol., 1999, vol. 65, p. 4949-4956.
Katz, J., A. "Management of inflammatory bowel disease in adults", Journal of Digestive Diseases, 2007, vol. 8, p. 65-71 . Labus, J.S., et al. "The Visceral Sensitivity Index: development and validation of a gastrointestinal symptom-specific anxiety scale", Alimentary
Pharmacology & Therapeutics, 2004, vol. 20, p. 89-97.
Longstreth, G.F., et al. "Functional Bowel Disorders", Gastroenterology, 2006, vol. 130, p. 1480-1491 .
Maslowski, K.M., et al. "Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43", Nature, 2009, vol. 461 , p. 1282-1286.
Okayasu, I., et al. "A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mic", Gastroenterology., 1990, vol. 98, p. 694-702.
Pathmakanthan, S., et al. "Lactobacillus plantarum 299: Beneficial in vitro immunomodulation in cells extracted from inflamed human colon", Journal of Gastroenterology and Hepatology, 2004, vol. 19, p. 166-173. Rezaie, A., et al. "Oxidative Stress and Pathogenesis of Inflammatory Bowel Disease: An Epiphenomenon or the Cause?", Digestive Diseases and
Sciences, 2007, vol. 52, p. 2015-2021 .
Rodas, A.M., et al. "Polyphasic study of wine Lactobacillus strains: taxonomic implications", Int J Svst Evol Microbiol, 2005, vol. 55, p. 197-207. Roessner, A., et al. "Oxidative stress in ulcerative colitis-associated
carcinogenesis", Pathol Res Pract., 2008, vol. 204, p. 51 1 -524.
Sasaki, M., et al. "Reversal of experimental colitis disease activity in mice following administration of an adenoviral IL-10 vector", Journal of
Inflammation, 2005, vol. 2, p. 13. Scheinin, T., et al. "Validation of the interleukin-10 knockout mouse model of colitis: antitumour necrosis factor-antibodies suppress the progression of colitis.", Clin Exp Immunol, 2003, vol. 133, p. 38-43.
Tazoe, H., et al. "Roles of short-chain fatty acids receptors, GPR41 and GPR43 on colonic functions", J Physiol Pharmacol., 2008, vol. 59, p. 251 -262. Tedelind, S., et al. "Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: a study with relevance to inflammatory bowel disease", World J Gastroenterol., 2007, vol. 13, p. 2826-2832.
Vanhoutvin, S.A., et al. "The effects of butyrate enemas on visceral perception in healthy volunteers", Neurogastroenterology & Motility, 2009, vol. 21 , p. 952- e976.
Wang, Q., et al. "Naive Bayesian Classifier for Rapid Assignment of rRNA Sequences into the New Bacterial Taxonomy", Appl. Environ. Microbiol., 2007, vol. 73, p. 5261 -5267. Weisburg, W.G., et al. "16S ribosomal DNA amplification for phylogenetic study", J. Bacteriol., 1991 , vol. 173, p. 697-703.
Yamada, Y., et al. "A comparative analysis of two models of colitis in rats", Gastroenterology, 1992, vol. 102, p. 1524-1534.
Zhang, L, et al. "Alive and Dead Lactobacillus rhamnosus GG Decrease Tumor Necrosis Factor-alpha-lnduced lnterleukin-8 Production in Caco-2 Cells", J. Nutr., 2005, vol. 135, p. 1752-1756.
WO96/29083 EP 554418
EP 415941
US 7195906
Print Out (Original in Electronic Form)
(This sheet is not part of and does not count as a sheet of the international application)
Figure imgf000040_0001
1 The indications made below relate to
the deposited microorganism(s) or
other biological material referred to in
the description on:
1-1 Paragraph number 3
1-3 Identification of deposit
1 -3-1 Name of depositary institution CECT Coleccion Espanola de Cultivos Tipo
1 -3-2 Address of depositary institution Universidad de Valencia, Edificio de
Investigacion, Campus de Burjasot, 46100 Burjasot (Valencia) , Spain
1 -3-3 Date of deposit 04 February 2009 (04.02.2009)
1 -3-4 Accession Number CECT 7483,7484,7485
1-5 Designated States for Which
Indications are Made All designations
FOR RECEIVING OFFICE USE ONLY
0-4 This form was received with the
international application:
(yes or no) yes
0-4-1 Authorized officer
Pasche, Constantinus
FOR INTERNATIONAL BUREAU USE ONLY
0-5 This form was received by the
international Bureau on: 17 February 201 1 (17.02.201 1 )
0-5-1 Authorized officer
HAPPE, Isabel

Claims

1 . A composition comprising an effective amount of Lactobacillus plantarum CECT 7484, Lactobacillus plantarum CECT 7485, and Pediococcus acidilactici CECT 7483 or mutant or variants thereof.
2. A composition comprising an effective amount of Lactobacillus plantarum CECT 7484, Lactobacillus plantarum CECT 7485, and Pediococcus acidilactici CECT 7483.
3. The composition as defined in any one of the claims 1 -2 for use as a probiotic.
4. The composition as defined in any one of the claims 1 -2 for use as a medicament.
5. The composition as defined in any one of the claims 1 -2 for use as an immunomodulatory agent.
6. The composition as defined in any one of the claims 1 -2 for use in the prevention and/or treatment of bowel inflammation.
7. Use of the composition as defined in any one of the claims 1 -2 for the manufacture of a medicament for the prevention and/or treatment of bowel inflammation.
8. The composition according to claim 6 or the use according to claim 7, in the prevention and/or treatment of Inflammatory Bowel Disease.
9. The composition according to any one of the claims 1 -2 for use in the prevention and/or treatment of Irritable Bowel Syndrome.
10. The composition according to any one of the claims 1 -2 for use in the prevention and/or treatment of abdominal distension and bloating.
1 1 . A pharmaceutical product comprising an effective amount of the composition as defined in any of the claims 1 -2, together with appropriate amounts of pharmaceutically acceptable excipients.
12. A veterinary product comprising an effective amount of the composition as defined in any of the claims 1 -2, together with appropriate amounts of veterinary acceptable excipients.
13. An edible product comprising an effective amount of the composition as defined in any of the claims 1 -2, together with appropriate amounts of other edible ingredients.
14. The edible product according to claim 13, which is a dietary supplement.
15. A strain of Lactobacillus plantarum deposited in the Spanish Type Culture Collection under the accession number CECT 7484, or a mutant or variant thereof.
16. A strain of Lactobacillus plantarum deposited in the Spanish Type Culture Collection under the accession number CECT 7485, or a mutant or variant thereof.
17. A strain of Pediococcus acidilactici deposited in the Spanish Type Culture Collection under the accession number CECT 7483, or a mutant or variant thereof.
18. A Pediococcus acidilactici strain for use in the prevention and/or treatment of a gastrointestinal disease in a human.
19. The Pediococcus acidilactici according to claim 18 for use in the prevention and/or treatment of bowel inflammation.
20. The Pediococcus acidilactici according to claim 19, wherein the
gastrointestinal disease is the Inflammatory Bowel Disease (IBD).
21 . The Pediococcus acidilactici according to claim 18 for use in the prevention or treatment of Irritable Bowel Syndrome (IBS).
22. The Pediococcus acidilactici according to claim 18 for use in the treatment of abdominal distension and bloating.
23. The Pediococcus acidilactici according to any one of the claims 18-22 which is the strain of Pediococcus acidilactici deposited in the Spanish Type Culture Collection under the accession number CECT 7483.
PCT/EP2011/051170 2010-01-28 2011-01-27 Probiotic composition for use in the treatment of bowel inflammation WO2011092261A1 (en)

Priority Applications (15)

Application Number Priority Date Filing Date Title
KR1020127021713A KR101840239B1 (en) 2010-01-28 2011-01-27 Probiotic composition for use in the treatment of bowel inflammation
JP2012550449A JP5777640B2 (en) 2010-01-28 2011-01-27 Probiotic compositions for use in the treatment of intestinal inflammation
UAA201210207A UA109123C2 (en) 2010-01-28 2011-01-27 PROBIOTIC COMPOSITION FOR THE APPLICATION IN THE TREATMENT OF INTESTINAL INFLAMMATION
CN201180007399.0A CN102905712B (en) 2010-01-28 2011-01-27 Be used for the treatment of the probiotic composition of enteritis
ES11702426.5T ES2437940T3 (en) 2010-01-28 2011-01-27 Probiotic composition for use in the treatment of inflammation of the intestine
US13/575,865 US20130101566A1 (en) 2010-01-28 2011-01-27 Probiotic composition for use in the treatment of bowel inflammation
CA2787544A CA2787544C (en) 2010-01-28 2011-01-27 Probiotic composition for use in the treatment of bowel inflammation
MX2012008817A MX2012008817A (en) 2010-01-28 2011-01-27 Probiotic composition for use in the treatment of bowel inflammation.
BR112012018813-5A BR112012018813B1 (en) 2010-01-28 2011-01-27 composition comprising bacterial strains with anti-inflammatory, immunomodulating, anti-abdominal or anti-distension properties, use of these strains, as well as pharmaceutical, veterinary and edible products comprising said composition
RU2012136615/10A RU2563525C2 (en) 2010-01-28 2011-01-27 Probiotic composition for application in treatment of intestinal inflammation
AU2011209407A AU2011209407B2 (en) 2010-01-28 2011-01-27 Probiotic composition for use in the treatment of bowel inflammation
PL11702426T PL2528610T3 (en) 2010-01-28 2011-01-27 Probiotic composition for use in the treatment of bowel inflammation
EP11702426.5A EP2528610B1 (en) 2010-01-28 2011-01-27 Probiotic composition for use in the treatment of bowel inflammation
ZA2012/05683A ZA201205683B (en) 2010-01-28 2012-07-27 Probiotic composition for use in the treatment of bowel inflammation
US15/134,558 US10155015B2 (en) 2010-01-28 2016-04-21 Probiotic compositions for use in the treatment of bowel diseases

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US29911610P 2010-01-28 2010-01-28
EP10151998 2010-01-28
EP10151998.1 2010-01-28
US61/299,116 2010-01-28

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/575,865 A-371-Of-International US20130101566A1 (en) 2010-01-28 2011-01-27 Probiotic composition for use in the treatment of bowel inflammation
US15/134,558 Continuation US10155015B2 (en) 2010-01-28 2016-04-21 Probiotic compositions for use in the treatment of bowel diseases

Publications (1)

Publication Number Publication Date
WO2011092261A1 true WO2011092261A1 (en) 2011-08-04

Family

ID=42243204

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/051170 WO2011092261A1 (en) 2010-01-28 2011-01-27 Probiotic composition for use in the treatment of bowel inflammation

Country Status (18)

Country Link
US (2) US20130101566A1 (en)
EP (1) EP2528610B1 (en)
JP (1) JP5777640B2 (en)
KR (1) KR101840239B1 (en)
CN (1) CN102905712B (en)
AU (1) AU2011209407B2 (en)
BR (1) BR112012018813B1 (en)
CA (1) CA2787544C (en)
CL (1) CL2012002062A1 (en)
CO (1) CO6511276A2 (en)
ES (1) ES2437940T3 (en)
MX (1) MX2012008817A (en)
MY (1) MY162344A (en)
PL (1) PL2528610T3 (en)
RU (1) RU2563525C2 (en)
UA (1) UA109123C2 (en)
WO (1) WO2011092261A1 (en)
ZA (1) ZA201205683B (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012170915A1 (en) 2011-06-10 2012-12-13 Prothera Inc. Pharmaceutical compositions containing pediococcus and methods for reducing the symptoms of gastroenterological syndromes
JP2014031325A (en) * 2012-08-02 2014-02-20 Maruzen Pharmaceut Co Ltd Anti-inflammatory agent
WO2014168474A1 (en) * 2013-04-08 2014-10-16 N.V. Nutricia Fermented nutritional composition with thiol protease inhibitor
WO2015099617A1 (en) 2013-12-23 2015-07-02 Medis, D.O.O. New strains of the genus lactobacillus and use thereof
US20160151434A1 (en) * 2011-06-10 2016-06-02 Amorepacific Corporation Novel lactobacillus plantarum isolated from leaves of camellia sinensis
WO2016083450A3 (en) * 2014-11-25 2016-07-21 Dsm Ip Assets B.V. Lactobillus for use as probiotic and blood cell populations used for evaluating immune response to agents, e.g. probiotics
KR102036275B1 (en) * 2018-10-02 2019-10-24 서울대학교 산학협력단 A new Pediococcus acidilactici LDTM 5201 having advanced anti-inflammation(KCTC 13563BP)
WO2022171304A1 (en) 2021-02-12 2022-08-18 Ab-Biotics, S.A. Probiotic composition for the treatment of covid-19

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014096901A1 (en) * 2012-12-18 2014-06-26 Compagnie Gervais Danone Strain of bifidobacterium animalis ssp. animalis
SG11201604840TA (en) * 2014-02-28 2016-09-29 Meiji Co Ltd Prophylactic and/or therapeutic agent for functional gastrointestinal disorders
US10967013B2 (en) * 2014-04-23 2021-04-06 National Yang-Ming University Lactic acid bacterium for prophylaxis or treatment of a stress-induced disorder and a composition containing the same
US10188684B2 (en) * 2015-04-20 2019-01-29 National Yang-Ming University Method for preventing or treating functional gastrointestinal disorder by lactic acid bacterium
NZ732197A (en) * 2014-11-28 2022-07-01 Anagenix Ip Ltd Gold kiwifruit compositions and methods of preparation and use therefor
US10946050B2 (en) 2015-03-16 2021-03-16 Imagilin Technology Llc Compositions comprising probiotics and methods of use thereof
US10195237B2 (en) * 2015-03-16 2019-02-05 Imagilin Technology Llc Compositions and methods for treating inflammatory related diseases or conditions using Pediococcus acidilactici probiotics
MA41010B1 (en) * 2015-06-15 2020-01-31 4D Pharma Res Ltd Compositions comprising bacterial strains
US10857190B2 (en) 2015-10-06 2020-12-08 Albert Einstein College Of Medicine Microbial hyperswarmers and uses thereof
KR101999259B1 (en) 2017-01-31 2019-07-11 경희대학교 산학협력단 Novel lactic acid bacteria and use thereof
KR102464397B1 (en) * 2017-09-15 2022-11-08 (주)아모레퍼시픽 Composition for preventing and improving gastrointestinal disorder containing lactobacillus plantarum
RU2764639C1 (en) * 2018-02-02 2022-01-19 Кобиолабс, Инк. Lactobacillus plantarum strain kbl396 and application thereof
AU2019236242A1 (en) * 2018-03-15 2020-10-01 University Of Southern California Fasting-mimicking diet (FMD) but not water-only fasting promotes reversal of inflammation and IBD pathology
AR115757A1 (en) * 2018-07-13 2021-02-24 Cj Cheiljedang Corp COMPOSITION INCLUDING LACTOBACILLUS PLANTARUM STRAIN CJLP475 AND LACTOBACILLUS PLANTARUM STRAIN CJLP17 AND THE USE OF THE SAME
KR102118343B1 (en) * 2018-07-13 2020-06-03 롯데푸드 주식회사 A milk and fermented milk having anti-inflammatory effects on intestinal epithelial cells and having excellent ability to reach to intestinal epithelial cells
KR102038695B1 (en) * 2018-10-30 2019-10-30 주식회사 종근당바이오 Composition for Preventing or Treating Alcoholic Intestinal Damage Comprising Probiotics as Effective Ingredients
WO2020124088A1 (en) * 2018-12-14 2020-06-18 The University Of North Carolina At Chapel Hill Lachnospiraceae mitigates against radiation-induced hematopoietic/gastrointestinal injury and death, and promotes cancer control by radiation
KR102094415B1 (en) * 2018-12-17 2020-03-30 강원대학교산학협력단 Pediococcus acidilactici having probiotic activity and antimicrobial activity
KR102500854B1 (en) * 2019-12-17 2023-02-17 한국 한의학 연구원 Composition for relieving hangover comprising Pediococcus inopinatus as effective component
WO2023176849A1 (en) * 2022-03-17 2023-09-21 株式会社カネカ Glucuronidase activator, pharmaceutical composition, edible composition, and composition for oral application
CN114921351B (en) * 2022-05-06 2023-06-13 大连工业大学 Pichia pastoris DPUY-F1 with probiotics function and application thereof in relieving colonitis symptoms
CN116083277B (en) * 2022-09-17 2024-02-13 浙江大学 Pediococcus acidilactici ZJUIDS13 with ulcerative colitis relieving effect and application thereof
WO2024136615A1 (en) * 2022-12-23 2024-06-27 주식회사 씨티씨바이오 Lactobacillus having prophylactic or therapeutic activity for colitis disease and antibacterial activity against harmful gut bacteria and use thereof

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0415941A1 (en) 1988-03-09 1991-03-13 Kabivitrum Ab Nutrient composition and method for the preparation thereof.
EP0554418A1 (en) 1991-07-25 1993-08-11 Probi Ab Intestine colonizing lactobacilli
WO1996029083A1 (en) 1995-03-23 1996-09-26 Probi Ab Epithelial adhesive lactobacilli
WO2004110466A2 (en) * 2003-06-13 2004-12-23 N.V. Nutricia Pediocin-producing pediococci
US7195906B2 (en) 1999-01-15 2007-03-27 Enterprise Ireland (Trading As Bioresearch Ireland) Bifidobacterium in the treatment of inflammatory disease
WO2007150052A1 (en) * 2006-06-23 2007-12-27 Equine Research Associates, Ltd Animal nutritional supplement and method
JP2008013534A (en) * 2006-07-07 2008-01-24 Health Vision:Kk Probiotics product for pet animal
US20100196323A1 (en) * 2005-06-14 2010-08-05 Erber Aktiengesellschaft Probiotic health or fitness promoting human or animal foodstuff and/or drinking water additive and use thereof
WO2010114864A1 (en) * 2009-04-01 2010-10-07 Little Calumet Holdings, Llc Probiotic oral dosage forms

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2813790B1 (en) * 2000-09-11 2004-03-12 Dolisos Lab PHARMACEUTICAL PREPARATIONS CONTAINING SOY ISOFLAVONE EXTRACTS AND PROBIOTIC MICROORGANISMS
EP1384483A1 (en) * 2002-07-23 2004-01-28 Nestec S.A. Probiotics for treatment of irritable bowel disease (IBS) through improvement of gut neuromuscular function
DE602004030922D1 (en) * 2003-08-21 2011-02-17 Otsuka Pharma Co Ltd MILKY ACID BACTERIA WITH MUCOSA IMMUNOTHOTE EFFECT
EP1714660A1 (en) * 2005-04-21 2006-10-25 N.V. Nutricia Uronic acid and probiotics
ES2460891T3 (en) * 2005-10-06 2014-05-14 Probi Ab Use of lactobacillus for the treatment of autoimmune diseases
ATE483366T1 (en) * 2005-11-14 2010-10-15 Unilever Nv EDIBLE PRODUCT CONTAINING GINSENG POLYSACCHARIDES AND HEALTHY BACTERIA
SG188887A1 (en) * 2008-03-14 2013-04-30 Nestec Sa Synbiotic mixture
US8298526B2 (en) * 2010-06-02 2012-10-30 Genmont Biotech Inc. Lactobacillus strain, composition and use thereof for improving the syndrome of diabetes and complication thereof
CA2923982A1 (en) * 2013-08-12 2015-02-19 Mansel Griffiths Antiviral methods and compositions comprising probiotic bacterial molecules

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0415941A1 (en) 1988-03-09 1991-03-13 Kabivitrum Ab Nutrient composition and method for the preparation thereof.
EP0554418A1 (en) 1991-07-25 1993-08-11 Probi Ab Intestine colonizing lactobacilli
WO1996029083A1 (en) 1995-03-23 1996-09-26 Probi Ab Epithelial adhesive lactobacilli
US7195906B2 (en) 1999-01-15 2007-03-27 Enterprise Ireland (Trading As Bioresearch Ireland) Bifidobacterium in the treatment of inflammatory disease
WO2004110466A2 (en) * 2003-06-13 2004-12-23 N.V. Nutricia Pediocin-producing pediococci
US20100196323A1 (en) * 2005-06-14 2010-08-05 Erber Aktiengesellschaft Probiotic health or fitness promoting human or animal foodstuff and/or drinking water additive and use thereof
WO2007150052A1 (en) * 2006-06-23 2007-12-27 Equine Research Associates, Ltd Animal nutritional supplement and method
JP2008013534A (en) * 2006-07-07 2008-01-24 Health Vision:Kk Probiotics product for pet animal
WO2010114864A1 (en) * 2009-04-01 2010-10-07 Little Calumet Holdings, Llc Probiotic oral dosage forms

Non-Patent Citations (41)

* Cited by examiner, † Cited by third party
Title
AGRAWAL ANURAG ET AL: "Fermented milk containing the probiotic bifidobacterium animalis, DN-173 010 (FM) improves abdominal distension, bloating and transit in irritable bowel syndrome with constipation (IBS-C)", GASTROENTEROLOGY, ELSEVIER, PHILADELPHIA, PA, vol. 134, no. 4, Suppl. 1, 1 April 2008 (2008-04-01), pages A546, XP009107533, ISSN: 0016-5085, DOI: DOI:10.1016/S0016-5085(08)62549-4 *
ALTSCHUL, S.F. ET AL.: "Basic local alignment search tool", J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ANADON, A. ET AL.: "Opinion of the Scientific Panel on Additives and Products or Substances used in Animal Feed on the updating of the criteria used in the assessment of bacteria for resistance to antibiotics of human veterinary importance", THE EFSA JOURNAL, vol. 233, 2005, pages 1 - 12
ANDREOLETTI, O. ET AL.: "The maintenance of the list of QPS microorganisms intentionally added to food or feed. Question no: EFSA-Q-2008-006", THE EFSA JOURNAL, vol. 923, 2008, pages 1 - 48
ARAYA, M. ET AL.: "Guidelines for the Evaluation of Probiotics in Food - Joint FAO/WHO Working Group", 2002, FAO/WHO
BADIA, X. ET AL.: "Adaptacion al espanol del cuestionario IBSQoL para la medicion de la calidad de vida en pacientes con sindrome de intestino irritable", REV ESP ENFERM DIG, vol. 92, 2000, pages 637 - 643
BAROUEI J ET AL: "Prophylactic role of maternal administration of probiotics in the prevention of irritable bowel syndrome", MEDICAL HYPOTHESES, EDEN PRESS, PENRITH, US, vol. 73, no. 5, 1 November 2009 (2009-11-01), pages 764 - 767, XP026680495, ISSN: 0306-9877, [retrieved on 20090528], DOI: DOI:10.1016/J.MEHY.2009.04.023 *
BORIES, G. ET AL.: "Update on the criteria used in the assessment of bacterial resistance to antibiotics of human or veterinary importance", THE EFSA JOURNAL, vol. 732, 2008, pages 1 - 15
COLLADO, M. ET AL.: "Probiotic Strains and Their Combination Inhibit In Vitro Adhesion of Pathogens to Pig Intestinal Mucosa", CURRENT MICROBIOLOGY, vol. 55, 2007, pages 260 - 265
COOPER, H.S. ET AL.: "Clinicopathologic study of dextran sulfate sodium experimental murine colitis", LAB INVEST., vol. 69, 1993, pages 238 - 249
DANIEL, C. ET AL.: "Selecting Lactic Acid Bacteria for Their Safety and Functionality by Use of a Mouse Colitis Model", APPL. ENVIRON. MICROBIOL., vol. 72, 2006, pages 5799 - 5805
D'ARGENIO, G., MAZZACCA, G.: "Short-chain fatty acid in the human colon. Relation to inflammatory bowel diseases and colon cancer", ADV EXP MED BIOL, vol. 472, 1999, pages 149 - 158
DATABASE WPI Week 200845, Derwent World Patents Index; AN 2008-H07141, XP002625809 *
DEAN, B.B. ET AL.: "Impairment in work productivity and health-related quality of life in patients with IBS", AM J MANAQ CARE., vol. 11, 2005, pages 17 - 26
FITZPATRICK, L.R. ET AL.: "Effects of the probiotic formulation VSL#3 on colitis in weanling rats", J PEDIATR GASTROENTEROL NUTR., vol. 44, 2007, pages 561 - 570
FUKUMOTO, S. ET AL.: "Short-chain fatty acids stimulate colonic transit via intraluminal 5-HT release in rats", AM J PHYSIOL REQUL INTEQR COMP PHYSIOL, vol. 284, 2003, pages R1269 - 1276
GRABIG, A. ET AL.: "Escherichia coli Strain Nissle 1917 Ameliorates Experimental Colitis via Toll-Like Receptor 2- and Toll-Like Receptor 4-Dependent Pathways", INFECT. IMMUN., vol. 74, 2006, pages 4075 - 4082
GUARNER, F., SCHAAFSMA, G.J.: "Probiotics", INT J FOOD MICROBIOL., vol. 39, 1998, pages 237 - 238
HART AILSA L ET AL: "Use of probiotics in the treatment of inflammatory bowel disease", JOURNAL OF CLINICAL GASTROENTEROLOGY, RAVEN PRESS LTD., NEW YORK, NY, US, vol. 36, no. 2, 1 February 2003 (2003-02-01), pages 111 - 119, XP009127371, ISSN: 0192-0790, DOI: DOI:10.1097/00004836-200302000-00005 *
JACOBSEN, C.N. ET AL.: "Screening of Probiotic Activities of Forty-Seven Strains of Lactobacillus spp. by In Vitro Techniques and Evaluation of the Colonization Ability of Five Selected Strains in Humans", APPL. ENVIRON. MICROBIOL., vol. 65, 1999, pages 4949 - 4956
JONKERS DAISY ET AL: "Probiotics and inflammatory bowel disease.", April 2003, JOURNAL OF THE ROYAL SOCIETY OF MEDICINE APR 2003 LNKD- PUBMED:12668702, VOL. 96, NR. 4, PAGE(S) 167 - 171, ISSN: 0141-0768, XP002587820 *
KATZ, J., A.: "Management of inflammatory bowel disease in adults", JOURNAL OF DIQESTIVE DISEASES, vol. 8, 2007, pages 65 - 71
LABUS, J.S. ET AL.: "The Visceral Sensitivity Index: development and validation of a gastrointestinal symptom-specific anxiety scale", ALIMENTARY PHARMACOLOQV & THERAPEUTICS, vol. 20, 2004, pages 89 - 97
LONGSTRETH, G.F. ET AL.: "Functional Bowel Disorders", GASTROENTEROLOQV, vol. 130, 2006, pages 1480 - 1491
MASLOWSKI, K.M. ET AL.: "Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43", NATURE, vol. 461, 2009, pages 1282 - 1286
OKAYASU ET AL., GASTROENTEROL, 1990
OKAYASU, I. ET AL.: "A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mic", GASTROENTEROLOQV., vol. 98, 1990, pages 694 - 702
PATHMAKANTHAN, S. ET AL.: "Lactobacillus plantarum 299: Beneficial in vitro immunomodulation in cells extracted from inflamed human colon", JOURNAL OF GASTROENTEROLOAY AND HEPATOLOQY, vol. 19, 2004, pages 166 - 173
REZAIE, A. ET AL.: "Oxidative Stress and Pathogenesis of Inflammatory Bowel Disease: An Epiphenomenon or the Cause?", DIQESTIVE DISEASES AND SCIENCES, vol. 52, 2007, pages 2015 - 2021
RODAS, A.M. ET AL.: "Polyphasic study of wine Lactobacillus strains: taxonomic implications", INT J SVST EVOL MICROBIOL, vol. 55, 2005, pages 197 - 207
ROESSNER, A. ET AL.: "Oxidative stress in ulcerative colitis-associated carcinogenesis", PATHOL RES PRACT., vol. 204, 2008, pages 511 - 524
SASAKI, M. ET AL.: "Reversal of experimental colitis disease activity in mice following administration of an adenoviral IL-10 vector", JOURNAL OF INFLAMMATION, vol. 2, 2005, pages 13
SCHEININ, T. ET AL.: "Validation of the interleukin-10 knockout mouse model of colitis: antitumour necrosis factor-antibodies suppress the progression of colitis", CLIN EXP IMMUNOL, vol. 133, 2003, pages 38 - 43
SHANAHAN FERGUS: "Probiotics in inflammatory bowel disease--therapeutic rationale and role", ADVANCED DRUG DELIVERY REVIEWS, ELSEVIER BV, AMSTERDAM, NL LNKD- DOI:10.1016/J.ADDR.2003.11.003, vol. 56, no. 6, 19 April 2004 (2004-04-19), pages 809 - 818, XP002283007, ISSN: 0169-409X *
TAZOE, H. ET AL.: "Roles of short-chain fatty acids receptors, GPR41 and GPR43 on colonic functions", J PHYSIOL PHARMACOL., vol. 59, 2008, pages 251 - 262
TEDELIND, S. ET AL.: "Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: a study with relevance to inflammatory bowel disease", WORLD J GASTROENTEROL., vol. 13, 2007, pages 2826 - 2832
VANHOUTVIN, S.A. ET AL.: "The effects of butyrate enemas on visceral perception in healthy volunteers", NEUROQASTROENTEROLOQY & MOTILITY, vol. 21, 2009, pages 952 - E976
WANG, Q. ET AL.: "Naive Bayesian Classifier for Rapid Assignment of rRNA Sequences into the New Bacterial Taxonomy", APPL. ENVIRON. MICROBIOL., vol. 73, 2007, pages 5261 - 5267
WEISBURG, W.G. ET AL.: "16S ribosomal DNA amplification for phylogenetic study", J. BACTERIOL., vol. 173, 1991, pages 697 - 703
YAMADA, Y. ET AL.: "A comparative analysis of two models of colitis in rats", GASTROENTEROLOQV, vol. 102, 1992, pages 1524 - 1534
ZHANG, L. ET AL.: "Alive and Dead Lactobacillus rhamnosus GG Decrease Tumor Necrosis Factor-alpha-Induced Interleukin-8 Production in Caco-2 Cells", J. NUTR., vol. 135, 2005, pages 1752 - 1756

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160228480A1 (en) * 2011-06-10 2016-08-11 Amorepacific Corporation Novel lactobacillus plantarum isolated from leaves of camellia sinensis
EP2776048A1 (en) * 2011-06-10 2014-09-17 Prothera, Inc. Pharmaceutical compositions containing pediococcus and methods for reducing the symptoms of gastroenterological syndromes
AU2012267516B2 (en) * 2011-06-10 2017-08-10 Prothera Inc. Pharmaceutical compositions containing pediococcus and methods for reducing the symptoms of gastroenterological syndromes
WO2012170915A1 (en) 2011-06-10 2012-12-13 Prothera Inc. Pharmaceutical compositions containing pediococcus and methods for reducing the symptoms of gastroenterological syndromes
EP3598979A1 (en) * 2011-06-10 2020-01-29 Prothera, Inc. Pharmaceutical compositions containing pediococcus for reducing the symptoms of gastroenterological syndromes
EP2776048A4 (en) * 2011-06-10 2015-08-19 Prothera Inc Pharmaceutical compositions containing pediococcus and methods for reducing the symptoms of gastroenterological syndromes
US20160151434A1 (en) * 2011-06-10 2016-06-02 Amorepacific Corporation Novel lactobacillus plantarum isolated from leaves of camellia sinensis
US9895402B2 (en) * 2011-06-10 2018-02-20 Amorepacific Corporation Lactobacillus plantarum isolated from leaves of Camellia sinensis
US9782446B2 (en) * 2011-06-10 2017-10-10 Amorepacific Corporation Lactobacillus plantarum isolated from leaves of Camellia sinensis
US20160228479A1 (en) * 2011-06-10 2016-08-11 Amorepacific Corporation Novel lactobacillus plantarum isolated from leaves of camellia sinensis
US9895401B2 (en) * 2011-06-10 2018-02-20 Amorepacific Corporation Lactobacillus plantarum isolated from leaves of Camellia sinensis
US9889167B2 (en) * 2011-06-10 2018-02-13 Amorepacific Corporation Lactobacillus plantarum isolated from leaves of Camellia sinensis
US20160228478A1 (en) * 2011-06-10 2016-08-11 Amorepacific Corporation Novel lactobacillus plantarum isolated from leaves of camellia sinensis
JP2014031325A (en) * 2012-08-02 2014-02-20 Maruzen Pharmaceut Co Ltd Anti-inflammatory agent
WO2014168474A1 (en) * 2013-04-08 2014-10-16 N.V. Nutricia Fermented nutritional composition with thiol protease inhibitor
US10098916B2 (en) 2013-12-23 2018-10-16 Medis, D.O.O. Strains of the genus Lactobacillus and use thereof
WO2015099617A1 (en) 2013-12-23 2015-07-02 Medis, D.O.O. New strains of the genus lactobacillus and use thereof
WO2016083450A3 (en) * 2014-11-25 2016-07-21 Dsm Ip Assets B.V. Lactobillus for use as probiotic and blood cell populations used for evaluating immune response to agents, e.g. probiotics
US10226492B2 (en) 2014-11-25 2019-03-12 Dsm Ip Assets B.V. Probiotic and new biomarker
KR102036275B1 (en) * 2018-10-02 2019-10-24 서울대학교 산학협력단 A new Pediococcus acidilactici LDTM 5201 having advanced anti-inflammation(KCTC 13563BP)
WO2022171779A1 (en) 2021-02-12 2022-08-18 Ab-Biotics, S.A. Probiotic composition for the treatment of covid-19
WO2022171304A1 (en) 2021-02-12 2022-08-18 Ab-Biotics, S.A. Probiotic composition for the treatment of covid-19

Also Published As

Publication number Publication date
RU2012136615A (en) 2014-03-10
MY162344A (en) 2017-06-15
AU2011209407B2 (en) 2014-02-27
CL2012002062A1 (en) 2013-10-04
CN102905712A (en) 2013-01-30
MX2012008817A (en) 2012-11-23
US20160220620A1 (en) 2016-08-04
US10155015B2 (en) 2018-12-18
RU2563525C2 (en) 2015-09-20
BR112012018813B1 (en) 2020-12-08
BR112012018813A2 (en) 2017-06-20
US20130101566A1 (en) 2013-04-25
ES2437940T3 (en) 2014-01-15
PL2528610T3 (en) 2014-02-28
CA2787544A1 (en) 2011-08-04
CA2787544C (en) 2019-07-09
EP2528610B1 (en) 2013-11-13
JP2013517784A (en) 2013-05-20
CN102905712B (en) 2015-12-16
CO6511276A2 (en) 2012-08-31
KR20130002372A (en) 2013-01-08
ZA201205683B (en) 2013-08-28
JP5777640B2 (en) 2015-09-09
KR101840239B1 (en) 2018-03-20
UA109123C2 (en) 2015-07-27
AU2011209407A1 (en) 2012-09-20
EP2528610A1 (en) 2012-12-05

Similar Documents

Publication Publication Date Title
US10155015B2 (en) Probiotic compositions for use in the treatment of bowel diseases
EP2485743B1 (en) Lactobacillus plantarum strains as hypocholesterolemic agents.
CA2837479C (en) Pharmaceutical compositions containing pediococcus and methods for reducing the symptoms of gastroenterological syndromes
KR101353692B1 (en) Compositions comprising probiotic and prebiotic components and mineral salts, with lactoferrin
US20200121740A1 (en) Pharmaceutical compositions containing pediococcus and methods for reducing the symptoms of gastroenterological syndromes
BR112012011315B1 (en) Formulation comprising probiotic bifidobacteria strain
JP2004097222A (en) Bacillus bifidus and preparation containing the same
EP2220210B1 (en) Strains of lactobacillus plantarum as probiotics with immunomodulatory specific effect

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180007399.0

Country of ref document: CN

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

Ref document number: 11702426

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2787544

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2012002062

Country of ref document: CL

Ref document number: 12125095

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 12012501540

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2012550449

Country of ref document: JP

Ref document number: 13575865

Country of ref document: US

Ref document number: MX/A/2012/008817

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20127021713

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2011702426

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2011209407

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2381/KOLNP/2012

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: A201210207

Country of ref document: UA

WWE Wipo information: entry into national phase

Ref document number: 2012136615

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2011209407

Country of ref document: AU

Date of ref document: 20110127

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112012018813

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112012018813

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20120727