WO2010027344A1 - Extraits immunomodulateurs de bactéries lactobacillus et procédés de fabrication et d’utilisation de ceux-ci - Google Patents

Extraits immunomodulateurs de bactéries lactobacillus et procédés de fabrication et d’utilisation de ceux-ci Download PDF

Info

Publication number
WO2010027344A1
WO2010027344A1 PCT/US2008/010350 US2008010350W WO2010027344A1 WO 2010027344 A1 WO2010027344 A1 WO 2010027344A1 US 2008010350 W US2008010350 W US 2008010350W WO 2010027344 A1 WO2010027344 A1 WO 2010027344A1
Authority
WO
WIPO (PCT)
Prior art keywords
extract
lactobacillus
hours
extracts
bacterial
Prior art date
Application number
PCT/US2008/010350
Other languages
English (en)
Inventor
Jacques Alain Bauer
Marco Salvagni
Jean-Pierre Leon Vigroux
Laetitia Leela Gisele Chalvet
Carlo Chiavaroli
Original Assignee
Om Pharma
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 JP2011526017A priority Critical patent/JP2012502026A/ja
Priority to CN2008801309602A priority patent/CN102143756A/zh
Priority to BRPI0822997-0A priority patent/BRPI0822997A2/pt
Priority to AU2008361375A priority patent/AU2008361375B2/en
Priority to PCT/US2008/010350 priority patent/WO2010027344A1/fr
Priority to RU2011112781/15A priority patent/RU2500412C2/ru
Application filed by Om Pharma filed Critical Om Pharma
Priority to MX2011002080A priority patent/MX2011002080A/es
Priority to KR1020117007527A priority patent/KR20110051268A/ko
Priority to CA2733249A priority patent/CA2733249A1/fr
Publication of WO2010027344A1 publication Critical patent/WO2010027344A1/fr
Priority to ZA2011/00899A priority patent/ZA201100899B/en
Priority to IL211041A priority patent/IL211041A0/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
    • 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
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • 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
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12RINDEXING SCHEME ASSOCIATED WITH SUBCLASSES C12C - C12Q, RELATING TO MICROORGANISMS
    • C12R2001/00Microorganisms ; Processes using microorganisms
    • C12R2001/01Bacteria or Actinomycetales ; using bacteria or Actinomycetales
    • C12R2001/225Lactobacillus

Definitions

  • Embodiments of the present invention include extracts from
  • Lactobacillus bacteria which may produce immunomodulatory effects in subjects.
  • Embodiments of the invention may be used, for example, as nutraceuticals or pharmaceuticals for treatment of diseases, such as those related to an imbalance of the production of anti-inflammatory or proinflammatory cytokines, such as infections, allergies, autoimmunity disorders, and inflammation, or as adjuvants providing healthful benefits in subjects.
  • the invention also includes, inter alia, methods of making and using such extracts.
  • the invention also relates to particular strains of Lactobacillus bacteria.
  • lmmunomodulation is a global term that refers to a wide range of immune intervention that alters normal or abnormal immune responses.
  • Microbes produce and secrete a wide range of molecules that can modulate eukaryotic immune responses (Lavelle et al., Curr Top Med Chem. 2004, 4(5), 499-508). These include factors that subvert protective mechanisms in order to facilitate pathogen colonization and persistence.
  • Viral, bacterial and parasite-derived molecules that can inhibit inflammatory responses have been identified.
  • potent immune activators may also be of microbial origin themselves. These include bacterial enterotoxins, parasite-derived excretory-secretory products, and viral nucleic acids.
  • TLRs toll-like receptors
  • Figure 1 provides a list of TLR ligands (Gay and Gangloff, Ann. Rev. Biochem., 2007, 76:141-65). TLRs recognize a wide range of molecules also known as pathogen-associated molecular patterns (PAMP) produced by viruses, bacteria and fungi (Tse and Homer, Ann Rheum Dis. 2007 Nov; 66 Suppl 3:iii77-80). TLR-linked immunomodulation has been applied in the development of novel therapies for a wide spectrum of pathologies, including infectious, malignant, autoimmune and allergic diseases.
  • PAMP pathogen-associated molecular patterns
  • TLR agonists and antagonists have been studied as potential therapeutics for the prevention and treatment of diseases.
  • TLR agonists have been used as adjuvants for vaccines aimed at preventing infections, extinguishing allergic hypersensitivities and clearing malignant cells.
  • TLR agonists have also been investigated as monotherapies and adjunct therapies for the treatment of patients with infectious, allergic and malignant diseases.
  • the use of TLR antagonists have also been studied in preclinical studies and clinical trials as potential therapeutics for autoimmune diseases and sepsis.
  • Probiotics are live microorganisms that may provide health benefits to a subject when administered in adequate amounts (Mottet et al., Digestive and Liver Disease, 2005, 37:3-6; Ezendam et al., Nutr Rev, Jan. 2006, 64(1):1 -14; Gill and Prasad, Adv Exp Med Biol, 2008, 606:423-54).
  • the biological mechanisms involved in immune response stimulation by probiotic microorganisms and certain cellular components of those microorganisms have been a subject of study.
  • gram-positive bacteria have a characteristic cell wall comprising macromolecules such as lipoteichoic acid (LTA).
  • LTA may be associated with immunostimulatory activity (e.g., Bhakdi et al., Infect. Immun., 1991 , 59:4614-4620; Setoyama et al., J Gen Microbiol, 1985, 131 (9):2501-2503; Cleveland et al., Infect Immun, 1996, 64(6):1906-1912). See also (Deininger et al., Clin Vaccine Immunol, 2007, 14(2): 1629-1633).
  • probiotic bacteria may contain a variety of TLR ligands with immunomodulatory characteriistics. Cell wall fragments from various bifidobacteria!
  • live cell extracts may comprise large proteins and lipopeptides whose size hinders efficient absorption by the subject, thus limiting the local concentration of helpful molecules from probiotic bacteria in the body.
  • Conditions inside the body may also destroy active bacterial components or otherwise modify the chemical structures of those components, rendering them inactive.
  • Risks associated with the oral administration of live probiotic microorganisms include bacteremia and sepsis (Lactobacillus Sepsis Associated With Probiotic Therapy, Pediatrics, Jan. 2005, 115 (1 ):178-181 ).
  • the present invention relates to Lactobacillus extracts, some embodiments of which may display strong immununomodulatory activities.
  • embodiments of the present invention relate to extracts from bacterial strains that may be useful as nutraceuticals or as pharmaceuticals, in some cases, to treat infectious diseases, allergy, respiratory disorders, and inflammatory pathologies, or to act as an adjunct in connection with a treatment protocol.
  • the present invention also relates to compositions comprising the extracts, and processes of making the extracts, for example using media that do not pose a risk of prion diseases.
  • Processes according to the invention include, for example, lysis of cells under alkaline conditions, or under alkaline conditions followed by acidic conditions.
  • the extracts of the invention are soluble extracts, meaning that they do not contain significant amounts of solid or particulate matter.
  • the extracts contain chemically modified TLR ligands.
  • alkaline treatment may cause chemical modification of cellular materials including TLR ligands, cell wall components, proteins, lipoteichoic acids, lipopeptides and phospholipids.
  • Some embodiments of the invention may comprise extracts obtained from one or more of the following species:
  • Lactobacillus fermentum Lactobacillus rhamnosus, Lactobacillus plantarum, Lactobacillus johnsonii, Lactobacillus helveticus, Lactobacillus casei defensis, Lactobacillus casei ssp. casei, Lactobacillus paracasei, Lactobacillus bulgaricus, Lactobacillus paracasei, Lactobacillus acidophilus, Lactobacillus reuteri, Lactobacillus salivarius, Lactobacillus lactis, and Lactobacillus delbrueckii.
  • the extracts comprise at least one strain from each of the above species of bacteria, while in other embodiments, one or more specific strains from the list above may be removed or substituted with one or more different strains.
  • Some embodiments of the present invention comprise an extract obtained from one or more of the following bacterial strains: Lactobacillus fermentum 1-3929, Lactobacillus rhamnosus 71.38, Lactobacillus plantarum 71.39, Lactobacillus johnsonii 103782, and Lactobacillus helveticus 103146. The strains above are deposited according to the Budapest Treaty.
  • Lactobacillus fermentum 1-3929 Lactobacillus rhamnosus 71.38, Lactobacillus plantarum 71.39, Lactobacillus johnsonii 103782, and Lactobacillus helveticus 103146 are each deposited at the Collection Nationale de Culture des Microorganismes at the lnstitut Pasteur, 25 rue du Dr. Roux, 75724 Paris, France. Lactobacillus fermentum 1-3929 was deposited on February 27, 2008. The other strains are among the depository's collections and may be obtained by contacting the depository.
  • This invention also relates, inter alia, to the strain Lactobacillus fermentum 1-3929, extracts obtained from that strain, methods of making such extracts, and the uses thereof. That strain was obtained by allowing strains from Lactobacillus plantarum and Lactobacillus fermentum Xo undergo chromosomal exchange, thus producing a novel Lactobacillus strain. Extracts obtained from Lactobacillus fermentum 1-3929 were found to be active in several different in vivo and in vitro models correlating to infection and immunological disorders.
  • an extract is obtained from only one specific bacterial strain. Alternatively, more than one strain may be used. In other embodiments, one or more extracts from a different type of microorganism, such as from an non- Lactobacillus bacterial species, may be added.
  • the extracts may be obtained by lysing bacterial cells in specific conditions after the cells are grown to a suitable concentration in a culture medium.
  • the bacteria are grown in a medium that does not pose a risk of prion-related diseases or a risk of other diseases that may be transmitted through ingesting products obtained from animal-based media.
  • a vegetable-based medium is used to grow the cells, such as a soya-based medium.
  • the lysates may also be filtered to remove nucleic acids and larger cellular debris, such as insoluble or particulate matter.
  • the amount of nucleic acids present in the extracts is less than 100 ⁇ g/mL.
  • the resulting extract comprises soluble molecular components and does not contain significant amounts of insoluble or particulate material.
  • Membrane and cell wall molecules may be dissolved or suspended in the extracts, including lipoproteins, lipopeptides, peptidoglycans, lipooligosaccharides, lipoteichoic acids, and teichoic acids.
  • molecules in the cells such as in membranes and cell walls, may become chemically modified, for example, cleaved into smaller structures, by alkaline treatment.
  • embodiments of the invention may retain their biological activity in comparison to whole cells, or such embodiments may even demonstrate enhanced biological activities in comparison to whole cells.
  • alkaline treatment may be used to lyse cells, or may be applied to cells that have previously been lysed by another method.
  • L- amino acids found in natural proteins and lipopeptides are at least partially racemized to D-amino acids.
  • D-amino acids can be beneficial in increasing the time of effectiveness of the extracts, as they are not efficiently digested in the mammalian gut. D-amino acids may also protect smaller peptides and proteins from degradation during digestion.
  • D-amino acids examples include protein- bound D-amino acids, and to a lesser extent lysinoalanine (de Vrese et al., J Nutrition, 2000, 2026-2031).
  • antigenic molecules in the extracts chemically modified during lysis to contain D-amino acids may remain in the patient's body for a longer time, potentially allowing for a stronger immunostimulating effect in some embodiments.
  • a filtration process may also influence the properties of the resulting extracts, as the pore size of the filter, and in some cases, the chemical properties of the filter surface (i.e, its polarity), may alter the type of materials that are removed and retained.
  • some embodiments of the instant invention use a filtration process designed to retain molecules of interest but remove other molecular components such as nucleic acids or insoluble or particulate matter.
  • the filtered extracts may also be further purified by organic extraction, organo-aqueous extraction, chromatography, ultracentrifugation, ultrafiltration, or a combination of thereof.
  • FIG. 1 Ligands for the family of 11 toll-like receptors (TLRs), expressed by the host organism.
  • FIG. 2 A diagram of a tangential flow filtration (TFF) system for preparation of bacterial extracts following lysis of bacteria.
  • the diagram shows two different configurations for filters: a parallel mode where all filters work simultaneously and a serpentine mode where filters are configured in a serial mode.
  • FIG. 3 Generalized correlation between operating parameters and flux, inicating the areas of pressure control and mass transfer control for tangential flow filtration process (TFF).
  • Figure 4 Stimulation of spleen cells cultured for 48 hours in the presence of different dilutions of the lysates AFer300, CFer300, and DFer300, and ARahr300, CRahr300, and DRahr300. After the addition of 30 ⁇ l/well Alamar blue® solution diluted 1 :1 in cell culture medium, cells were further incubated (a) 8.5 h (first experiment); (b) 24 h (second experiment). Shown is the mean emission value at 590 nm ⁇ standard deviation of duplicate cultures.
  • Figure 5 Induction of nitric oxide (NO) production in mice treated with extracts of Lactobacillus fermentum I-3929 and Lactobacillus rhamnosus 71.38 in (a) the first assay, and (b) the second assay. Results are expressed in ⁇ M nitric oxide (NO) as mean value ⁇ standard deviation.
  • PBS phosphate-buffered saline
  • Extract means material obtained following lysis of one or more bacterial strains. In some cases, the extract is obtained from only one strain while in others the extract is obtained from a mixture of several different strains.
  • the extract is a soluble extract, meaning that it does not contain significant amounts of particulate and insoluble materials, such as particulate or solid cell wall fragments. Instead, components from cell walls, organelles, and cell membranes may be comprised in the extracts to the extent that they are dissolved or suspended. For example, the extract may be treated to remove particulate and insoluble materials, such as via filtration, centrifugation, or another separation technique.
  • Chemical lysis This is a method of lysing bacterial cells under basic, acidic, and/or osmotic conditions.
  • Lysate As used herein, this term means an extract of bacteria obtained from a cell lysis procedure.
  • a filtration process means a passage of an extract or a mixture of extracts, through one or more filters such as microfilters (i.e., microfiltration) and/or ultrafilters (i.e., ultrafiltration). Such filtration may not necessarily remove 100% of the components it is designed to remove, but may, in some embodiments, render the extracts substantially free of those components. In some cases, filtration is repeated in several passes or cycles.
  • filters such as microfilters (i.e., microfiltration) and/or ultrafilters (i.e., ultrafiltration).
  • Such filtration may not necessarily remove 100% of the components it is designed to remove, but may, in some embodiments, render the extracts substantially free of those components. In some cases, filtration is repeated in several passes or cycles.
  • Initial pH That term means the pH measured at the start of a procedure, such as bacterial lysis or filtration.
  • Saccharides A saccharide, as defined herein, includes monosaccharides, disaccharides, as well as larger saccharides such as linear and branched polysaccharides. Saccharides also include substituted or chemically modified saccharides, such as lipopolysaccharides (LPS) and their chemically modified variants.
  • LPS lipopolysaccharides
  • Lipoproteins This term refers to macromolecules that comprise both protein or peptide chains and lipids, for example, a protein or peptide covalently bound to a lipid. Lipoprotein, as used herein, also includes lipopeptides.
  • Peptidoglycans This term refers to polymers comprising sugars and amino acids.
  • Lipoteichoic acid This term refers to a surface-associated adhesion amphiphilic molecule present in gram-positive bacterial strains.
  • Teichoic acid This term refers to polymers of glycerol phosphate or ribitol phosphate linked together via phosphodiester bonds.
  • D-amino acids This term refers to amino acids that exist in dextra-rotatory isomeric forms, as opposed to biosynthetically produced L-amino acids, which exist in levo-rotatory isomeric forms.
  • Racemization This term indicates at least partial chemical modification of L-amino acids to D-amino acids.
  • Medium that avoids the risk of prion-based diseases means a culture medium used at any stage of the preparation of the extracts that does not comprise materials such as serum or meat extracts taken from animals such as cows or sheep, or from any other animal that can transmit prion-based diseases.
  • examples of such media include vegetable-based or synthetic media and also media using horse serum or media comprising materials taken from animal species that do not transmit prion diseases.
  • Examples of prion-based diseases include, for example, mad cow disease, scrapie, and Creutzfeld-Jacob disease.
  • a non-animal medium is a medium that does not include components derived from animals. Examples include a vegetable-based (i.e. vegetal) medium, such as a soya medium, and a synthetic medium.
  • Nutraceutical as used herein, means any composition that may have healthful effects in a subject upon administration wherein the composition is, for example, available to a subject without a doctor's prescription.
  • Treatment means both treatment of current diseases or disorders as well as prevention of or protection from the development of new diseases or disorders, for example.
  • Adjuvant as used herein to refer to embodiments of the invention, refers to an embodiment of the invention provided to a subject in conjunction with a medical treatment plan.
  • Immunomodulation, immunomodulatory, and like terms, as used herein, refer to the ability to modify the immune responses in a subject in a manner that may have healthful benefits, such as to produce an anti-inflammatory or immunostimulatory effect.
  • Anti-inflammatory, and like terms, as used herein, refer to immunomodulatory effects serving to reduce inflammation.
  • Immunostimulatory and like terms refer to the stimulation of the immune system.
  • Protective immunity means that an embodiment is provided to a subject so as to provide protection from subsequent challenge with an infective agent or allergen.
  • the level of infective agent or allergen in the subject is sufficiently low in concentration so as to not significantly compromise the subject's health.
  • the length of time in which such protection from challenge is effective may be limited, such as for a period of hours, days, or weeks.
  • Subject means any animal subject, including mammalian subjects, such as humans and domestic animals.
  • domestic animals for example, may include mammals such as dogs, cats, horses, pigs, cows, sheep, goats, or other livestock, and also may include non-mammals such as birds, for example chickens, ducks, geese, turkeys and other livestock birds.
  • the specific bacterial strains identified herein and used in the invention may include the strain obtained from the original deposit recited herein or a genetic clone thereof, including a strain that has been re- deposited at a later time with a different deposit code name, but which is considered to be genetically the same strain as the originally deposited version.
  • the present invention includes an extract of one or more
  • Lactobacillus bacterial strains wherein the extract is a soluble extract, and wherein the extract comprises chemically modified bacterial molecules.
  • the extracts of the present invention may be prepared, for example, by culturing of cells followed by harvesting the resulting biomass, lysis and purification. For each strain, to obtain a sufficient amount of material, the fermentation cultures may be started from a working seed lot followed by inoculation into larger fermentation containers.
  • the media used may be the same for each species.
  • a medium that avoids the risk of prion-based diseases may be used for growing all strains to be used.
  • the resulting biomass from one strain or from a set of strains may be inactivated by a heat treatment, concentrated, and frozen.
  • the starting material used to form the extracts in some embodiments, may be un-lysed whole cells.
  • the starting material used to prepare the extracts may be biomass obtained from cells already at least partially lysed mechanically, enzymatically, or chemically.
  • the starting material may be a fraction of such previously lysed cells, such as a cell wall- containing fraction.
  • the starting material is treated with an alkaline medium, such as from a strong base, such as hydroxide, or other strong mineral or organic bases.
  • a strong base such as hydroxide, or other strong mineral or organic bases.
  • un-lysed cells in the starting material are lysed while, in some embodiments, cellular components may be chemically modified.
  • the chemically modified bacterial molecules are obtained by base treatment, such as strong base treatment of the one or more Lactobacillus bacterial strains from which the extract is obtained (i.e. base treatment of un-lysed cells or components or fractions from bacterial cells, as just explained).
  • a biomass dry weight concentration of 2 to 90 g/L of may be subjected to base treatment, such as from about 2 to about 80 g/L, or from about 3 to about 40 g/L, such as 3, 5, 10, 15, 20, 25, 30, 35, or 40 g/L, or even about 5 to 50 g/L or other ranges bounded by the concentrations listed above.
  • base treatment such as from about 2 to about 80 g/L, or from about 3 to about 40 g/L, such as 3, 5, 10, 15, 20, 25, 30, 35, or 40 g/L, or even about 5 to 50 g/L or other ranges bounded by the concentrations listed above.
  • about 40 to about 80 g/L is subjected to base treatment, such as 40, 50, 60, 70, or 80 g/L or ranges bounded by the concentrations listed above.
  • the biomass dry weight is defined here by the dry weight of material in g per liter of sample. It may be measured by drying the sample in a small porcelain dish at about 105 0 C until it reaches a constant mass.
  • the temperature may be from 30 to 60°C, such as from 30 to
  • the base treatment temperature may be from 35 to 60 0 C, such as 35 to 55°C, 35 to 50 0 C, 35 to 45°C, or 35 to 40 0 C, for example.
  • the base treatment temperature may be 31 0 C, 32°C, 33°C, 34°C, 35°C, 36 0 C, 37°C, 38°C, 39°C, or even 40 0 C, or ranges bounded by the temperatures listed above.
  • the time of the base treatment may vary from 2 hours to several days, such as 1 , 2, 3, 4, 5 or even 10 days, or from 3 to 120 hours, or from 3 to 48 hours, such as 3, 5, 8, 15, 14, 16, 18, 20, 22, 24, 26, 28, 30, 36, 40, 44, or 48 hours, or from 15 to 120 hours, such as 60 to 120 hours, such as 60, 72, 84, 96, 108, or 120 hours, or ranges bounded by the times listed above. It is understood that these ranges of time include any fractional number of days, hours, or minutes, therein.
  • 1.0 N is used, such as, from 0.001 N to 0.6 N, or from 0.10 N to 0.8 N, or from 0.6 N to 1.0 N, or a range starting or ending from 0.001 , 0.002, 0.003, or 0.1 N, or from 0.1 N to 0.6 N, or a range starting or ending from 0.6, 0.7, 0.8, 0.9, 1.0, or 1.0 N, or other ranges bounded by the concentrations listed above.
  • a base concentration is used so as to achieve a initial pH of greater than 9.0, or a pH of greater than 9.5, a pH greater than 10.0 and less than 13.5, such as greater than 11.5, greater than 12.0, greater than 12.5, greater than 13.0, or from pH 9.0 to pH 13.5.
  • a base concentration may be used so as to achieve an initial pH greater than 10.0 and less than 13.0, or from pH 9.0 to pH 13.0, for example.
  • the pH during the base treatment may be decreased upon the extraction of soluble components.
  • the initial pH may be a basic pH, such as pH 9.0 to pH 13.0, or pH 9.5 to pH 12.5.
  • the base treatment may be allowed to proceed for a certain period of time, such as 3 to 120 hours, such as 3 to 48 hours, or for a period of time as listed above, at a temperature as listed above.
  • the pH may optionally be rendered acidic by the addition of, for instance, hydrochloric acid, so as to obtain a pH between 2.0 and 4.5, or a pH between 2.5 and 4.5, or a pH between 2.5 and 4.0, such as 2.5, 3.0, 3.5, 4.0, or a range bounded by any of the pH's listed above.
  • the second treatment at low pH may be carried out at a temperature from 30 to 60°C, 35 to 55, or 35 to 45°C, such as 35°C, 36°C, 37°C, 38°C, 39°C, 40°C, 41 °C, 42°C, 43°C, 44°C, or even 45°C.
  • the time of acidic treatment may vary from 1 hour to several hours up to 72 hours, for example between 1 hour and 24 hours, or between 1 hour and 6 hours, or between 3 hours and 48 hours, or between 3 hours and 24 hours, or between 4 and 72 hours, or even between 24 hours and 72 hours, or any range of time bounded by the times listed above.
  • an alkaline treatment is performed on a bacterial biomass comprising, for example, material from Lactobacillus fermentum, having a biomass dry weight between 10 g /L to 40 g/L.
  • the alkaline treatment is performed on a bacterial biomass comprising a mixture of Lactobacillus strains and having a biomass dry weight between 10 g/L and 40 g/L.
  • the alkaline treatment may be performed at a hydroxide ion concentration between 0.025 N and 0.25 N, or at a pH between 9.5 and 12.5 at a temperature of 35 to 45°C for a time of between 3 hours and 48 hours.
  • the alkaline treatment is performed on bacterial biomass comprising material from one or more Lactobacillus strains at a hydroxoide ion concentration between 0.025 N and 0.20 N, between 0.025 and 0.15 N, between 0.025 and 0.10 N, between 0.05 N and 0.25 N, between 0.05 N and 0.20 N, between 0.05 and 0.15 N, between 0.05 N and 0.10 N, between 0.10 N and 0.25 N, between 0.10 N and 0.20 N, between 0.10 N and 0.15 N, between 0.15 N and 0.25 N, between 0.15 N and 0.20 N, or even between 0.20 N and 0.25 N.
  • Such embodiments may, for example, have a pH between 9.5 and 12.0, between 9.5 and 11.5, between 9.5 and 11.0, between 9.5 and 10.5, between 9.5 and 10.0, between 10.0 and 12.5, between 10.0 and 12.0, between 10.0 and 11.5, between 10.0 and 11.0, between 10.0 and 10.5, between 10.5 and 12.5, between 10.5 and 12.0, between 10.5 and 11.5, between 10.5 and 11.0, between 11.0 and 12.5, between 11.0 and 12.0, between 11.0 and 11.5, between 11.5 and 12.5, between 11.5 and 12.0, or even a pH between 12.0 and 12.5.
  • the time of alkaline treatment for such embodiments may be between 3 hours and 36 hours, between between 3 hours and 24 hours, between 3 hours and 18 hours, between 3 hours and 12 hours, between 3 hours and 6 hours, between 6 hours and 48 hours, between 6 hours and 36 hours, between 6 hours and 24 hours, between 6 hours and 18 hours, between 6 hours and 12 hours, between 6 hours and 8 hours, between 8 hours and 48 hours, between 8 hours and 36 hours, between 8 hours and 24 hours, between 8 hours and 18 hours, between 8 hours and 12 hours, between 12 hours and 48 hours, between 12 hours and 36 hours, between 12 hours and 18 hours, between 18 hours and 48 hours, between 18 hours and 36 hours, between 18 hours and 24 hours, between 24 hours and 48 hours, between 24 hours and 36 hours, or between 36 hours and 48 hours.
  • the alkaline treatment may be performed for any of the time periods bounding the ranges above, for example, 3, 6, 8, 12, 18, 24, 36, or even 48 hours. Such conditions may provide for a moderate alkaline treatments [061 ]
  • 10 g/L and 40 g/L of biomass dry weight from one or more Lactobacillus strains may be subjected to a hydroxide ion concentration between 0.15 N and 0.50 N or a pH between 11.5 and 13.5, at a temperature of 35 to 45°C for a time of between 15 hours and 120 hours.
  • the hydroxide concentration may be between 0.15 N and 0.45 N, between 0.15 N and 0.40 N, between 0.15 N and 0.35 N, between 0.15 N and 0.30 N, between 0.15 N and 0.25 N, between 0.15 N and 0.20 N, between 0.20 N and 0.50 N, between 0.20 N and 0.40 N, between 0.20 N and 0.30 N, between 0.25 N and 0.50 N, between 0.30 N and 0.50 N, between 0.30 N and 0.40 N, or between 0.40 N and 0.50.
  • Such embodiment may have a pH between 11.5 and 13.0, between 1 1.5 and 12.5, between 11.5 and 12.0, between 12.0 and 13.5, between 12.0 and 13.0, between 12.0 and 12.5, between 12.5 and 13.5, between 12.5 and 13.0, between 13.0 and 13.5, for example.
  • the time period for alkaline treatment may be between 15 hours and 100 hours, between 15 hours and 90 hours, between 15 hours and 75 hours, between 15 hours and 60 hours, between 15 hours and 48 hours, between 15 hours and 36 hours, between 24 hours and 120 hours, between 24 hours and 100 hours, between 24 hours and 90 hours, between 24 hours and 75 hours, between 24 hours and 60 hours, between 24 hours and 48 hours, between 36 hours and 120 hours, between 36 hours and 100 hours, between 36 hours and 90 hours, between 36 hours and 75 hours, between 36 hours and 60 hours, between 36 hours and 48 hours, between 48 hours and 120 hours, between 48 hours and 100 hours, between 48 hours and 90 hours, between 48 hours and 75 hours, between 48 hours and 60 hours, between 60 hours and 120 hours, between 60 hours and 100 hours, between 60 hours and 90 hours, between 60 hours and 75 hours, between 75 hours and 120 hours, between 75 hours and 100 hours, between 75 hours and 90 hours, between 90 hours and 120 hours, or between 100 and 120 hours, for example. Time periods also contemplated for alkaline treatment in such embodiments include 15,
  • between 10 g/L and 40 g/L of starting biomass dry weight may be treated with a hydroxide concentration between 0.025 N and 0.25 N or a pH between 9.5 and 12.5, at a temperature of 35 to 45°C for a time of between 3 hours and 48 hours.
  • the pH may then be adjusted to between 2.5 and 4.0 by the addition of acid, such as hydrochloric acid (HCI), comprising an acidic treatment.
  • HCI hydrochloric acid
  • the acidic treatment may be performed at a temperature of between 35 and 45°C for a time of between 1 hour and 24 hours.
  • the alkaline treatment of bacterial biomass comprising one or more Lactobacillus strains may be performed with a hydroxide concentration between 0.025 N and 0.20 N, between 0.025 and 0.15 N, between 0.025 and 0.10 N, between 0.05 N and 0.25 N, between 0.05 N and 0.20 N, between 0.05 and 0.15 N, between 0.05 N and 0.10 N, between 0.10 N and 0.25 N, between 0.10 N and 0.20 N, between 0.10 N and 0.15 N, between 0.15 N and 0.25 N, between 0.15 N and 0.20 N, or even between 0.20 N and 0.25 N.
  • such embodiments may, for example, have a pH between 9.5 and 12.0, between 9.5 and 11.5, between 9.5 and 11.0, between 9.5 and 10.5, between 9.5 and 10.0, between 10.0 and 12.5, between 10.0 and 12.0, between 10.0 and 11.5, between 10.0 and 11.0, between 10.0 and 10.5, between 10.5 and 12.5, between 10.5 and 12.0, between 10.5 and 11.5, between 10.5 and 11.0, between 11.0 and 12.5, between 11.0 and 12.0, between 11.0 and 11.5, between 11.5 and 12.5, between 11.5 and 12.0, or even a pH between 12.0 and 12.5.
  • the time of alkaline treatment for such embodiments may be between 3 hours and 36 hours, between between 3 hours and 24 hours, between 3 hours and 18 hours, between 3 hours and 12 hours, between 3 hours and 6 hours, between 6 hours and " 48 hours, between 6 hours and 36 hours, between 6 hours and 24 hours, between 6 hours and 18 hours, between 6 hours and 12 hours, between 6 hours and 8 hours, between 8 hours and 48 hours, between 8 hours and 36 hours, between 8 hours and 24 hours, between 8 hours and 18 hours, between 8 hours and 12 hours, between 12 hours and 48 hours, between 12 hours and 36 hours, between 12 hours and 18 hours, between 18 hours and 48 hours, between 18 hours and 36 hours, between 18 hours and 24 hours, between 24 hours and 48 hours, between 24 hours and 36 hours, or between 36 hours and 48 hours.
  • the alkaline treatment may be performed for any of the time periods bounding the ranges above, for example, 3, 6, 8, 12, 18, 24, 36, or even 48 hours.
  • the pH may then be adjusted to between 2.5 and 3.5, between 2.5 and 3.0, between 3.0 and 4.0, between 3.0 and 3.5, or between 3.5 and 4.0 by the addition of acid, for the acidic treatment following the alkaline lysis.
  • the acidic treatment may be performed for a time of between 1 hour and 18 hours, between 1 hour and 12 hours, between 1 hour and 6 hours, between 1 hour and 3 hours, between 3 hours and 24 hours, between 3 hours and 18 hours, between 3 hours and 12 hours, between 3 hours and 6 hours, between 6 hours and 24 hours, between 6 hours and 18 hours, between 6 hours and 12 hours, between 12 hours and 24 hours, between 12 hours and 18 hours, between 18 hours and 24 hours. Times also contemplated for the acidic treatment include 1 , 3, 6, 12, 18, and 24 hours.
  • the lysates obtained following the base treatment described above may next be purified by centrifugation and/or filtration, for example, to remove particulate and insoluble components.
  • lysates may be centrif uged at 9000 x gravity, followed by one or more rounds of filtration on a 0.2 micron filter. In some cases, successive rounds of filtration on larger pore filters followed by filtration on a 0.2 micron filter may be used.
  • Ultrafiltration methods may also be employed in order to help extract soluble materials from the extract, for example, recirculating the ultrafiltration permeate for further microfiltration.
  • a tangential flow filtration (TFF) method may be used to filter the lysates and to extract soluble molecules from larger cellular debris ( Figure 2). See, e.g., Separations Technology, Pharmaceutical and Biotechnology Applications, Wayne P. Olson, Editor, lnterpharm Press, Inc., Buffalo Grove, IL, U.S.A., p.126 to 135 - ISBN:0-935184-72-4.
  • a diluted bacterial lysate may be stored in a first tank.
  • the extract may be exposed to both a microfilter and an ultrafilter.
  • MF microfiltration
  • an ultrafiltration (UF) loop is started.
  • the UF permeate may be recirculated back to the first tank for continuous extraction of solubilized extracts from the lysate while the UF retentate is stored in the second tank.
  • the volumes in tanks 1 and 2 may be adjusted by regulation of flow rates of the microfiltration and ultrafiltration permeates.
  • the ultrafiltration loop may be shut down and the microfiltration loop may be run alone and the MF permeate transferred to tank 2.
  • Conditions of crossflow and transmembrane pressure are defined by the pressure independent and mass transfer controlled regions in film theory described, for example, by M.Cheryan (Ultrafiltration and Microfiltration Handbook, 2 nd Ed., Ch. 4, 1998).
  • the permeate flux and extraction yield are affected by filtration conditions (trans-membrane pressure (TMP), crossflow, temperature, etc.).
  • TMP trans-membrane pressure
  • the type of filter can also affect the filtration performance, as well as the type of plate system (cassette filter). Different configurations, including parallel mode and serpentine mode can be used (see Figure 2). Specific conditions are developed for optimized performances for each combination of type of mode and type of filter used.
  • the microfiltration loop may be fitted with filters of 1.2 microns to
  • the cross-flow may be between 100 and 3000 Liters/ hours m 2 (LHM), such as between 300 and 2500 LHM, or 2000 LHM with a TMP of 0.3 to 2 bar.
  • the ultrafiltration loop may be fitted with filters of from 10 KDa to 1000 KDa, such as from 10 KDa to 100 KDa, or from 10 KDa to 30 KDa, or from 30 KDa to 100 KDa.
  • the cross-flow may be between 30 and 1000 LHM, such as between 20 and 500 LHM with a TMP of 0.2 to 1.5 bar.
  • diafiltration volumes may be used to extract soluble components from bacterial cell walls.
  • between 8 and 15 volumes are used.
  • between 5 and 15 cycles of filtration may be used, in some cases between 8 and 15 cycles.
  • the extract may be further concentrated or centrifuged, if desired. For instance, a further microfiltration using a smaller pore filter may be performed, such as a 0.2 micron filter. Following filtration, the extract may be lyophilized prior to formulating it for use.
  • the extract can be purified in order to separate, to eliminate or increase the concentration of one or more modified components in the extract.
  • a strong ionic chromatography step can be used in order to remove charged components.
  • Other purification processes can be used, such as gel filtration, chromatography, ultracentrifugation, extraction and precipitation.
  • Base treatment may result in a variety of chemical modifications to cellular components.
  • proteins (1) peptide bonds may undergo partial cleavage generating smaller polypeptides; (2) natural L-amino acids may be at least partially racemized to D-amino acids; and (3) asparagine and glutamine residues may become deaminated, leading to changes in the protein isoelectric point.
  • Molecules such as lipoteichoic acids, lipopeptides, and phospholipids may undergo a base-catalyzed hydrolysis of ester bonds and/or amide bonds leading to modified amphiphilic structures which may have new physicochemical and immunological properties.
  • RNA ribonucleic acid
  • Examples of other possible chemical modifications include partial solubilization of cell wall polysaccharides and complete hydrolysis of ribonucleic acid (RNA) into individual ribonucleotides, including rearrangement of phosphate groups.
  • RNA ribonucleic acid
  • Such molecular modifications may affect the biological activities of the extracts.
  • base treatment of bacteria according to the present invention may result in partial hydrolysis of proteins as well as deamination, deamidation, and/or partial racemization of amino acids from L to D.
  • peaks representing D- aspartic acid, D-glutamic acid, D-serine, D-methionine, D-histidine, D-alanine, D- arginine, D-phenylalanine, D-tyrosine, D-leucine, and D-lysine were each observed.
  • the percentage of D-amino acids of those species in that study ranged from 3% to 40%.
  • some embodiments of the invention allow for racemization of one or more of serine, threonine, histidine, alanine, arginine, tyrosine, phenylalanine, leucine, and lysine, such as all of the above amino acids, or any selection of more than one but less than all of the above amino acids, such as, for example, alanine, phenylalanine and lysine.
  • at least 10% of one or more of the above amino acids may become racemized from D to L.
  • at least 40% of one or more of the above amino acids may become racemized.
  • extracts of the present invention may comprise between 1 and 90% D-amino acids, such as between 1 and 80%, or between 1 and 60%. In some embodiments, the extract comprises between 10 and 45% D-amino acids, such as between 25 and 35% D-amino acids.
  • the extracts of the invention may comprise at least one D-amino acid selected from the group consisting of D-aspartic acid, D- asparagine, D-glutamic acid, D-glutamine, D-serine, D-methionine, D-histidine, D- alanine, D-arginine, D-phenylalanine, D-tyrosine, D-leucine, D-lysine, D-valine, and D-threonine.
  • the concentration of any one D-amino acid comprises between 1 and 50 %, such as between 10 and 40 %, or even between 15 and 35 %.
  • Some extracts of the present invention comprise Lactobacillus bacterial cell wall and membrane components, such as lipoteichoic acids, teichoic acid, peptidoglycan, or a combination thereof. In some embodiments, those components are chemically modified. Some extracts also comprise cell wall and/or cell membrane components such as lipoproteins, which may also be chemically modified. In some embodiments, the cell wall components or cell membrane components, for example lipoproteins, are dissolved or suspended in the extracts and thus are not present in particulate or insoluble form.
  • an extract according to the present invention may comprise, for example, from 10 to 100 mg/mL soluble dry weight (SDW) of material, 1 to 30 mg/mL of protein (Prot.), 0.5 to 4.0 mg/mL of sugar and less then 100 ⁇ g/mL DNA.
  • SDW soluble dry weight
  • Prot. protein
  • some embodiments contain about 15 to 35 mg/mL of soluble dry weight, 3 to 7 mg/mL of protein, 1.0 to 3.0 mg/mL of sugar and 10 to 40 ⁇ g/mL of DNA.
  • An extract according to the present invention may contain, for example, 30 mg/mL of soluble dry weight, 9.6 mg/mL protein, 2.4 mg/mL of sugar and 33 ⁇ g/mL of DNA or another example containing 32.4 mg/mL of soluble dry weight, 5.8 mg/mL protein, 2.3 mg/mL of sugar and less than 100 ⁇ g/mL of DNA.
  • Soluble Dry Weight (SDW) in g/L or mg/mL is determined by obtaining 5 mL of the soluble fraction resulting from the lysis or base treatment and drying it to a constant mass in a porcelain dish at 105°C.
  • the extracts comprise at least 0.3 mg/mL of saccharides, such as between 0.3 and 4.5 mg/mL of saccharides.
  • at least one saccharide is chosen from monosaccharides, disaccharides, and polysaccharides.
  • Some extracts of the invention comprise at least one branched polysaccharide.
  • at least one saccharide is chemically modified.
  • Lysis or base treatment of bacteria may result in a decrease of the average molecular weight of component macromolecules to a range of, for instance, I kDa to between 300 kDa and 100 kDa, or to a range of 1 kDa to between 60 kDa and 10 kDa.
  • the extract comprises at least one protein with a molecular weight of less than 50 kDa or less than 30 kDa, such as less than 10 kDa.
  • Extracts according to the invention may have immunomodulatory activities. For example, some extracts may stimulate the immune system. Some extracts may have anti-inflammatory activities. The specific effects of an extract may depend upon the manufacturing conditions and the species or strain of Lactobacillus, or mixture of species or strains, from which the extract is obtained. Accordingly, some extracts according to the invention may show potent immunostimulatory activity, and may thus be useful in treating infections, or as adjuncts to such treatment, while other embodiments may show weaker immunostimulatory activity but show anti-inflammatory activity, thus being useful in treating inflammatory disorders such as allergies, asthma, autoimmune diseases, colitis, and inflammatory bowel diseases, or as adjuncts to such treatment.
  • inflammatory disorders such as allergies, asthma, autoimmune diseases, colitis, and inflammatory bowel diseases, or as adjuncts to such treatment.
  • extracts according to the invention may be effective to treat patients suffering from disorders including, but not limited to, microbial infections, allergic diseases, and digestive tract disorders.
  • Some extracts according to the invention may also be provided to a patient as nutraceuticals, for example, as adjuvants in the treatment of a variety of conditions including, but not limited to, microbial infections, allergic diseases, and digestive tract disorders.
  • the range of the biological activiies of the extracts may be determined by several in vitro and in vivo assays.
  • the AlamarBlueTM-Assay incorporates a fluorometric/colorimetric growth indicator based on the detection of metabolic activity by an oxidation-reduction (REDOX) indicator in response to chemical reduction resulting from cell growth (Example 4).
  • REDOX oxidation-reduction
  • nitric oxide nitric oxide
  • the extracts may stimulate NO production in murine macrophages, leading to measured NO concentrations ranging from 3 ⁇ M to 60 ⁇ M, such as 5 ⁇ M to 40 ⁇ M.
  • the NO concentration may be above 30 ⁇ M.
  • the type of bacterial species may also affect these results. For example, extracts from Lactobacillus fermentum may induce a larger nitric oxide production than Lactobacillus rahmnosus (see, e.g. Example 5 below).
  • tests of the bacterial extracts of the invention may be performed on human peripheral blood mononuclear cells (PBMC).
  • PBMC peripheral blood mononuclear cells
  • the release of both IL12p70 (an inflammatory cytokine) and IL10 (an anti-inflammatory cytokine) may be measured, and the IL-10/IL-12 ratio calculated (Example 6).
  • the invention includes extracts capable of achieving a calculable IL10/IL12 ratio in human peripheral blood mononuclear cells, wherein the ratio is equal to or greater than the IL10/IL12 ratio achieved by a live Lactobacillus strain from which the extract is obtained.
  • the immune responses of the extracts of the invention may also be tested by examining their effects on Toll-like receptors (TLRs),
  • TLRs Toll-like receptors
  • extracts may be tested in HEK293 cells in the presence or absence of the TLR2 agonist Pam3Cys, or in the presence or absence of the TLR4 agonist LPS (Example 7).
  • the HEK293 cell line enables efficient monitoring of TLR activity using ELISA analyses such as IL-8 titration or reporter-based systems that monitor TLR-induced NF- ⁇ B activation.
  • Some embodiments of the invention may act as TLR 2/6 antagonists in HEK TLR 2/6 cells. Thus some embodiments may be useful to fight infections in a subject, whereas other embodiments may be developed for use against inflammation and/or autoimmunity disorders.
  • the extracts presently disclosed may also be screened for TLR and NOD2 receptor activity (Example 8). Some embodiments of the invention activate TLR and or NOD2 receptors in vitro, indicating that they may be able to activate the immune system, via TLRs and/or NOD2.
  • the plaque forming cells (PFC) technique may be employed to evaluate a non-specific stimulation of B-lymphocytes (Example 9). Certain lymphoid cells release hemolytic antibodies which diffuse and cause the lysis of the neighboring red blood cells by forming a lysis plaque in the presence of complement. Some embodiments of the invention may increase the secretion of immunoglogulins by B cells, and thus can potentially be used prophylactically for priming the immune system in subjects suffering from recurrent infections.
  • the anti-infective efficacy of the extracts of the instant invention may be tested, for example upon Salmonella infection of subjects, such infection of mice (Example 10).
  • Some embodiments of the invention may provide protective immunity against infections, such as bacterial infections, i.e. Salmonella infections.
  • some embodiments may decrease the mortality induced in mice by the injection of Salmonella thyphimurium.
  • the extract decreases the eosinophil cell number, neutrophil cell number, lymphocyte cell number, or any combination thereof, in an asthmatic murine subject by a factor of at least 1.5 with respect to an asthmatic non-treated control.
  • Some embodiments of the invention may decrease eosinophilia in asthmatic subjects, and concomitantly decrease the level of Th2 cytokines (such as IL4, IL5, IL13), which are markers of asthma.
  • Th2 cytokines such as IL4, IL5, IL13
  • such embodiments may have anti-inflammatory activities in subjects suffering from immunological disorders, such as allergic disorders, including asthma.
  • compositions Comprising the Bacterial Extracts
  • Extracts according to the invention may be formulated in a number of different ways for eventual administration.
  • oral tablets, capsules, pills may be prepared, as well as liquid formulations or aerosols.
  • Formulations for infusion or injection may also be prepared.
  • Embodiments of this invention can be formulated, for example, as solid dosage forms or liquid dosage forms.
  • Exemplary solid dosage forms may include, for example, a tablet, e.g. coated tablet, chewable tablet, effervescent tablet, sublingual tablet, granulates, powder, or a capsule) containing the extract.
  • Solid dosage forms may also contain diluents, fillers, and/or other excipients. Other excipient components may be added such as preservatives, colorants, flavourings, and sweeteners.
  • excipients such as preservatives, colorants, flavourings, and sweeteners.
  • the extracts of the present invention may be included in one or more nutraceutical compositions, such as nutritional and/or dietary supplements and food additives, or one or more pharmaceutical compositions.
  • a dose comprising at least one extract of the present invention may be administered to a subject suffering from or at risk of developing at least one disorder chosen from digestive tract disorder, respiratory tract disorder, urinary tract disorder, and allergic conditions.
  • the extracts may be administered to subjects suffering from, or at risk of developing, pulmonary upper an lower infections, obstructive pulmonary disease with acute lower respiratory infection, obstructive pulmonary disease with acute exacerbation, nasopharyngitis, sinusitis, pharyngitis, tonsillitis, laryngitis, tracheitis, laryngopharyngitis, influenza, pneumonia, bronchopneumonia, bronchitis, rhinitis, nasopharyngitis, pharyngitis, sinusitis, tonsillitis, laryngitis, laryngotracheitis, bronchitis, allergic rhinitis, allergic asthma, atopic dermatitis, urinary tract infections due to o
  • the extracts are administered to a subject in the form of a nutraceutical composition such as a nutritional supplement and/or food additive.
  • the extracts are administered to a subject in the form of a pharmaceutical composition.
  • the administration may comprise a single dose or multiple dose administration.
  • an extract may be provided in a therapeutically effective dose to treat a subject suffering from one or more of the conditions above. In some embodiments, an extract may be provided as an adjunct to other medical treatment.
  • Example 1 Bacterial Cultures
  • Example 1.1 Culture of Lactobacillus fermentum I-3929
  • Culture media was prepared by dissolving in purified water the following components: Sodium chloride: 3 g/L; Sodium monohydrogen phosphate: 2 g/L; Sodium acetate: 1 g/L; Soya peptone 50 g/L; Glucose: 12 g/L; Calcium chloride: 0.1 g/L; Potassium chloride: 0.1 g/L; Sodium bicarbonate: 0.5 g/L; pyruvate: 0.1 g/L; Glutamate: 0.2 g/L; Metal solution (copper sulfate: 3 mg/l; iron chloride: 830 mg/l; zinc sulfate: 860 mg/l; sulfuric acid: 1.1 mg/L): 0.5 ml_/L.
  • Polypropylene glycol (0.02 mL/L, density 1.005 g/mL) was then added. After dissolution, the pH was not adjusted. After sterilizing the media, small Erlenmeyer flasks were individually inoculated with the content of frozen vials (containing 1.5 mL of frozen bacteria) and incubated at 37°C for 8 hours. Then 20 ml aliquots of this culture were transferred to larger Erlenmeyer flasks containing 1000 mL of culture media, and incubated again in the same conditions. After 16 hours growth, the content of the 1 liter Erlenmeyer flask was transferred to the prefermenter.
  • Example 1.2 Culture of Lactobacillus helveticus 103146
  • Culture media was prepared by dissolving in purified water the following components: Sodium chloride: 3 g/L; Sodium monohydrogen phosphate: 2 g/L; Sodium acetate: 1 g/L; Soya peptone 50 g/L; Glucose: 12 g/L; Calcium chloride: 0.1 g/L; Potassium chloride: 0.1 g/L; Sodium bicarbonate: 0.5 g/L; pyruvate: 0.1 g/L; Glutamate: 0.2 g/L; Metal solution (copper sulfate: 3 mg/l; iron chloride: 830 mg/l; zinc sulfate: 860 mg/l; sulfuric acid: 1.1 mg/L): 0.5 ml_/L.
  • Polypropylene glycol (0.02 mL/L) was then added. After dissolution, the pH was not adjusted. After sterilizing the media, small Erlenmeyer flasks were individually inoculated with the content of frozen vials (containing 1.5 mL of frozen bacteria) and incubated at 37°C for 9 hours. Then 20 ml aliquots of this culture were transferred to larger Erlenmeyer flasks containing 1000 mL of culture media, and incubated again under the same conditions. After 15 hours, growth, the content of the 1 liter Erlenmeyer flask was transferred to the prefermenter.
  • Polypropylene glycol (0.02 ml_/L) was then added. After dissolution, the pH was not adjusted. Incubation temperature was regulated at 37°C, with 100 rpm stirring and no aeration. The pH was not regulated during the culture. After 9 hours, 7 liters from the prefermenters were transferred to a fermenter. (OD at 700 nm prefermenter culture after 9 hours: 0.14). The cultures of the prefermenters were transferred under sterile conditions into fermenters.
  • 70 liters of culture media were prepared by dissolving in purified water the following components: Sodium chloride: 3 g/L; Sodium monohydrogen phosphate: 2 g/L; Sodium acetate: 1 g/L; Soya peptone 50 g/L; Glucose: 12 g/L; Calcium chloride: 0.1 g/L; Potassium chloride: 0.1 g/L; Sodium bicarbonate: 0.5 g/L; pyruvate: 0.1 g/L; Glutamate: 0.2 g/L; Metal solution (copper sulfate: 3 mg/l; iron chloride: 830 mg/l; zinc sulfate: 860 mg/l; sulfuric acid: 1.1 mg/L): 0.5 ml_/L. Polypropylene glycol (0.02 ml_/L) was then added. After dissolution, the pH was not adjusted.
  • Example 1.3 Culture of Lactobacillus plantarum 71.39
  • Culture media was prepared by dissolving in purified water the following components: Sodium chloride: 3 g/L; Sodium monohydrogen phosphate: 2 g/L; Sodium acetate: 1 g/L; Soya peptone 50 g/L; Glucose: 12 g/L; Calcium chloride: 0.1 g/L; Potassium chloride: 0.1 g/L; Sodium bicarbonate: 0.5 g/L; pyruvate: 0.1 g/L; Glutamate: 0.2 g/L; Metal solution (copper sulfate: 3 mg/l; iron chloride: 830 mg/l; zinc sulfate: 860 mg/l; sulfuric acid: 1.1 mg/L): 0.5 mL/L.
  • Polypropylene glycol (0.02 mL/L) was then added. After dissolution, the pH was not adjusted. After sterilizing the media, small Erlenmeyer flasks were individually inoculated with the content of frozen vials (containing 1.5 mL of frozen bacteria) and incubated at 35°C for 9 hours. Then 20 ml aliquots of this culture were transferred to larger Erlenmeyer flasks containing 1000 mL of culture media, and incubated again under the same conditions. After 15 hours growth, the content of the 1 liter Erlenmeyer flask was transferred to the prefemnenter.
  • 70 liters of culture media were prepared by dissolving in purified water the following components: Sodium chloride: 3 g/L; Sodium monohydrogen phosphate: 2 g/L; Sodium acetate: 1 g/L; Soya peptone 50 g/L; Glucose: 12 g/L; Calcium chloride: 0.1 g/L; Potassium chloride: 0.1 g/L; Sodium bicarbonate: 0.5 g/L; pyruvate: 0.1 g/L; Glutamate: 0.2 g/L; Metal solution (copper sulfate: 3 mg/l; iron chloride: 830 mg/l; zinc sulfate: 860 mg/l; sulfuric acid: 1.1 mg/L): 0.5 ml_/L. Polypropylene glycol (0.02 mL/L) was then added. After dissolution, the pH was not adjusted.
  • Example 1.4 Culture of Lactobacillus rhamnosus 71.38
  • Culture media was prepared by dissolving in purified water the following components: Sodium chloride: 3 g/L; Sodium monohydrogen phosphate: 2 g/L; Sodium acetate: 1 g/L; Soya peptone 50 g/L; Glucose: 12 g/L; Calcium chloride: 0.1 g/L; Potassium chloride: 0.1 g/L; Sodium bicarbonate: 0.5 g/L; pyruvate: 0.1 g/L; Glutamate: 0.2 g/L; Metal solution (copper sulfate: 3 mg/l; iron chloride: 830 mg/l; zinc sulfate: 860 mg/l; sulfuric acid: 1.1 mg/L): 0.5 mL/L.
  • Polypropylene glycol (0.02 mL/L) was then added. After dissolution, the pH was not adjusted. The incubation temperature was regulated at 35°C, with 100 rpm stirring and no aeration. The pH was not regulated during the culture. After 9 hours, 7 liters from the prefermenters were transferred to a fermenter. (OD at 700 nm prefermenter culture after 9 hours: 3.75). The cultures of the prefermenters were transferred under sterile conditions into fermenters.
  • 70 liters of culture media were prepared by dissolving in purified water the following components: Sodium chloride: 3 g/L; Sodium monohydrogen phosphate: 2 g/L; Sodium acetate: 1 g/L; Soya peptone 50 g/L; Glucose: 12 g/L; Calcium chloride: 0.1 g/L; Potassium chloride: 0.1 g/L; Sodium bicarbonate: 0.5 g/L; pyruvate: 0.1 g/L; Glutamate: 0.2 g/L; Metal solution (copper sulfate: 3 mg/l; iron chloride: 830 mg/l; zinc sulfate: 860 mg/l; sulfuric acid: 1.1 mg/L): 0.5 mLJL. Polypropylene glycol (0.02 ml_/L) was then added. After dissolution, the pH was not adjusted.
  • Example 1.5 Culture of Lactobacillus iohnsonii 103782
  • Culture media was prepared by dissolving in purified water the following components:.
  • Sodium chloride 3 g/L; Sodium monohydrogen phosphate: 2 g/L; Sodium acetate: 1 g/L; Soya peptone 50 g/L; Glucose: 12 g/L;
  • Calcium chloride 0.1 g/L; Potassium chloride: 0.1 g/L;
  • Sodium bicarbonate 0.5 g/L; pyruvate: 0.1 g/L; Glutamate : 0.2 g/L;
  • Metal solution (copper sulfate: 3 mg/l; iron chloride: 830 mg/l; zinc sulfate: 860 mg/l; sulfuric acid : 1.1 mg/L): 0.5 ml_/L.
  • Polypropylene glycol (0.02 mL/L) was then added. After dissolution, the pH was not adjusted. After sterilizing the media, small Erlenmeyer flasks were individually inoculated with the content of frozen vials (containing 1.5 mL of frozen bacteria) and incubated at 33°C for 10 hours. Then 20 ml aliquots of this culture were transferred to larger Erlenmeyer flasks containing 1000 mL of culture media, and incubated again in the same conditions. After 14 hours growth, the content of the 1 liter Erlenmeyer flask was transferred to the prefermenter.
  • Polypropylene glycol (0.02 ml_/L) was then added. After dissolution, the pH was not adjusted. Incubation temperature was regulated at 35°C, with 100 rpm stirring and no aeration. The pH of the culture was adjusted to 5.6 with acetic acid. After 24 hours, 7 liters from the prefermenters were transferred to a fermenter. (OD at. 700 nm prefermenter culture after 24 hours: 0.47). The cultures of the prefermenters were transferred under sterile conditions into fermenters.
  • Lactobacillus fermentum I-3929 biomass from Example 1.1 containing 6 g of bacterial dry weight was thawed to room temperature, then diluted with purified water to reach 12 g/L of bacterial biomass dry weight. Alkalinization was performed with 0.03 M sodium hydroxide. The pH measured at the beginning of the lysis was 10.3. Then the lysis was incubated for 6 hours at 40°C under continuous stirring. After the incubation, the pH was 9.9.
  • Lactobacillus rhamnosus 71.38 biomass from Example 1.4 containing 20 g of bacterial dry weight was thawed to room temperature, then diluted with purified water to reach 40 g/L of bacterial biomass dry weight. Alkalinization was performed with 0.03 M sodium hydroxide. The pH measured at the beginning of the lysis was 10.3. Then the lysis was incubated for 6 hours at 40 0 C under continuous stirring. After the incubation, the pH was 9.7.
  • Lactobacillus fermentum I-3929 biomass from Example 1.1 containing 6 g of bacterial dry weight was thawed to room temperature, then diluted with purified water to reach 12 g/L of bacterial biomass dry weight. Alkalinization was performed with 0.06 M sodium hydroxide. The pH measured at the beginning of the lysis was 12.3. Then the lysis was incubated for 6 hours at 40°C under continuous stirring. After the incubation, the pH was 11.8.
  • Lactobacillus fermentum I-3929 biomass from Example 1.1 containing 6 g of bacterial dry weight was thawed to room temperature, then diluted with 0.2 N NaCI solution to reach 12 g/L of bacterial biomass dry weight. The final concentration of NaCI solution was 0.15N.
  • the pH measured at the beginning of the lysis was 6.4. Then the lysis was incubated for 6 hours at 40°C under continuous stirring. After the incubation, the pH was 6.3.
  • Example 2.1 An aliquot of the lysate of Example 2.1 was taken to continue the lysis process. The volume was adjusted at pH 3.6 with HCI 25% and then incubated in a water-bath at 40 0 C for 1 hour under static conditions.
  • Example 2.4 An aliquot of the lysate of Example 2.4 was taken to continue the lysis process. The volume was adjusted at pH 12.4 with NaOH 10N and then incubated in a water-bath at 40 0 C for 1 hour under static conditions.
  • Lactobacillus johnsonii 103782 biomass from Example 1.5 containing 0.3 g of bacterial dry weight was thawed to room temperature then diluted with purified water to reach 6 g/L of bacterial biomass dry weight. Alkalinization was performed with 0.06 M sodium hydroxide. The pH measured at the beginning of the lysis was 12.5. Then the lysis was incubated for 2 hours at 40 0 C under continuous stirring. After the incubation, the pH was 12.3.
  • Lactobacillus fermentum I-3929 biomass from Example 1.1 containing 6.8 g of bacterial dry weight was thawed at room temperature, then diluted with purified water to reach 7 g/L of bacterial biomass dry weight. Alkalinization was performed with 0.08 M sodium hydroxide. The pH measured at the beginning of the lysis was 12.2. Then the lysis was incubated for 7 hours at 40 0 C under continuous stirring. After the incubation, the pH was 12.0. The lysis pH was adjusted to 9.8 with HCI.
  • the lysis comprised solubilised dry weight (SDW): 20.5 mg/g, proteins (Prot): 4.9 mg/g.
  • Lactobacillus fermentum I-3929 biomass from Example 1.1 containing 6.8 g of bacterial dry weight was thawed to room temperature then diluted with purified water to reach 7 g/L of bacterial biomass dry weight. Alkalinization was performed with 0.15 M sodium hydroxide. The pH measured at the beginning of the lysis was 13.0. Then the lysis was incubated for 63 hours at 40 0 C under continuous stirring. After the incubation, the pH was 12.6. The lysis pH was adjusted to 9.9 with HCI. [0129] The lysis comprised SDW: 23.7 mg/g, Prot: 5.0 mg/g.
  • Lactobacillus fermentum I-3929 biomass from Example 1.1 containing 98 g of bacterial dry weight was thawed to room temperature, then diluted with purified water to reach 10 g/L of bacterial biomass dry weight. Alkalinization was performed with 0.08 M sodium hydroxide. The pH measured at the beginning of the lysis was 12.0. Then the lysis was incubated for 24 hours at 40°C under continuous stirring. After the incubation, the pH was 11.1. The lysis pH was adjusted to 11.5 with NaOH.
  • the lysis comprised SDW: 23.7 mg/g.
  • Lactobacillus fermentum I-3929 biomass from Example 1.1 containing 39 g of bacterial dry weight was thawed to room temperature, then diluted with a 0.2N NaCI solution to reach 7.6 g/L of bacterial biomass dry weight in order to produce an osmotic lysate.
  • the pH measured at the beginning of the lysis was 4.4.
  • the bacterial osmotic lysat was incubated for 24 hours at 40°C under continuous stirring. After the incubation, the pH was 4.2.
  • the bacterial lysate was then made alkaline with addition of NaOH to reach 0.06 N.
  • the pH measured at the beginning of the alkaline lysis was 10.6.
  • the lysis was re- incubated for 2.25 hours at 40°C under continuous stirring. After the incubation, the pH was 10.2.
  • Example 2 The lysate of Example 2.5 was centrifuged for 20 minutes at 3000 x g. Then the supernatant was adjusted to pH 6.8 with NaOH 10N and filtered through successive filters with porosities of 0.45 ⁇ m and 0.2 ⁇ m, and finally with a sterile filter 0.22 ⁇ m.
  • the concentrate comprised Prot: 2.8 mg/mL; DNA: 17.4 ⁇ g/mL.
  • Example 2.6 The lysate of Example 2.6 was centrifuged for 20 minutes at 3000 x g. Then the supernatant was adjusted to pH 7 with HCI and filtered through successive filters with porosities of 0.45 ⁇ m and 0.2 ⁇ m, and finally with a sterile filter 0.22 ⁇ m.
  • the concentrate comprised Prot: 12.3 mg/mL; DNA: 27.7 ⁇ g/mL.
  • the concentrate comprised SDW: 30.0 mg/g; Prot: 9.6 mg/mL; DNA: 32.8 ⁇ g/mL.
  • D-Amino acid percentage 14.3% D-AIa, 13.9% D-Pro, 14.1% D- Asp, 36.9% D-Ser.
  • Example 2.2 300 ml of the lysate of Example 2.2 was adjusted to pH 7.0 with HCI. The extract was filtered as described in Example 3.3 with a constant flow at 350 mL/min. The yield volume was more than 75%. The concentrate was finally sterile filtered through a PES membrane 0.2 ⁇ m (Nalgene).
  • the concentrate comprised SDW: 49.2 mg/g; Prot: 14.2 mg/mL; DNA: 34.1 ⁇ g/mL.
  • D-Amino acid percentage 16.0% D-AIa, 13.3% D-Pro, 14.2% D- Asp.
  • NO production in mg of active dry weight /mL 0.003 mg/mL (C1 )
  • C2 4.1 ⁇ M
  • C3 26.3 ⁇ M.
  • Example 2.3 300 ml of the lysate of Example 2.3 was first centrifuged at 3000xg for 20 minutes. The supernatant was adjusted to pH 7.1 with HCI. The extract was filtered as described in Example 3.3 with a constant flow at 330-350 mL/min. The yield volume was more than 75%. The concentrate was finally sterile filtered through a PES membrane 0.2 ⁇ m (Nalgene).
  • the concentrate was composed of SDW: 34.5 mg/g; Prot: 8.9 mg/mL; DNA: 16.8 ⁇ g/mL.
  • D-Amino acid percentage 16.4% D-AIa, 24.3% D-Pro, 17.3% D-Asp.
  • C2 - 0.03 mg/mL
  • C3 0.3 mg/mL (C3) of active dry weight /mL: C1 : 3.0 ⁇ M, C2: 36.4 ⁇ M, C3: 69.5 ⁇ M.
  • the concentrate comprised SDW: 32.7 mg/g; Prot: 6.5 mg/g; Sugar: 2.4 mg/g.
  • the sugar concentration was assayed according to the procedure by Herbert et al. (Meth. Microbiol, 1971 , 5B:266 et seq.).
  • PBMC Peripheral Blood Mononuclear Cells
  • the bacterial lysate mixture of Example 2.10 was transferred into a microfiltration (MF) tank after being adjusted to pH 10.2.
  • the microfiltration (MF) unit used a 0.45 ⁇ m tangential flow filtration (TFF) filter (Sartocon Slice Sartorius).
  • the cross flow was adjusted to 290 L/h m 2 (LHM) and the Trans Membrane Pressure (TMP) to 0.4-0.5 bar.
  • the permeate was transferred to an ultrafiltration (UF) tank.
  • the UF unit was started.
  • the permeate of UF filters was used as washing buffer in MF tank.
  • the volumes of both the MF and UF tanks were maintained at the same level.
  • the UF was stopped, and the bacterial lysate was concentrated in the MF tanks.
  • the recovered volumeric yield was 83%.
  • the recovered extract in UF tank was adjusted to pH 7.3 with HCI and then filtered through a 0.2 ⁇ m sterile filter.
  • the concentrate comprised SDW: 17.5 mg/g; Prot: 4.3 mg/g.
  • Example 2.7 The lysate of Example 2.7 was centrifuged for 15 minutes at 9384 x g. Then the supernatant was adjusted to pH 7.3 with HCI and filtered through successive filters with porosities of 0.45 ⁇ m and 0.2 ⁇ m, and finally with a sterile filter 0.22 ⁇ m.
  • PBMC Peripheral Blood Mononuclear Cells
  • Example 2.8 The lysate of Example 2.8 was centrifuged for 15 minutes at 9384 x g. Then supernatant was adjusted to pH 7.4 with HCI and filtered through successive filters with porosities of 0.45 ⁇ m and 0.2 ⁇ m, and finally with a sterile filter 0.22 ⁇ m.
  • PBMC with 10 times diluted product 904 pg/mL IL-10, 0 pg/mL IL-12, 4995 pg/mL IL-6.
  • Example 2.9 The lysate of Example 2.9 was centrifuged for 15 minutes at 9384 x g. Then the supernatant was adjusted to pH 7.6 with HCI and filtered through successive filters with porosities of 0.45 ⁇ m and 0.2 ⁇ m, and finally with a sterile filter 0.22 ⁇ m.
  • PBMC with 10 times diluted product 476 pg/mL IL-10, 0 pg/mL IL-12, 4924 pg/mL IL-6.
  • Example 2.11 The lysate of Example 2.11 was transferred to an MF tank after being adjusted to pH 10.4.
  • the TFF installation was similar to Example 3.7.
  • the cross flow was adjusted to 290 L/h m 2 and the TMP to 0.4-0.5 bar.
  • the UF was stopped after 10 diaf iltration volumes.
  • the recovered volume yield was 86%.
  • the final product was adjusted to pH 7.3 with HCI.
  • the concentrate obtained comprised SDW: 24.2 mg/g; Prot: 4.8 mg/g.
  • Example 2.12 The lysate of Example 2.12 was transferred to an MF tank after being adjusted to pH 11.5.
  • the TFF installation was similar to Example 3.7.
  • the cross flow was adjusted to 290 Uh m 2 and the TMP to 0.4 bar.
  • the UF was stopped after 10 diaf iltration volumes.
  • the recovered volume yield was 74%.
  • the final product was adjusted to pH 7.2 with HCI.
  • the concentrate obtained comprised SDW: 32.4 mg/g; Prot: 5.8 mg/g; Sugar: 2.3 mg/g.
  • PBMC Peripheral Blood Mononuclear Cells
  • Results obtained at 0.5 mg of active dry weight /ml_ were TNF- ⁇ : 37 pg/mL; IL-6: 1092 pg/mL; IL-12: 81 pg/mL (in the presence of 10 ng/ml IFN- ⁇ ); and IL-10: 160 pg/mL (in the presence of 10 ng/ml IFN- ⁇ ).
  • Protein concentration was measured by the DC Protein assay (BioRad, DC Protein assay, kit no. 500-0116). The microplate assay protocol was performed.
  • Samples to analyze were diluted with 1.9 ml_ of 25 mM phosphate buffer pH 11.
  • a protein standard curve was prepared each time the assay was performed with a bovine serum albumine solution at 2 mg BSA /ml (BSA, Pierce ref 23210).
  • BSA solution was diluted with 25 mM phosphate buffer pH 11 to obtain the following concentrations: 0.18, 0.24, 0.3, 0.36 and 0.42 mg BSA/ml.
  • Samples (20 ⁇ l) and BSA standards (20 ⁇ l) were added in quadruplicate into a clean dry microtiter plate. Reagent A (25 ⁇ l) from the DC Protein assay kit was added into each well.
  • the soluble dry weight (SDW) was determined by obtaining 5 mL of the soluble fraction resulting from the lysis and drying it to a constant mass in a porcelain dish at 105°C.
  • the AlamarBlueTM-Assay is designed to measure quantitatively the growth of human or animal cells.
  • This assay incorporates a fluorometric/colorimetric growth indicator based on the detection of metabolic activity by an oxidation-reduction (REDOX) indicator in response to chemical reduction resulting from cell growth.
  • REDOX oxidation-reduction
  • the REDOX indicator causes changes from the oxidized (non-fluorescent, blue) form to the reduced (fluorescent, red) form.
  • mice Balb/c mice (female, 6-8 weeks of age) were received from Charles River Laboratories, Sulzfeld, Germany, and were sacrificed by cervical dislocation. Spleens were homogenized using a potter; cell suspensions were washed by centrifugation (280 x g, 4°C, 10 min) and resuspended in 5 ml RPMI 1640 containing 5% FCS, 100 U/ml penicillin and 100 ⁇ g/ml streptomycin.
  • spleen cells 100 ⁇ l spleen cells (2 x 10 6 /ml) were incubated with 50 ⁇ l of bacterial extract dilutions in 96-well plates (Falcon 3072) for 48 h at 37°C and 5 % CO 2 in cell culture medium.
  • AlamarBlueTM After the addition of 30 ⁇ l AlamarBlueTM solution diluted 1 :1 with culture medium, the chemical reduction of AlamarBlueTM was measured with a Fluoroskan Ascent Reader (ThermoLabsystems, Frankfurt, Germany) at 544 nm excitation and 590 nm emission wavelengths.
  • Afer300 extract from Lactobacillus ferment ⁇ m 1-3929 (12 g/L) obtained as described in Example 3.5 (31.6 mg of active dry weight /g);
  • Cfer300 extract from Lactobacillus fermentum 1-3929 (12 g/L) obtained as described in Example 3.3 (28.4 mg of active dry weight /g);
  • Dfer300 extract from Lactobacillus fermentum 1-3929 (12 g/L) obtained as described in Example 2.4 and purified using the same conditions as Example 3.3 (29.5 mg of active dry weight /g). This example was used as a non- alkaline lysed control.
  • ARahr300 extract from Lactobacillus rhamnosus 71.38 (40 g/L) obtained by an alkaline lysis with NaOH 0.03 M at 40°C for 6 hours (49.3 mg of active dry weight /g);
  • CRahr300 extract from Lactobacillus rhamnosus 71.38 (40 g/L) obtained as described in Example 3.4 (46.4 mg of active dry weight /g);
  • DRahr300 extract from Lactobacillus rhamnosus 71.38 (40 g/L) lysed by osmotic stress in 0.15 N NaCI solution at 40 0 C for 6 hours (46.2 mg of active dry weight /g). This example was used as a non-alkaline lysed control.
  • ARahr300, CRahr300, DRahr300 Three extracts obtained from a second strain, Lactobacillus rhamnosus 71.38 (ARahr300, CRahr300, DRahr300).
  • ARahr300 and CRahr300 are within the scope of the invention while DRahr is an osmotic lysis control.
  • the ARahr300 extract was effective in two independent assays at a dilution of 1 :300, whereas the CRahr300 and DRahr300 extracts showed a weaker but significant stimulation in one of the two assays at the same dilution.
  • Lactobacillus fermentum I-3929 extracts were more effective in stimulating spleen cell growth in comparison to the Lactobacillus rhamnosus 71.38 extracts.
  • Example 5 Production of Nitric Oxide by Bone Marrow-Derived Macrophages
  • Macrophages were then detached with ice-cold PBS, washed and resuspended in DH medium supplemented with 5% fetal calf serum (FCS), amino acids and antibiotics (DHE medium).
  • FCS fetal calf serum
  • DHE medium amino acids and antibiotics
  • the cell density was adjusted to 700000 cells/mL
  • Aqueous solutions of the extracts were serially diluted in DHE medium directly in microtiter plates.
  • the extracts of the invention were tested in triplicates and each microtiter plate comprised the medium as negative control.
  • the final volume in each well was 100 ⁇ l_.
  • 100 ⁇ l_ of the cell suspension was added to the diluted extracts and the cells were incubated for 22 hours in an incubator at 37°C, under 8% CO 2 and a moisture-saturated atmosphere.
  • 100 ⁇ l_ of supernatant was transferred to another microtiter plate and the nitrite concentration produced in each supernatant was determined by running a Griess reaction.
  • 100 ⁇ l_ of Griess reagent (5 mg/mL of sulfanilamide + 0.5 mg/mL of N-(1 - naphtyl)ethylene-diamine hydrochloride) in 2.5% aqueous phosphoric acid, was added to each well.
  • the microtiter plates were read with a spectrophotometer (SpectraMax Plus, Molecular Devices) at 562 nm against a reference at 690 nm. The nitrite concentration was proportional to nitric oxide content being formed.
  • nitrite content was determined based on a standard curve of sodium nitrite (1 to 70 ⁇ M NaNO 2 ). The results were given in ⁇ M nitric oxide (NO) as mean value ⁇ standard deviation and plotted as a dose response curve. Test articles [0181 ] The following extracts were tested: First assay:
  • OP0701 B4_CFer300 extract from Lactobacillus fermentum I- 3929 (12 g/L) obtained as described in Example 3.3;
  • OP0701 B4_Dfer300 extract from Lactobacillus fermentum I- 3929 (12 g/L) obtained as described in Example 2.4 and purified using the same conditions as Example 3.3. This example was used as a non-alkaline lysed control.
  • OP0701 B4_CRahr300 extract from Lactobacillus rhamnosus 71.38 (40 g/L) obtained as described in Example 3.4
  • OP0701 B4_ DRahr300 extract from Lactobacillus rhamnosus 71.38 (40 g/L) obtained by osmotic stress in 0.15 N NaCI solution at 40 °C for 6hours. This example was used as a non-alkaline lysed control.
  • OP0701 C_10G0.5P4H extract from Lactobacillus fermentum I- 3929 at 10 g/l of biomass dry weight with 0.037M NaOH incubated at 40 0 C for 4 hours.
  • OP0701 C_1 OG 1 P4H extract from Lactobacillus fermentum I- 3929 at 10 g/l of biomass dry weight with 0.075M NaOH incubated at 40°C for 4 hours.
  • OP0701 C_10G2P4H extract from Lactobacillus fermentum I- 3929 at 10 g/l of biomass dry weight with 0.150 M NaOH incubated at 40 0 C for 4 hours.
  • OP0701 C_1 OG 1 P21 H extract from Lactobacillus fermentum I- 3929 at 10 g/l of biomass dry weight with 0.075M NaOH incubated at 4O 0 C for 21 hours.
  • OP0701 C_10G0.5P21 H extract from Lactobacillus fermentum I- 3929 at 10 g/l of biomass dry weight with 0.037M NaOH incubated at 40 0 C for 21 hours.
  • OP0701 C_10G2P21 H extract from Lactobacillus fermentum I- 3929 at 10 g/l of biomass dry weight with 0.150 M NaOH incubated at 40 0 C for 21 hours.
  • OP0701 C_10G2P45H extract from Lactobacillus fermentum I- 3929 at 10 g/l of biomass dry weight with 0.150 M NaOH incubated at 40°C for 45 hours. Results first assay
  • Graded amounts of the 6 bacterial extracts were diluted in cell culture medium in the presence of 10 ng/ml of IFN- ⁇ , a cytokine known to enhance the production of IL-12p70, starting with an initial dose of 1 mg/ml (highest final dose tested).
  • PBMCs isolated from the blood of healthy donors were tested as serial dilutions of active dry weight extracts from 100 ng/ml to 1 mg/ml.
  • PBMCs were isolated from peripherical blood. Briefly, after a Ficoll gradient centrifugation (Pharmacia, Uppsala, Sweden), mononuclear cells were collected, washed in RPMI 1640 medium (Live technologies, Paisley, Scotland) and adjusted to 2 x 10 6 cells/mL in RPMI 1640 supplemented with gentamicin (150 ⁇ g/mL), L-glutamine (2 mmol/L), and 10% foetal calf serum (FCS) (Gibco-BRL). Induction of cytokines
  • PBMCs (2 x 10 6 cells/mL) were seeded in 24-well tissue culture plates (Corning, NY). Cells were stimulated as described above. After 24 hours stimulation at 37°C in an atmosphere of air with 5% CO 2 , the culture supernatants were collected, clarified by centrifugation and stored at -20 0 C until cytokine analysis. Cytokines were measured by ELISA using pharmingen antibody pairs (BD Biosciences, San Jose, Ca, USA) for IL-10 and IL-12p70, according to the manufacturer's recommendations. Test articles
  • OP0701 C_10G2P45H (A): extract from Lactobacillus fermentum I-3929 lysis at 10g of biomass dry weight / liter of lysis, 0.15 M NaOH at 40°C for 45 hours.
  • OP0701 B4_BFer300 extract from Lactobacillus fermentum I- 3929 lysis at 6 g of biomass dry weight / liter of lysis, 0.075 M NaOH at 40 0 C for 6 hours.
  • the aim of this test was to compare the in vitro immunostimulating or anti-inflammatory potential of embodiments of the invention with a live Lactobacillus strain.
  • the quantity of each cytokine IL-10 and IL-12 released from PBMCs was compared, as well as the IL10/IL12 ratio.
  • Table 2 in vitro pro-inflammatory (immunostimulating) or anti-inflammatory (immunomodulatory) potential of the bacterial extracts in human PBMCs.
  • Table 3 in vitro pro-inflammatory (immunostimulating) or anti-inflammatory (immunomodulatory) potential of the live Lactobacillus fermentum Strain I-3929 in human PBMCs.
  • Embodiments of the invention are obtained from gram positive bacteria, and therefore are expected to act via TLR2 receptors.
  • TLR receptors are expressed principally, but not exclusively, by immune cells such as monocytes, macrophages, dendritic cell, T-cells etc, and are key sensors of microbial products, which can be recognized as signal dangers by the host. Even though they trigger first an unspecific innate immunity, TLR activation initiates a full immunological cascade which results, in the presence of antigens, to the development of acquired immunity.
  • the HEK293 cell line was chosen for its null or low basal expression of the TLR genes. These cells enable efficient monitoring of TLR activity using ELISA analysis such as IL-8 titration or reporter-based systems that monitor TLR-induced NF- ⁇ B activation.
  • HEK-TLR2/6 cells (Invivogen, Toulouse, France) are engineered HEK293 cells stably transfected with multiple genes from the TLR2/6 pathway that include TLR2, TLR6 and genes participating in the recognition or involved in the signaling cascade. These cells secrete IL-8 after TLR2/6 stimulation. Experiments were performed according to the manufacturer's instructions.
  • OP0701 B4_Afer50 extract from Lactobacillus fermentum I-3929 lysis at 12g of biomass dry weight / liter of lysis, 0.075 M NaOH at 40°C for 4 hours, purified as described in Example 3.6.
  • OP0701 C-BtI LAC extract from Lactobacillus fermentum 1-3929 obtained as described in Example 3.7.
  • the cell line HEK TLR2/6 responded to the TLR2 agonist Pam3Cys.
  • the TLR 4 agonist from E coli was inactive, even at the high dose of 0.01 ⁇ g/ml.
  • IL-8 (pg/ml) secretion from supernatants obtained on HEK TLR2/6 cells stimulated at the indicated concentrations with the controls (medium, LPS, or Pam3Cys), with the bacterial extract
  • TLR4 was extensively studied since it is the major receptor involved in the recognition of lipopolysaccharide (LPS) responsible for sceptic shock.
  • LPS lipopolysaccharide
  • HEK-TLR4-MD2 -CD14 cells are highly sensitive to LPS. They were obtained by stable transfection of HEK293 cells with the TLR4, MD2 and CD14 genes and an NF- ⁇ B-inducible reporter system. These cells secrete IL-8.
  • the cell line responds clearly only to the TLR4 agonist LPS K12. In contrast, as expected, the TLR2 agonist from Pam3Cys was inactive, even at the high dose of 0.01 ⁇ g/ml.
  • Table 7 IL-8 (pg/ml) secretion from supernatants obtained on HEK TLR4 cells stimulated at the indicated concentrations with the controls (medium, LPS 1 or Pam3Cys), with the bacterial extracts
  • Table 8 IL-8 (pg/ml) secretion from supernatants obtained on HEK TLR4 cells stimulated at the indicated concentrations with the controls (medium, LPS, or Pam3Cys), with the bacterial extracts
  • Bacterial extracts according to the invention were tested in 96 well microplates.
  • the extracts were diluted in DMEM culture medium and 20 ⁇ l of each dilution was tested in duplicate.
  • a volume of 180 ⁇ l of HEK293 cell suspensions containing 25000 or 50000 cells in DMEM culture medium + 10% FCS one HEK293 cell line specific for each TLR with reporter gene secreted alkaline phosphatase under the control of NF-kB
  • FCS one HEK293 cell line specific for each TLR with reporter gene secreted alkaline phosphatase under the control of NF-kB
  • Enzymatic reaction with secreted alkaline phosphatase was performed for 30 to 60 min for the different series of TLRs expressing cell lines. Read out was performed using a microplatereader at 630 nm. Results are expressed in OD at 630 nm.
  • TLR2 PAM2 100 ng/ml; TLR3 PoIy(I :C) 100 ng/ml; TLR4 E. coli K12 LPS 1 ⁇ g/ml; TLR5 S. typhimurium flagellin 1 ⁇ g/ml; TLR7 R848 10 ⁇ g/ml; TLR8 R848 10 ⁇ g/ml; TLR9 CpG ODN 2006 10 ⁇ g/ml; NOD2 Muramyldipeptide 1 ⁇ g/ml. Negative controls
  • a recombinant HEK-293 cell line for the reporter gene only (NFkB) was used as a negative control for the TLR cell lines.
  • the negative control value for each clone was the background signal of these non-induced clones.
  • TNF- alpha was used as a positive control for this non-TLR expressing cell line.
  • OP0701 B4_CFer300 extract from Lactobacillus fermentum I- 3929 (12 g/L) obtained as described in Example 3.3 (F);
  • OP0701 B4_CRahr300 extract from Lactobacillus rhamnosus 71.38 (40 g/L) obtained as described in Example 3.4 (G);
  • OP0701 D_10L1 PswitchA extract from Lactobacillus fermentum /-3929 (10 g/L) obtained as described in Example 3.12 (I);
  • OP0701 D_5LOSMOConc extract from osmotic lysis Lactobacillus fermentum 1-3929 (7.6 g/L) obtained by osmotic stress in 0.16 N NaCI solution at 40 °C for 24 hours (H) as a control.
  • Bacterial extract F (OP0701 B4Cfer300)
  • Table 10 activation of TLR's and NOD2 receptors by OP0701 B4Cfer300 extracts.
  • Bacterial extract F activated specifically hTLR2, hTLR4 and hNod2, and to a lesser extent, hTLR5.
  • Bacterial extract G (OP0701 B4CRahr300)
  • Table 11 activation of TLR's and NOD2 receptors by OP0701 B4CRahr300 extracts.
  • Bacterial extract G activated hTLR2, hNOD2, and to a much lesser extent, hTLR4.
  • Bacterial extract H (OP0701 D 5LOSMOConc)
  • Table 12 activation of TLR's and NOD2 receptors by OP0701 D_5LOSMOConc extracts.
  • Bacterial extract I (OP0701 D 10L1 PswitchA)
  • Table 13 activation of TLR's and NOD2 receptors by OP0701 D_10LPswitchA extracts.
  • Bacterial extract I strongly activates hTLR2, hTLR5, hNOD2, and to a lesser extent, hTLR4 and hTLR9.
  • Table 15 activation of TLR's and NOD2 receptors by different bacterial extracts obtained from different process conditions according to the present invention.
  • the bacterial extracts according to the present invention may help to activate the immune system via various TLRs, and also Nod2. Therefore the extracts of the invention may be good activators of immune response.
  • Extract I acted on TLR5, which indicates a potential application in radioatherapy. Indeed, radiation therapy is a well-established and highly effective treatment for certain types of cancer. Its side effects can be devastating, however, as it can destroy healthy cells in the body, especially bone-marrow cells and cells in the gastrointestinal tract. Burdelya et al. reported recently that a CBLB502 peptide binds to TLR5 and activates the nuclear factor- ⁇ B signaling pathway, a pathway that cancer cells often activate to avoid cell death (Burdelya et al., "An agonist of toll-like receptor 5 has radioprotective activity in mouse and primate models," Science, 2008, 320:226-30).
  • mice and rhesus monkeys treated with CBLB502 shortly before exposure to lethal doses of total body irradiation exhibited less damage to healthy bone marrow and gastrointestinal cells, and survived significantly longer than controls. Importantly, in tumor-bearing mice, CBLB502 did not compromise the antitumor efficacy of radiation therapy.
  • Example 9 Ability of extract to produce plaque forming cells (against sheep red blood cells) in mice
  • the plaque forming cells (PFC) technique enables the evaluation of a non-specific stimulation of B-lymphocytes. This technique was first described by Cunningham and Seenberg (Immunology, 1968, 14, 599). The presence of hemolytic antibodies around antibody-forming cells was demonstrated as described below. Murine lymphoid cells, as well as a dense population of foreign (sheep) erythrocytes, were simultaneously introduced on a microscope slide. Certain lymphoid cells release hemolytic antibodies which diffuse and cause the lysis of the neighboring red blood cells by forming a lysis plaque in the presence of complement. At the end of the experiment, the number of PFC reported to 10 6 cells or to the spleen was counted.
  • Group (a) 8 mice receiving 1.2 mg/mouse of extract 1 obtained as described in Example 3.5; volume 0.2 ml Group (b) 8 mice receiving 0.6 mg/mouse of extract
  • mice received the extracts described above, at the doses indicated, daily per os (via the oral route) for 5 consecutive days (day 1 to day 5). Two more intubations occurred on days 19 and 20.
  • the antigen (10 6 sheep red blood cells) in 0.2 ml isotonic saline (0.9% NaCI, Alsever solution) was injected invtraveneously through the caudal vein of the animal; 4 days later (day 33) the spleen cell suspensions were prepared.
  • Control animals received Sheep Red Blood Cells (SRBC) only. Animals of other groups received the bacterial extracts per os as indicated previously. To this end, each extract was dissolved in water.
  • SRBC Sheep Red Blood Cells
  • Spleen cells suspension 4 days after the last antigen (SRBC) injection, a suspension of spleen cells was prepared for each mouse according to the following procedure:
  • the animal was anesthetized with ether and then sacrificed by cervical dislocation.
  • the spleen was removed and crushed in a glass tissue homogenizer with 2 ml BME at pH 6.75-6.80.
  • 3 ml of sterile distilled water were added, and the mixture was stirred for 20 seconds.
  • the suspension obtained was further diluted with 10 BME and centrifuged at 1200 rpm for 10 minutes at 4°C.
  • the supernatant was discarded and the precipitate was suspended in 2 ml BME.
  • the cells were allowed to recover 5-10 minutes on ice.
  • a viability count of the mononucleated cells was performed in the following way:
  • a 1/20 dilution of an aliquot of the cell suspension was prepared with saline containing a 0.1 % solution of trypan blue .
  • the non-colored cells, i.e. the living cells, were counted in a Neubauer chamber.
  • a fraction of the spleen cell suspension was adjusted to 25,000 mononucleated cells per mm 3 .
  • a mixture of the following elements (added in the order described) was prepared by means of an automatic pipette:
  • the lysis plaques were easy to recognize. 5 vertical optic strips were examined on each slide and the number of lysis plaques was counted. The number of cells (N) forming direct lysis plaques per 10 6 cells was calculated by extrapolation. If x is the number of observed plaques and X the number of examined cells, the number of direct lysis plaques per 10 6 cells is equal to:
  • N (X / X) * 10 6
  • L length of an optic strip
  • the extracts of the invention appeared to be B-cell activators when tested at concentrations of 1.2 mg/mouse/per day or 0.6 mg/mouse/day. A slight dose-dependent effect was observed. Hence, some extracts of the invention could potentially be used to prime the immune system in patients suffering from recurrent infections.
  • Example 10 Effect of CFer300 in intraperitoneal Salmonella typhimurium infection in balb/c mice
  • mice were housed in the facilities of the Institute for Immunology, Moscow.
  • non-inbred laboratory grade white mice were purchased from Stolbovaya, the Russian State Scientific Centre for Biomedical Technologies (Russian Academy of Medical Sciences). Upon arrival, the mice had a body weight of 12-14 g. Throughout the experiments, mice were maintained at pathogen-free conditions on standard rodent diet and water.
  • Study groups (main experiment) [0290] Two groups of 22 mice per group were used to test the anti- infective efficacy of the bacterial extracts prepared using an experimental model of Salmonella typhimurium infection in mice.
  • One group was treated orally with a solution of CFer300, and the second group received a sham treatment (water) as a negative control.
  • Group 1 Mice treated with CFer300 given per os as single 2 mg (0.5 ml) doses.
  • Group 2 Mice received a sham treatment using oral administration of 0.5 ml water daily for 10 days.
  • the bacterial extract tested was OP0701 B4CFer300 ("CFer300"); the extract from Lactobacillus fermentum I-3929 (12 g/L) obtained as described in Example 3.3 (28.4 mg of active dry weight /g).
  • the purpose of the preliminary experiment was to determine the dose of the infectious agent that would induce mortality close to 50% three weeks after the challenge.
  • a suspension of Salmonella enterica, serovar typhimurium strain 415 (I. Mechnikov, Institute for Vaccines and Sera, Russian Academy of Medical Sciences) was injected intraperitoneally into each mouse.
  • the challenge dose ranged from 10 3 to 10 5 CFU of Salmonellae per mouse.
  • mice were kept under standard conditions for laboratory animals. Daily observations and records of death were recorded during a period of 21 days post infection.
  • the anti-infective efficacy of the preparations was estimated according to the post infection survival rate (SR), the post infection average duration of life (ADL), the defense factor (DF), and the preparation efficacy index (El), calculated for each experimental group.
  • the SR was taken as percent of animals alive in the experimental groups on day 21 post infection.
  • ADL is the average duration of life
  • X1 to Xn are the durations of life post-infection for experimental mice 1 to n.
  • N is the total number of animals in the experimental group.
  • DF is the defense factor
  • CD is the percent of death in the control group
  • ED is the percent of death in the experimental group.
  • El is the preparation efficacy index
  • DF is the defense factor
  • Table 17 Preliminary experiment to determine the challenge dose of Salmonella typhimurium corresponding to approximately 50% death rate.
  • Example 11 Effect of two Lactobacillus extracts in a model of LACK-induced asthma
  • mice [0307] A total of 27 mice were divided into 4 groups as follows:
  • Group A untreated, LACK-sensitized and saline- challenged mice; LACK is a protein from the parasite Leishmania major (4 mice).
  • Group B untreated LACK-sensitized and challenged mice (8 mice)
  • Group C OM-1009A-treated (8 mg of dry weight residue per administration), LACK-sensitized and challenged mice (8 mice)
  • Group D OM-1009B-treated (8 mg of dry weight residue per administration), LACK-sensitized and challenged mice (7 mice) Treatment and schedule with test or control article
  • mice were treated from day -3 to day 22. On day 0 and day 7, mice were sensitized intraperitoneal ⁇ (i.p.) with 10 ⁇ g of LACK in the presence of 2 mg of alum 3. From day 17-21 , mice were exposed to a daily 20-min aerosol challenge of a LACK solution (0.15%) (Groups B, C, and D), or a saline solution (Group A) as control. On day 22, mice were analyzed for their ability to develop AHR upon methacholine inhalation. On day 23, the mice were sacrificed and lung inflammation was assessed.
  • a LACK solution 0.15%
  • mice mice were analyzed for their ability to develop AHR upon methacholine inhalation.
  • mice were sacrificed and lung inflammation was assessed.
  • mice were therapeutically treated three times as described above. Lavages were performed in individual mice bled with a canula inserted into their trachea. Lungs were washed 3 times with 1 ml of warmed PBS. Cells were washed with PBS, resuspended in 300 ⁇ l, and counted using a Burker-T ⁇ rk chamber. For differential BAL cell counts, cytospin preparations were made and stained with Wright/Giemsa coloration.
  • mice LACK-sensitized and PBS-challenged wild type mice (Control), LACK-sensitized and challenged wt mice (asthmatic animals), OM-1009A-treated wt mice, and OM-1009B-treated wt mice.
  • the total cell number in BAL was determined by microscopic examination of cytospin preparations stained with Wright/Giemsa coloration.
  • mice were analyzed for their ability to develop AHR upon methacholine inhalation at the doses indicated. The results are reported in Figure 5. The results show that OM-1009-B restored basal Penh values similar to the PBS untreated non-asthmatic group. OM-1009-A was more efficient, since the Penh values obtained were even lower than those observed in the non-asthmatic control group. b) Total and differential cell number in broncho-alveolar lavages
  • mice were sacrificed and lung inflammation was assessed.
  • the total cell number in BAL is reported in Table 20 (right column), and also the differential cell numbers: Eosinophils number (Eo), Neutrophils number (Neutro), Lymphocytes number (Lympho), and Other cells number (Other).
  • Table 20 Total and differential cell number in BAL upon treatment with two extracts of the invention (Groups C and D). Mean values and standard error mean values are provided.
  • Both extracts decreased significantly the total cell number in BAL (p ⁇ 0.01 for OM-1009A; and p ⁇ 0.03 for OM-1009B when compared to group B).
  • the eosinophils numbers were decreased 3- and 1.7-fold respectively.
  • the neutrophils numbers were decreased 1.8- and 3.6-fold respectively.
  • the reduction of the eosinophil and neutrophil numbers indicates a lower concentration of inflammatory cells in BAL of animals pretreated with the extracts.
  • Th2 cytokines IL4, IL5, and IL13
  • IL4, IL5, and IL13 Th2 cytokines
  • IL4 cytokines IL4, IL5, and IL13
  • OM-1009A bacterial extracts obtained by a strong base treatment
  • OM-1009-A may be less active than those obtained under a moderate base treatment
  • OM-1009A may be a better candidate than OM-1009B for treatment or prevention of conditions related to Th2-related diseases, such as allergic diseases and atopy, or symptoms thereof.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Pulmonology (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Otolaryngology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

La présente invention concerne des extraits de bactéries Lactobacillus, qui peuvent produire des effets immunomodulateurs chez des sujets. Des modes de réalisation de l’invention peuvent être utilisés, par exemple, en tant qu’agents nutraceutiques ou pharmaceutiques pour le traitement de maladies ou en tant qu’adjuvants dans un traitement médical, tel que ceux associés à un déséquilibre de la production de cytokines anti-inflammatoires ou pro-inflammatoires. Les affections pour lesquelles les extraits de l’invention peuvent être utiles comprennent des infections, des allergies, des troubles auto-immuns, et l’inflammation, ou en tant qu’adjuvants produisant des bénéfices pour la santé chez des sujets. L’invention comprend en outre, entre autres, des procédés de fabrication et d’utilisation de tels extraits. L’invention concerne en outre des souches particulières de bactéries Lactobacillus.
PCT/US2008/010350 2008-09-04 2008-09-04 Extraits immunomodulateurs de bactéries lactobacillus et procédés de fabrication et d’utilisation de ceux-ci WO2010027344A1 (fr)

Priority Applications (11)

Application Number Priority Date Filing Date Title
CN2008801309602A CN102143756A (zh) 2008-09-04 2008-09-04 来自乳杆菌属细菌的免疫调节提取物及其制备及使用方法
BRPI0822997-0A BRPI0822997A2 (pt) 2008-09-04 2008-09-04 Extratos imunomoduladores de bactérias lactobacillus e métodos para fabricação e uso dos mesmos
AU2008361375A AU2008361375B2 (en) 2008-09-04 2008-09-04 Immunomodulatory extracts from Lactobacillus bacteria and methods of manufacturing and use thereof
PCT/US2008/010350 WO2010027344A1 (fr) 2008-09-04 2008-09-04 Extraits immunomodulateurs de bactéries lactobacillus et procédés de fabrication et d’utilisation de ceux-ci
RU2011112781/15A RU2500412C2 (ru) 2008-09-04 2008-09-04 ИММУНОМОДУЛИРУЮЩИЕ ЭКСТРАКТЫ ИЗ БАКТЕРИЙ Lactobacillus И СПОСОБЫ ИХ ПОЛУЧЕНИЯ И ПРИМЕНЕНИЯ
JP2011526017A JP2012502026A (ja) 2008-09-04 2008-09-04 ラクトバチルス属細菌の免疫調節抽出物及びその製造法と使用法
MX2011002080A MX2011002080A (es) 2008-09-04 2008-09-04 Extractos inmunomoduladores de bacteria de lactobacillus y metodos de manufactura y uso de los mismos.
KR1020117007527A KR20110051268A (ko) 2008-09-04 2008-09-04 락토바실러스 박테리아 유래 면역조절 추출물 및 이의 제조 방법 및 이의 용도
CA2733249A CA2733249A1 (fr) 2008-09-04 2008-09-04 Extraits immunomodulateurs de bacteries lactobacillus et procedes de fabrication et d'utilisation de ceux-ci
ZA2011/00899A ZA201100899B (en) 2008-09-04 2011-02-03 Immunomodulatory extracts from lactobacillus bacteria and methods of manufacturing and use thereof
IL211041A IL211041A0 (en) 2008-09-04 2011-02-03 Immunomodulatory extracts from lactobacillus bacteria and methods of manufacturing and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2008/010350 WO2010027344A1 (fr) 2008-09-04 2008-09-04 Extraits immunomodulateurs de bactéries lactobacillus et procédés de fabrication et d’utilisation de ceux-ci

Publications (1)

Publication Number Publication Date
WO2010027344A1 true WO2010027344A1 (fr) 2010-03-11

Family

ID=41797341

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/010350 WO2010027344A1 (fr) 2008-09-04 2008-09-04 Extraits immunomodulateurs de bactéries lactobacillus et procédés de fabrication et d’utilisation de ceux-ci

Country Status (11)

Country Link
JP (1) JP2012502026A (fr)
KR (1) KR20110051268A (fr)
CN (1) CN102143756A (fr)
AU (1) AU2008361375B2 (fr)
BR (1) BRPI0822997A2 (fr)
CA (1) CA2733249A1 (fr)
IL (1) IL211041A0 (fr)
MX (1) MX2011002080A (fr)
RU (1) RU2500412C2 (fr)
WO (1) WO2010027344A1 (fr)
ZA (1) ZA201100899B (fr)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012103637A1 (fr) * 2011-02-02 2012-08-09 Justbio Inc. Aliments et boissons fonctionnels ayant des propriétés synergiques pour induire l'homéostase
CN103562381A (zh) * 2011-03-29 2014-02-05 雀巢产品技术援助有限公司 缺失d-乳酸产生并且具有改进的贮藏期限的约氏乳杆菌菌株cncm i-1225的衍生物
CN103649306A (zh) * 2011-03-29 2014-03-19 雀巢产品技术援助有限公司 在d乳酸生产中有缺陷以及具有改善的免疫特征的约氏乳杆菌菌株cncm i-1225的天然衍生物
US9713630B2 (en) 2012-01-16 2017-07-25 Labyrinth Holdings, Llc Compositions and methods for the treatment of hepatic diseases and disorders
US9931398B2 (en) 2012-01-16 2018-04-03 Labyrinth Holdings, Llc Naturally-occurring CpG oligonucleotide compositions and therapeutic applications thereof
US10004770B2 (en) 2011-07-21 2018-06-26 Biogaia Ab Production and use of bacterial histamine
WO2020182970A1 (fr) 2019-03-14 2020-09-17 Om Pharma Sa Procédé de fabrication d'extraits bactériens stables et leur utilisation en tant que produits pharmaceutiques
CN112057479A (zh) * 2020-11-16 2020-12-11 招远新兴化工有限公司 一种能够缓解抗生素引起腹泻的益生菌组合物
WO2021142514A1 (fr) * 2020-01-13 2021-07-22 Monash University Compositions et méthodes
US11259539B2 (en) * 2017-06-01 2022-03-01 Meiji Co., Ltd. Method for producing cheese-flavored substance
CN114774302A (zh) * 2022-03-09 2022-07-22 重庆第二师范学院 一种植物乳杆菌cqpc02及其分离方法和应用
US11583560B2 (en) 2017-10-31 2023-02-21 Amorepacific Corporation Immunomodulatory composition comprising lactobacillus-derived extracellular vesicles

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150017208A1 (en) * 2013-07-12 2015-01-15 National Yang-Ming University Lactic acid bacterium having immunomodulatory and anti-allergic effects
JP6491422B2 (ja) * 2013-09-26 2019-03-27 雪印メグミルク株式会社 免疫疾患予防剤
JP6821643B2 (ja) * 2013-09-26 2021-01-27 雪印メグミルク株式会社 免疫疾患予防剤
CN104957260B (zh) * 2014-03-07 2018-07-24 内蒙古伊利实业集团股份有限公司 具有缓解咽喉部炎症功效的益生菌食品组合物及食品
KR101627806B1 (ko) * 2014-06-23 2016-06-07 한국식품연구원 락토바실러스 속 균주의 면역증강 활성을 증가시키는 배양 방법
TWI731209B (zh) * 2018-01-09 2021-06-21 柯順議 益生菌組合物及其用途
BR112020022749A2 (pt) 2018-05-09 2021-03-02 Ko Biolabs, Inc. cepa de lactobacillus paracasei e uso da mesma
CN112469403A (zh) * 2018-06-07 2021-03-09 国立大学法人金泽大学 肾损伤的预防或治疗用的药物组合物
WO2019235590A1 (fr) * 2018-06-07 2019-12-12 国立大学法人金沢大学 Composition pharmaceutique pour la prévention ou le traitement d'une lésion rénale
CN109007842A (zh) * 2018-10-29 2018-12-18 广州普维君健药业有限公司 具有防治过敏功效的益生菌组合物及其应用
CN109628359B (zh) * 2019-02-22 2021-03-02 江南大学 一株可缓解过敏性哮喘的罗伊氏乳杆菌及其应用
KR102156829B1 (ko) * 2019-02-22 2020-09-18 가톨릭대학교 산학협력단 마이크로바이옴 혼합물을 포함하는 면역 질환의 예방 및 치료용 조성물
CN110643541B (zh) * 2019-10-25 2021-08-24 江南大学 一株可调节过敏性哮喘Th2/Th1平衡的干酪乳杆菌及其应用
CN111053789A (zh) * 2019-11-26 2020-04-24 湖南营养树生物科技有限公司 调节免疫系统的方法和组合物
CN111548961A (zh) * 2020-05-15 2020-08-18 郑州和合生物工程技术有限公司 改善过敏性疾病的益生菌组合物、益生菌组合物菌粉及制备方法,应用
CN111991428A (zh) * 2020-06-12 2020-11-27 郑州和合生物工程技术有限公司 一种具有改善哮喘作用的益生菌组合物及其制备方法
KR102424594B1 (ko) * 2020-11-30 2022-07-25 파이토지노믹스 주식회사 항염 활성 및 유해 미생물에 대한 항균 활성을 가지는 락토바실러스 퍼멘텀 okbl-l.fe 1 균주 및 이의 용도
KR20230001127A (ko) 2021-06-28 2023-01-04 농업회사법인 산청바이오진생마을 주식회사 새싹 인삼 라떼 제조방법
KR20230131450A (ko) * 2022-03-03 2023-09-13 한국생명공학연구원 N-카바밀-l-글루탐산을 포함하는 염증성 질환의 치료용 조성물

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3821080A (en) * 1971-06-25 1974-06-28 Nestle Sa Extraction of proteins from microorganism cells
US20050196480A1 (en) * 2004-03-04 2005-09-08 Michael Sullivan Skin treatment method with lactobacillus extract
US7025998B2 (en) * 2003-05-30 2006-04-11 Rotta Research Laboratorium S.P.A. Phytoestrogens and probiotic for women's health
US7235395B2 (en) * 2001-03-02 2007-06-26 Nestec S.A. Lactic acid bacteria as agents for treating and preventing allergy
US7390519B2 (en) * 2001-07-26 2008-06-24 Alimentary Health Limited Probiotic Lactobacillus salivarius strains

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3004890B2 (ja) * 1995-03-28 2000-01-31 雪印乳業株式会社 病原体感染防御剤
RU2302458C2 (ru) * 2001-07-26 2007-07-10 Элиментери Хелс Лимитед Пробиотический штамм lactobacillus salivarius (варианты), пробиотический препарат на их основе и способ лечения или предупреждения с использованием штаммов lactobacillus salivarius
EP2261323A1 (fr) * 2005-05-31 2010-12-15 The Iams Company Lactobacilles probiotiques félins
JP2007269737A (ja) * 2006-03-31 2007-10-18 Morinaga Milk Ind Co Ltd インターロイキン産生調節剤、該インターロイキン産生調節剤を含む医薬組成物及び飲食品、並びにその製造方法
JP5019961B2 (ja) * 2006-06-26 2012-09-05 株式会社ヤクルト本社 インターロイキン10産生促進剤

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3821080A (en) * 1971-06-25 1974-06-28 Nestle Sa Extraction of proteins from microorganism cells
US7235395B2 (en) * 2001-03-02 2007-06-26 Nestec S.A. Lactic acid bacteria as agents for treating and preventing allergy
US7390519B2 (en) * 2001-07-26 2008-06-24 Alimentary Health Limited Probiotic Lactobacillus salivarius strains
US7025998B2 (en) * 2003-05-30 2006-04-11 Rotta Research Laboratorium S.P.A. Phytoestrogens and probiotic for women's health
US20050196480A1 (en) * 2004-03-04 2005-09-08 Michael Sullivan Skin treatment method with lactobacillus extract

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
RHEE ET AL.: "Effect of Environmental pH on Fermentation Balance of Lactobacillus bulgaricus", JOURNAL OF BACTERIOLOGY, vol. 144, no. 1, October 1980 (1980-10-01), pages 217 - 221 *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9770476B2 (en) 2011-02-02 2017-09-26 Justbio Inc. Functional foods and beverages with synergistic properties to promote homeostasis
WO2012103637A1 (fr) * 2011-02-02 2012-08-09 Justbio Inc. Aliments et boissons fonctionnels ayant des propriétés synergiques pour induire l'homéostase
US10028990B2 (en) 2011-02-02 2018-07-24 Justbio Inc. Functional foods and beverages with synergistic properties to promote homeostasis
CN103562381B (zh) * 2011-03-29 2016-04-06 雀巢产品技术援助有限公司 缺失d-乳酸产生并且具有改进的贮藏期限的约氏乳杆菌菌株cncm i-1225的衍生物
CN103649306B (zh) * 2011-03-29 2015-07-01 雀巢产品技术援助有限公司 在d乳酸生产中有缺陷以及具有改善的免疫特征的约氏乳杆菌菌株cncm i-1225的天然衍生物
CN103649306A (zh) * 2011-03-29 2014-03-19 雀巢产品技术援助有限公司 在d乳酸生产中有缺陷以及具有改善的免疫特征的约氏乳杆菌菌株cncm i-1225的天然衍生物
CN103562381A (zh) * 2011-03-29 2014-02-05 雀巢产品技术援助有限公司 缺失d-乳酸产生并且具有改进的贮藏期限的约氏乳杆菌菌株cncm i-1225的衍生物
US10004770B2 (en) 2011-07-21 2018-06-26 Biogaia Ab Production and use of bacterial histamine
US10898529B2 (en) 2011-07-21 2021-01-26 Biogaia Ab Production and use of bacterial histamine
US9713630B2 (en) 2012-01-16 2017-07-25 Labyrinth Holdings, Llc Compositions and methods for the treatment of hepatic diseases and disorders
US9931398B2 (en) 2012-01-16 2018-04-03 Labyrinth Holdings, Llc Naturally-occurring CpG oligonucleotide compositions and therapeutic applications thereof
US10149869B2 (en) 2012-01-16 2018-12-11 Labyrinth Holdings, Llc Compositions and methods for the treatment of hepatic diseases and disorders
US10688177B2 (en) 2012-01-16 2020-06-23 Labyrinth Holdings, Llc Naturally-occurring CpG oligonucleotide compositions and therapeutic applications thereof
US11857577B2 (en) 2012-01-16 2024-01-02 Labyrinth Holdings, Llc Compositions and methods for the treatment of hepatic diseases and disorders
US11259539B2 (en) * 2017-06-01 2022-03-01 Meiji Co., Ltd. Method for producing cheese-flavored substance
US11583560B2 (en) 2017-10-31 2023-02-21 Amorepacific Corporation Immunomodulatory composition comprising lactobacillus-derived extracellular vesicles
WO2020182970A1 (fr) 2019-03-14 2020-09-17 Om Pharma Sa Procédé de fabrication d'extraits bactériens stables et leur utilisation en tant que produits pharmaceutiques
CN113840615A (zh) * 2019-03-14 2021-12-24 Om药物公司 制备稳定细菌提取物的方法及其作为药物的用途
EP4234026A2 (fr) 2019-03-14 2023-08-30 OM Pharma SA Procédé de fabrication d'extraits bactériens stables et leur utilisation en tant que produits pharmaceutiques
US11801271B2 (en) 2019-03-14 2023-10-31 Om Pharma Sa Stable bacterial extracts as pharmaceuticals
WO2021142514A1 (fr) * 2020-01-13 2021-07-22 Monash University Compositions et méthodes
CN112057479A (zh) * 2020-11-16 2020-12-11 招远新兴化工有限公司 一种能够缓解抗生素引起腹泻的益生菌组合物
CN114774302A (zh) * 2022-03-09 2022-07-22 重庆第二师范学院 一种植物乳杆菌cqpc02及其分离方法和应用
CN114774302B (zh) * 2022-03-09 2023-06-16 善恩康生物科技(苏州)有限公司 植物乳杆菌cqpc02在制备用于预防和/或治疗狼疮性肾炎的产品中的应用

Also Published As

Publication number Publication date
MX2011002080A (es) 2011-03-29
JP2012502026A (ja) 2012-01-26
IL211041A0 (en) 2011-04-28
AU2008361375B2 (en) 2014-05-22
RU2500412C2 (ru) 2013-12-10
KR20110051268A (ko) 2011-05-17
AU2008361375A1 (en) 2010-03-11
ZA201100899B (en) 2012-05-30
CN102143756A (zh) 2011-08-03
BRPI0822997A2 (pt) 2015-06-23
CA2733249A1 (fr) 2010-03-11
RU2011112781A (ru) 2012-10-10

Similar Documents

Publication Publication Date Title
AU2008361375B2 (en) Immunomodulatory extracts from Lactobacillus bacteria and methods of manufacturing and use thereof
Sashihara et al. An analysis of the effectiveness of heat-killed lactic acid bacteria in alleviating allergic diseases
Galdeano et al. Mechanisms involved in the immunostimulation by probiotic fermented milk
US9463209B2 (en) Bacterial extract for respiratory disorders and process for its preparation
US9198961B2 (en) Bacterial extract for digestive or urinary tract disorders and process for its preparation
Burns et al. Impact of bile salt adaptation of Lactobacillus delbrueckii subsp. lactis 200 on its interaction capacity with the gut
US20100055082A1 (en) Immunomodulatory extracts from lactobacillus bacteria and methods of manufacturing and use thereof
CN113747908A (zh) 益生菌的治疗性微泡
EP2161329A1 (fr) Extraits immunomodulateurs de bactéries lactobacillus et leurs procédés de fabrication et d'utilisation
Velez et al. Modulation of gut immune response by probiotic fermented milk consumption to control IgE in a respiratory allergy model
TW201010711A (en) Immunomodulatory extracts from lactobacillus bacteria and methods of manufacturing and use thereof
AU2013200193B2 (en) Bacterial extract for digestive or urinary tract disorders and process for its preparation
Dimitrov et al. Selected lactic acid bacteria strains reduce the expression of interleukin-8 in epithelial cells.
ITMI20070597A1 (it) Metodo di frazionamento di organismi unicellulari e prodotti ottenuti da tale processo

Legal Events

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

Ref document number: 200880130960.2

Country of ref document: CN

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

Ref document number: 08815598

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 211041

Country of ref document: IL

Ref document number: 839/DELNP/2011

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2008361375

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2733249

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2008361375

Country of ref document: AU

Date of ref document: 20080904

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2011/002080

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2011526017

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20117007527

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2011112781

Country of ref document: RU

122 Ep: pct application non-entry in european phase

Ref document number: 08815598

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: PI0822997

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110228