[go: up one dir, main page]

WO2025158027A1 - 4-hydroxyphenyllactic acid for use in treating and reducing the risk of ige-mediated diseases - Google Patents

4-hydroxyphenyllactic acid for use in treating and reducing the risk of ige-mediated diseases

Info

Publication number
WO2025158027A1
WO2025158027A1 PCT/EP2025/051848 EP2025051848W WO2025158027A1 WO 2025158027 A1 WO2025158027 A1 WO 2025158027A1 EP 2025051848 W EP2025051848 W EP 2025051848W WO 2025158027 A1 WO2025158027 A1 WO 2025158027A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
composition
use according
ige
infant
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
PCT/EP2025/051848
Other languages
French (fr)
Inventor
Susanne Brix PEDERSEN
Rasmus Ibsen DEHLI
Pernille Neve MYERS
Carsten Eriksen
Anna Hammerich THYSEN
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Danmarks Tekniske Universitet
Original Assignee
Danmarks Tekniske Universitet
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Danmarks Tekniske Universitet filed Critical Danmarks Tekniske Universitet
Publication of WO2025158027A1 publication Critical patent/WO2025158027A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/405Indole-alkanecarboxylic acids; Derivatives thereof, e.g. tryptophan, indomethacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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

Definitions

  • the present invention relates to the use of compositions comprising 4- hydroxyphenyllactic acid for the prevention and/or treatment of IgE-mediated diseases.
  • the gut microbiota can impact host physiology in various ways via the production of metabolites. This can result in biochemical, immune and metabolic changes in the gut. Indeed, depletion of specific microbes, such as Bifidobacterium, in early life has been associated with increased risk of development of IgE-mediated diseases, such as allergy and asthma, in childhood, and suggested to compromise immune function and susceptibility to infectious disease. Consequently, there is a significant interest in pinpointing microbial elements that can promote a more robust imprinting of the immune system, potentially preventing the onset of allergies, autoimmunity, and other conditions related to immune function (Renz & Skevaki, 2020).
  • IgE-mediated diseases such as allergy and asthma
  • therapies such as antihistamines, corticosteroids, bronchodilators and immunomodulators primarily focus on alleviating symptoms rather than addressing the underlying causes of IgE-mediated diseases.
  • currently available therapeutics for IgE-mediated diseases are associated with highly variable patient responses, limited efficacy in severe cases and in some cases, such as with systemic corticosteroids, may exhibit immunosuppressive effects that can increase susceptibility to infections and other complications. Therefore, there is an unmet need for efficacious therapeutics for IgE-mediated diseases which target their fundamental aetiology. Summary
  • the present invention is directed to compositions for use to prevent and/or treat IgE-mediated diseases.
  • Aldh+ Bifidobacterium species convert aromatic amino acids (tryptophan, phenylalanine and tyrosine) into their respective aromatic lactic acids (indolelactic acid, phenyllactic acid and 4-hydroxyphenyllactic acid) via the aromatic lactate dehydrogenase (ALDH) enzyme (Laursen et al., 2021).
  • ADH aromatic lactate dehydrogenase
  • IgE is produced in plasma B cells following cross-linking of B cell receptors (BCR) by allergens and stimulation via IL-4.
  • BCR B cell receptors
  • the present invention is directed to compositions comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof, formula (II), wherein;
  • A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
  • the present invention is directed to a compound of formula (II) or a pharmaceutically acceptable salt thereof, formula (II), wherein;
  • A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in a method of reducing the risk of IgE-mediated disease in a subject, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or a systemic allergen-specific IgE level above 0.35 kll/L, and, b. Administering a composition comprising one or more compounds of formula (II) or a pharmaceutically acceptable salt thereof to the subject to achieve a total level of aromatic lactic acids in plasma of at least 5 pM.
  • Figure 1 A high gut bacterial load between week 1 and 6 months of age is linked to decreased incidence of IgE-mediated diseases later in life.
  • A Bacterial load in fecal samples of children from 3-6 days (1w), 3 weeks, 2 months, 6 months, 12 months, 18 months, 24 months (2 years), and 60 months (5 years) postpartum and in fecal samples from their mothers 2 months postpartum (Mother). Dots represent individual samples, and the line represents median abundance.
  • (B-E) Mean bacterial load in early life fecal samples (1 week to 6 months) of children which are not (No)/are (Yes), (B) sensitized to food allergens ⁇ 5 years postpartum (37/18), (C) diagnosed with atopic dermatitis as defined by SCORAD at 2 years of age (51/4), (D) repeatedly prescribed asthma medication ⁇ 14 years postpartum (45/11) and (E) repeatedly prescribed allergic rhinitis medication ⁇ 14 years postpartum (45/11). Dots represent individual samples. Statistics: Welch two-sample t-test.
  • B-E Maximum load of bifidobacterial species in early life (from 1 week to 6 months) in fecal samples from children which are not (No)/are (Yes)
  • B sensitized to food allergens ⁇ 5 years postpartum (37/18)
  • C diagnosed with atopic dermatitis as defined by SCORAD at 2 years of age (51/4)
  • D repeatedly prescribed asthma medication ⁇ 14 years postpartum (45/11)
  • E repeatedly prescribed allergic rhinitis medication ⁇ 14 years postpartum (45/11). Dots represent individual samples.
  • C-D Maximum fecal levels of 4-OH-PLA between 2 and 6 months in children which are not (No)/are (Yes)
  • C repeatedly prescribed asthma medication ⁇ 14 years postpartum (45/11) and
  • D repeatedly prescribed allergic rhinitis medication ⁇ 14 years postpartum (45/11).
  • E-F Fecal levels of PLA at 2 months in children which are not (No)/are (Yes)
  • E sensitized to food allergens ⁇ 5 years postpartum (79/59) and (F) diagnosed with atopic dermatitis at 2 years of age (125/12).
  • G-H Maximum fecal levels of PLA between 2 and 6 months in children which are not (No)/are (Yes)
  • G repeatedly prescribed asthma medication ⁇ 14 years postpartum (45/11) and
  • H repeatedly prescribed allergic rhinitis medication ⁇ 14 years postpartum (45/11).
  • I-J Fecal levels of ILA at 2 months in children which are not (No)/are (Yes)
  • I sensitized to food allergens ⁇ 5 years postpartum (77/59) and (J) diagnosed with atopic dermatitis at 2 years of age (123/12).
  • Figure 4 The metabolite 4-OH-PLA completely mediates the association of aldh+ bifidobacteria with decreased incidence of food allergen sensitization, while PLA does not.
  • the estimate corresponds to the change in log odds of food allergen sensitization until 5 years of age per one-unit increase. Accordingly, a 100 nmol/g feces (100-unit) increase in 2 months faecal 4-OH-PLA levels is estimated to significantly decrease the odds of developing food allergen sensitization during the first 5 years of life by 15% (C) Total effect and mediation model, including tests of significant associations proceeding the mediation analyses.
  • Association between max early life (0-6m) fecal aldh+ relative abundance and food allergen sensitization until 5 years of age (path c) and 2m fecal 4-OH-PLA or PLA levels and food allergen sensitization (path b) were tested with a generalized linear model.
  • the 2m fecal 4-OH-PLA or PLA levels were predicted by max early life (0-6m) fecal aldh+ relative abundance (path a) using a linear model.
  • Path c corresponds to the total effect in the mediation analyses.
  • FIG. 1 Levels of individual aromatic lactic acids (ALAs) and total ALA levels in fecal samples at 2 months of age in children with later development of food allergen sensitization (until 5 years of age) or diagnosis of atopic dermatitis at 2 years and at maximum 2-6 months of age in children with repeated prescriptions of asthma or allergic rhinitis medication until 14 years of age.
  • A Levels of the three ALAs and their sum in feces at 2 months of age in children that later are not (No)/are (Yes) i) sensitized to food allergens ⁇ 5 years postpartum and ii) diagnosed with atopic dermatitis as determined by SCORAD.
  • ILA levels detected in plasma at 6 months of age correlate with relative abundance of fecal aldh+ bacteria and with fecal ILA levels at 6 months of age.
  • FIG. 7 Both ILA and 4-OH-PLA decrease IgE production in vitro in blood- derived human plasma B cells.
  • Freshly isolated human blood-derived peripheral blood mononuclear cells were stimulated for 10 days with 50 ng/mL CD40L, 500 ng/mL cross-linking HA antibody and 50 ng/mL rhlL-4 to induce IgE production in plasma B cells.
  • Co-incubation with 200 pM ILA or 4-OH-PLA resulted in at least 50% inhibition of IgE production. Data represent mean +/- SEM of assay replicates in two different subjects.
  • Cryopreserved human blood- derived peripheral blood mononuclear cells were thawed and stimulated for 10 days with 50 ng/mL CD40L, 100 ng/mL cross-linking HA antibody and 50 ng/mL rhlL-4 to induce IgE production in plasma B cells.
  • Co-incubation with doses of 5, 50, 200 and 1000 pM ILA or 4-OH-PLA resulted in varying in vitro IgE inhibition patterns.
  • Human blood-derived peripheral blood mononuclear cells were stimulated for 10 days with 50 ng/mL CD40L, 100 ng/mL cross-linking HA antibody and 50 ng/mL rhlL-4 to induce IgE production in plasma B cells.
  • Co-incubation with high concentrations of 200 pM ILA or 200 pM 4-OH-PLA resulted in consistent in vitro inhibition of IgE production, while 200 pM D-PLA and L-PLA showed inconsistent effects across subjects, even at this high concentration. Cell viability was unaltered at these compound concentrations. Data represent mean +/- SEM of replicates from 6 subjects.
  • administering refers to any method of providing an agent to the subject (e.g., an IgE-reducing agent).
  • agents e.g., an IgE-reducing agent
  • Such methods include, but are not limited to, oral administration, transdermal administration, administration by inhalation, nasal administration, topical administration, intravaginal administration, ophthalmic administration, intra-aural administration, intracerebral administration, administration to spinal cord, administration to intracerebral fluid, rectal administration, and parenteral administration, including injectable such as intravenous administration, intra-arterial administration, intramuscular administration, and subcutaneous administration. Administration can be continuous or intermittent. In some instances an IgE-reducing agent can be administered therapeutically.
  • an IgE-reducing agent can be administered prophylactically, such as administered for prevention of a disease or condition in a subject, or for improvement of one or more immune cell (e.g., basophil) functions in a subject (e.g., in the lung of a subject).
  • the terms "effective amount” or “amount effective” or “therapeutically effective amount” refer to an amount that is sufficient to achieve the desired result (e.g., therapeutic benefit) or to have an effect on an undesired condition.
  • a “therapeutically effective amount” of a composition of the present invention may refer to an amount that is sufficient to achieve the desired result or to have an effect on a disease in a subject.
  • a "therapeutically effective amount" of a composition of the present invention may refer to an amount of the compositions of the present invention that is sufficient to reduce the level and/or activity of IgE in a subject (e.g., in a biological sample obtained from the subject), or to switch one or more functions of an immune cell (e.g., B cells) in a subject to whom a composition of the present invention is administered.
  • an immune cell e.g., B cells
  • the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including: the disorder being treated and the severity of the disorder; the age, body weight, general health, sex, diet, ethnic group and/or geographical location of the subject; the time of administration; the route of administration; the rate of excretion of the composition of the present invention employed; the duration of the treatment; drugs used in combination or coincidental with the specific composition of the present invention employed and like factors known in the medical arts.
  • the effective daily dose can be divided into multiple doses for purposes of administration. Consequently, single dose compositions can contain such amounts or submultiples thereof to make up the daily dose.
  • the dosage can be adjusted by the individual physician in the event of any contraindications. Dosage can vary, and can be administered in one or more dose administrations daily, for one or several days. In some instances, a composition of the present invention can be administered in a prophylactically effective amount.
  • the term “decreased level” or “decreased concentration” or “decreased” refers to a level that is detectably lower (e.g., by about 5-10 percent, 10-20 percent, 20-30 percent, 30-40 percent, 40-50 percent, 50- 60 percent, 60-70 percent, 70-80 percent, 80-90 percent, 85-95 percent, or more; such as, by about 5 percent, 10 percent, 15 percent, 20 percent, 25 percent, 30 percent, 35 percent, 40 percent, 45 percent, 50 percent, 55 percent, 60 percent, 65 percent, 70 percent, 75 percent, 80 percent, 85 percent, 90 percent, 95 percent, 97 percent, 99 percent, or more) compared to a reference level.
  • decreased IgE production in a subject may refer to detectably lower (e.g., by about 5- 10 percent, 10-20 percent, 20-30 percent, 30-40 percent, 40-50 percent, 50-60 percent, 60- 70 percent, 70-80 percent, 80-90 percent, or 85-95 percent, or more; such as, by about 5 percent, 10 percent, 15 percent, 20 percent, 25 percent, 30 percent, 35 percent, 40 percent, 45 percent, 50 percent, 55 percent, 60 percent, 65 percent, 70 percent, 75 percent, 80 percent, 85 percent, 90 percent, 95 percent, 97 percent, 99 percent, or more) level of IgE compared to a reference level of IgE.
  • level of IgE compared to a reference level of IgE.
  • treatment and/or prevention is to be understood as the treatment and/or prevention and/or reduction and/or alleviation of the indicated pathology, pathophysiological process and/or disease, and/or the treatment and/or prevention and/or reduction and/or alleviation of the risk factors for the indicated pathology, pathophysiological process and/or disease.
  • HCAR2 is to be understood as Hydroxycarboxylic acid receptor 2, also known as GPR109A, niacin receptor 1 (NIACR1), HCA2, HM74a, HM74b, NIACR1, PUMAG, and Puma-g.
  • HCAR3 Hydroxycarboxylic acid receptor 3, also known as niacin receptor 2 (NIACR2) and GPR109B.
  • AhR is to be understood as aryl hydrocarbon receptor, also known as AHR, ahr, ahR, AH receptor, or dioxin receptor.
  • Bifidobacteria is to be understood as microorganisms belonging to the genus Bifidobacterium. Examples include but are not limited to Bifidobacterium adolescentis, B. angulatum, B. animalis subsp. animalis, B. animalis subsp. lactis, B. asteroides, B. bifidum, B. bourn, B. breve, B. catenulatum, B. choerinum, B. coryneforme, B. cuniculi, B. denticolens, B. dentium, B. gallium, B. gallinarum, B. globosum; B.
  • indicum B. infantis, B. inopinatum, B. lactis, B. longum, B. magnum, B. merycicum, B. minimum, B. parvulorum, B. pseudocatenulatum, B. pseudoIongum subsp. globosum, B. pseudoIongum subsp. pseudoIongum, B. pullorum, B. ruminale, B. ruminantium, B. saeculare, B. scardovii, B. subtile, B. suis, B. thermacidophilum, and B. thermophilum.
  • Aldh+ bifidobacteria is to be understood as any microorganism belonging to the genus Bifidobacterium which contains the gene encoding the enzyme aromatic lactate dehydrogenase (Aldh), as defined in SEQ ID NO: 1.
  • ALAs are metabolites synthesized by certain bacteria, for example by microorganisms belonging to the genus Bifidobacterium, from dietary amino acids.
  • ALAs are indole-3-lactic acid (ILA), 4-hydroxyphenyllactic acid (4-OH-PLA), D-phenyllactic acid (D-PLA) and L- phenyllactic acid (L-PLA).
  • gut microbiota is to be understood as all the bacteria and other microorganisms within the gut.
  • gut microbiota dysbiosis refers to an imbalance in the relative abundance or presence of microbes (e.g., beneficial and/or pathogenic microbes) in the gut of a subject that can result in a variety of symptoms including, for example, one or more of bloating, flatus, spasms, inflammation with loss of intestinal permeability, dysplasia of a mucosal surface, insufficient reclamation of nutrients for buffering capacity or increasing risk for development of autoimmune or IgE-mediated diseases.
  • Dysbiosis can include a loss of beneficial microbes and/or an expansion of pathogenic microbes (e.g., pathobionts). Dysbiosis is believed to trigger pro-inflammatory effects and immune dysregulation associated with various disease states.
  • the term “pharmaceutical composition” refers to the combination of an active agent (e.g., a composition of the present invention) with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo.
  • the term “pharmaceutical composition” can be a formulation containing a composition of the present invention and/or an agent for reducing IgE production of the present disclosure in a form suitable for administration to a subject.
  • the pharmaceutical composition is in bulk or in unit dosage form.
  • the unit dosage form is any of a variety of forms, including, for example, a capsule, an intravenous bag, a tablet, a single pump on an aerosol inhaler or a vial.
  • the quantity of active ingredient in a unit dose of composition is an effective amount and can vary depending upon the particular treatment involved.
  • active ingredient e.g., a formulation of a disclosed agent for reducing IgE production alone or together with other immune modulating agents, as well as salt, hydrate, solvate or isomer thereof
  • the dosage will also depend on the route of administration. A variety of routes are contemplated, including, for example, oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like.
  • Dosage forms for the topical or transdermal administration of an IgE-reducing agent alone or together with other immune modulating agents include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • an IgE-reducing agent is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that are required.
  • an IgE-reducing agent is one or more compounds according to Formula I or Formula II as disclosed and defined herein.
  • pharmaceutically acceptable carrier refers to buffers, carriers, and excipients suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable carriers include any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions (e.g., such as an oil/water or water/oil emulsions), and various types of wetting agents.
  • Pharmaceutically acceptable carriers include buffers, solvents, dispersion media, coatings, isotonic and absorption delaying agents, and the like, that are compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is known in the art.
  • compositions of the present invention are used interchangeably and refer to the amount of a bioactive agent or combination of bioactive agents present in one or more pharmaceutical compositions as described herein that is needed to provide a desired level of active agent or agents in the bloodstream or at the site of action in an subject (e.g., the hepatic system, the renal system, the circulatory system, the lungs, the gastrointestinal system, the colorectal system, the vagina etc.) to be treated to give an anticipated physiological response when such composition is administered.
  • a bioactive agent or combination of bioactive agents present in one or more pharmaceutical compositions as described herein that is needed to provide a desired level of active agent or agents in the bloodstream or at the site of action in an subject (e.g., the hepatic system, the renal system, the circulatory system, the lungs, the gastrointestinal system, the colorectal system, the vagina etc.) to be treated to give an anticipated physiological response when such composition is administered.
  • immune modulating agent refers to nutrients that have been identified as critical for the growth and function of immune cells, for example vitamins and minerals.
  • the immune modulating agent may be a vitamin such as Vitamin A, C, D, B, E, p-carotene and others, and/or minerals such as zinc, magnesium, selenium, iron and others.
  • Allergen refers to a substance that may elicit an immune reaction (hypersensitive response such as antibody formation) in a subject. Common examples include, but are not limited to: grasses, pollen, milk, egg, seafood (such as shrimp), nuts, bee venom, drugs (such as penicillin), and natural rubber latex proteins.
  • immunoglobulin E refers to an immunoglobulin of class E which may be involved in a hypersensitivity response.
  • sample refers to biological material containing analytes to be assayed.
  • Biological material may be obtained from a subject and may include, for example, tissue, cells, feces, or bodily fluid.
  • the sample is a bodily fluid.
  • Sample can refer to any fluid that may be obtained from a subject, including, but not limited to saliva, blood, plasma, serum, feces, urine, interstitial fluid, lymph, gastric juices, bile, sweat, and spinal and brain fluids, including fluids that are processed or present in their natural form.
  • a “sample” from a subject is one that may contain circulating IgE.
  • a “sample” from a subject is one that is suspected to contain suboptimal levels of ALAs.
  • alkyl refers to a linear or branched hydrocarbon moiety.
  • cycloalkyl refers to a monocylcic or polycyclic system.
  • cycloalkyl also used herein can optionally contain one or more unsaturations or substituents.
  • heteroaryl refers to an aromatic ring containing from 5 to 6 ring atoms where at least one of the ring atoms are heteroatom(s).
  • heteroatom is intended to mean sulfur, oxygen or nitrogen.
  • aromatic refers to a cyclic or polycyclic moiety having a conjugated unsaturated (4r
  • stereochemistry configuration refers to the absolute configuration of that center, which can be R or S, as determined by the Cahn-lngold-Prelog priority rules.
  • the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, formula (I) wherein;
  • R 1 represents hydroxy group or -H
  • R 2 represents an alkyl, cycloalkyl, aryl or heteroaryl group, or -H,
  • A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
  • R 2 is selected from the group consisting of C1-C10 alkyl optionally substituted with one or more, identical or different, substituents, or a Ca-Cs cycloalkyl optionally substituted with one or more, identical or different, substituents.
  • the present invention relates to a compound of formula (II) or a pharmaceutically acceptable salt thereof, formula (II), wherein; A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
  • the compound is not phenyllactic acid.
  • R1 represents hydroxy
  • A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring.
  • A represents an unsubstituted indole or; wherein A represents; _
  • A represents; , wherein R is hydroxy.
  • A represents an unsubstituted indole.
  • R1 represents a -H, represents a double bond
  • A represents a substituted aromatic ring.
  • A represents; wherein R 3 is hydroxy
  • R 4 is methoxy, hydroxy or -H
  • R 5 is methoxy, hydroxy or -H.
  • A represents; wherein R 3 is hydroxy
  • R 4 is methoxy or-H
  • R 5 is methoxy or-H
  • A represents; wherein R 3 is hydroxy, R 4 is methoxy, R 5 is-H.
  • A represents; wherein R 3 is hydroxy
  • R 4 and R 5 are methoxy or hydroxy.
  • A represents; wherein R 3 is hydroxy, R 4 and R 5 are methoxy.
  • the compound binds hydroxycarboxylic acid receptor 3 (HCAR3), aryl hydrocarbon receptor (AHR), and/or hydroxycarboxylic acid receptor 2 (HCAR2).
  • the compound is an agonist of HCAR3, AHR and/or HCAR2receptors.
  • the compound has the structure of formula (1a) formula (1a), or a pharmaceutically acceptable salt thereof.
  • the compound has the structure of formula (1b) formula (1b), or a pharmaceutically acceptable salt thereof.
  • the compound does not have the structure of formula (1c) formula (1c).
  • the compound has the structure of formula (1d) formula (1 d), or a pharmaceutically acceptable salt thereof.
  • the compound has the structure of formula (1e) formula (1e), or a pharmaceutically acceptable salt thereof.
  • the compound has the structure of formula (1f) formula (1f), or a pharmaceutically acceptable salt thereof.
  • the present invention relates to a composition comprising or consisting of one or more compounds of formula (I) or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
  • the present invention relates to a composition comprising or consisting of one or more compounds of formula (II) or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
  • the composition comprises, or consists of; a. the compound of formula (1 a) formula (1a), or a pharmaceutically acceptable salt thereof, and/or b. the compound of formula (1 b) formula (1b), or a pharmaceutically acceptable salt thereof.
  • the composition comprises, or consists of; a. the compound of formula (1 a) formula (1a), or a pharmaceutically acceptable salt thereof, and b. the compound of (1 b) formula (1b), or a pharmaceutically acceptable salt thereof.
  • the composition comprises, or consists of; a. the compound of (1a) formula (1a), or a pharmaceutically acceptable salt thereof, or b. the compound of (1 b) formula (1 b), or a pharmaceutically acceptable salt thereof.
  • the compound is 4-hydroxyphenyllactic acid or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is indole-3-lactic acid or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is not phenyllactic acid. In some embodiments, the composition comprises 4-hydroxyphenyllactic acid and indole-3-lactic acid. In some embodiments, the composition comprises is 4-hydroxyphenyllactic acid or indole-3- lactic acid.
  • the compound is p-coumaric acid or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is Sinapinic acid or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is Ferulic acid or a pharmaceutically acceptable salt thereof.
  • the subject is a human. In some embodiments, the subject is a non-infant. In some embodiments, the subject is an infant. In some embodiments, the subject is a pre-term infant or a term infant. In some embodiments, the infant is an infant born by C-section. In some embodiments, the infant is born from a first-time mother. In some embodiments, the infant has one or both parents affected by an IgE-mediated disease.
  • the infant has a dysbiotic gut microbiota. In some embodiments, the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a low abundance of Aldh+ bacteria. In some embodiments, the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a low abundance of Aldh+ bacteria, wherein said Aldh+ bacteria comprises an Aldh gene encoding the enzyme aromatic lactate dehydrogenase, preferably an Aldh of SEQ ID NO: 1.
  • the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has an abundance of Aldh+ bacteria of at most 10% of total bacteria, such as at most 9% of total bacteria, such as at most 8% of total bacteria, such as at most 7% of total bacteria, such as at most 6% of total bacteria, such as at most 5% of total bacteria, such as at most 4% of total bacteria, such as at most 3% of total bacteria, such as at most 2% of total bacteria, such as at most 1% of total bacteria.
  • Aldh+ bacteria of at most 10% of total bacteria, such as at most 9% of total bacteria, such as at most 8% of total bacteria, such as at most 7% of total bacteria, such as at most 6% of total bacteria, such as at most 5% of total bacteria, such as at most 4% of total bacteria, such as at most 3% of total bacteria, such as at most 2% of total bacteria, such as at most 1% of total bacteria.
  • the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has an abundance of Aldh+ bacteria of at most 5% of total bacteria. In some embodiments, the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a low load of Aldh+ bacteria. In some embodiments, the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a load of Aldh+ bacteria of at most 3x10 9 Aldh+ bacteria/g feces.
  • the infant is between 3 weeks and 6 months of age, such as between 3 weeks and 5 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 3 months of age, such as between 3 weeks and 2 months of age, such as between 3 weeks and 1 month of age, such as between 1 month and 6 months of age, such as between 2 month and 6 months of age, such as between 3 month and 6 months of age, such as between 4 month and 6 months of age.
  • the infant has at most 3x10 9 Aldh+ bacteria/g feces. In some embodiments, the infant has at most 100 nmol/g feces of at least one aromatic lactic acid. In some embodiments, the infant has at most 100 nmol/g feces of at least one aromatic lactic acid, corresponding to a plasma concentration of at most 3 pM.
  • the infant has at most 100 nmol/g feces of at least one aromatic lactic acid, wherein the concentration of the at least one aromatic lactic acid is determined by LC-MS.
  • the infant has at most 100 nmol/g feces of at least one aromatic lactic acid, such as is 4-hydroxyphenyllactic acid and/or indole-3-lactic acid and/or phenyllactic acid. In some embodiments, the infant has at most 100 nmol/g feces of the compound of formula (1a), formula (1a), in the gut. In some embodiments, the infant has at most 100 nmol/g feces of 4- hydroxyphenyllactic acid in the gut. In some embodiments, the infant has at most 100 nmol/g feces of 4-hydroxyphenyllactic acid in the gut at between 3 weeks and 6 months of age, such as between 2-5 months of age.
  • the infant has less than 100 nmol/g feces of the compound of formula 1b, formula (1b), in the gut.
  • the infant has less than 100 nmol/g feces of indole-3-lactic acid in the gut.
  • the infant has less than 100 nmol/g feces of the compound of formula (1c), formula (1c), in the gut.
  • the infant has less than 100 nmol /g feces of phenyllactic acid in the gut.
  • the infant has less than 100 nmol/g feces of the compound or indole-3-lactic acid in the gut between 3 weeks and 6 months of age, such as between 3 weeks and 5 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 3 months of age, such as between 3 weeks and 2 months of age, such as between 3 weeks and 1 month of age, such as between 1 month and 6 months of age, such as between 2 month and 6 months of age, such as between 3 month and 6 months of age, such as between 4 month and 6 months of age.
  • 3 weeks and 6 months of age such as between 3 weeks and 5 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 3 months of age, such as between 3 weeks and 2 months of age, such as between 3 weeks and 1 month of age, such as between 1 month and 6 months of age, such as between 2 month and 6 months of age, such as between 3 month and 6 months
  • the compound is administered in an amount which is effective to decrease IgE production. effect
  • the compound decreases IgE production in human B cells and/or plasma cells. In some embodiments, the compound decreases IgE production in human B cells and/or plasma cells to at most 25 ng/mL.
  • the compound decreases IgE production in human B cells and/or plasma B cells, wherein IgE production is determined by human B cell assays either alone or together with other cells.
  • the compound decreases IgE production in human B cells.
  • the compound decreases IgE production in human B cells to at most 25 ng/mL.
  • the compound decreases IgE production in plasma B cells.
  • the compound decreases IgE production in plasma B cells to at most 25 ng/mL.
  • the compound decreases systemic IgE production to below 0.50 kU/L plasma IgE. In some embodiments, the compound decreases systemic IgE production to below 0.50 kU/L plasma IgE, such as to 0.35 or below kU/L, such as to 0.1 kU/L or below. In some embodiments, the compound decreases systemic IgE production to 0.50 kU/L plasma allergen-specific IgE or less.
  • the compound decreases systemic IgE production to 0.50 kU/L plasma allergen-specific IgE or below, such as to 0.35 kU/L or below, such as to about 0.1 kU/L. In some embodiments, IgE production in human B cells and/or plasma B cells is decreased systemically to about 0.1 kU/L, such as to 0.35 kU/L or below.
  • the IgE-mediated disease is an allergic response to an allergen.
  • the allergen is a pollen, a dust mite, a food allergen, a plant allergen, animal dander, insect stings, a fungus, a spore, a mold, latex, or a drug.
  • the IgE-mediated disease is allergic asthma, urticaria, angioedema, food allergy, an allergic response, atopic dermatitis, anaphylaxis, cutaneous mastocystosis, allergic rhinitis, allergic rhinoconjunctivitis, nasal polyposis, Kimura's disease, eosinophilic otitis media, eosinophilic gastroenteritis, latex allergy, bronchopulmonary allergic aspergillosis.
  • the IgE-mediated disease is acute or chronic.
  • the compound or composition is administered to the subject prophylactically. In some embodiments, the compound or composition is administered to the subject orally and/or parenterally. In some embodiments, the compound or composition is administered to the subject orally. In some embodiments, the compound or composition is administered to the subject parenterally. In some embodiments, the compound or composition is administered to the subject via intramuscular, and/or subcutaneous and/or IV administration. In some embodiments, the compound or composition is administered with at least one or more therapeutic agent(s). In some embodiments, the at least one or more therapeutic agent(s) is the standard of care treatment for IgE-mediated disease. In some embodiments, the at least one or more therapeutic agent(s) is administered simultaneously, sequentially or separately.
  • the composition is a pharmaceutical composition. In some embodiments, a therapeutically effective amount of the composition is administered to the subject. In some embodiments, a therapeutically effective amount of the compound or composition is administered to the infant.
  • the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in a subject, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kll/L, and, b. Administering a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM.
  • the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in a subject, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kll/L, and, b.
  • the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in an infant, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the infant, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM.
  • the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in an infant, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the infant until a level of at least 5 pM of one or more aromatic lactic acids in plasma is achieved until 6 months of age of the infant.
  • the present invention relates to a method of reducing the risk of IgE- mediated disease in a subject, comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kll/L, and, b. Administering a compound of formula (I) to the subject thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM and/or individual allergen-specific plasma IgE levels below 0.5 kll/L.
  • the present invention relates to a method of reducing the risk of IgE- mediated disease in a subject, comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut, and, b. Administering a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the subject to achieve a total level of aromatic lactic acids in plasma of at least 5 pM and/or until plasma IgE levels are at most 0.5 kll/L.
  • the present invention relates to a method of reducing the risk of IgE- mediated disease in an infant, comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the infant thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM.
  • the present invention relates to method of reducing the risk of IgE- mediated disease in an infant, comprising: a.
  • a reduced total level of aromatic lactic acids in the infant gut corresponds to, and is measured by, a level of one or more of the ALAs described herein of 100 nmol/g feces or less, as defined herein.
  • allergen-specific plasma IgE level uses an allergen-specific plasma IgE level above 0.35 kll/L as cutoff, wherein the assay is positive if the allergen-specific plasma IgE level is above 0.35 kll/L.
  • a subject presenting symptoms of IgE-mediated disease such as allergic asthma, urticaria, angioedema, food allergy, an allergic response, atopic dermatitis, anaphylaxis, cutaneous mastocystosis, allergic rhinitis, allergic rhinoconjunctivitis, nasal polyposis, Kimura's disease, eosinophilic otitis media, eosinophilic gastroenteritis, latex allergy, bronchopulmonary allergic aspergillosis, typically have allergen-specific IgE levels of 1 kll/L and above.
  • step a. comprises identifying an allergen-specific plasma IgE level of 0.5 kU/L or higher, such as of 1 kU/L or higher, such as of 10 kU/L or higher.
  • Reducing an allergen-specific plasma IgE level to 0.5 kU/L or less corresponds to reaching an allergen-specific plasma IgE level at which a subject does not feel, experience or show any allergic symptoms.
  • the administered compound is a compound of formula (la) or formula (lb), or a combination thereof.
  • the aromatic lactic acids are selected from the group consisting of 4-hydroxyphenyllactic acid, indole-3-lactic acid and/or phenyllactic acid.
  • the present invention relates to a method for the treatment and/or prevention of an IgE-mediated disease comprising administration of a therapeutically effective amount of a composition comprising or consisting of a compound of formula (I) or a pharmaceutically acceptable salt thereof, formula (I) wherein;
  • R 1 represents hydroxy group or -H
  • R 2 represents an alkyl, cycloalkyl, aryl or heteroaryl group, or -H,
  • A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, to a subject in need thereof.
  • the present invention relates to use of a composition
  • a composition comprising or consisting of a compound of formula (I) or a pharmaceutically acceptable salt thereof, formula (I) wherein;
  • R 1 represents hydroxy group or -H
  • R 2 represents an alkyl, cycloalkyl, aryl or heteroaryl group, or -H, represents a single or double bond
  • A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for the manufacture of a medicament for the treatment and/or prevention of an IgE- mediated disease.
  • a therapeutically effective amount of the compound is administered to the subject. In one embodiment, a therapeutically effective amount of the compound is administered to the infant.
  • R 1 represents hydroxy group or -H
  • R 2 represents an alkyl, cycloalkyl, aryl or heteroaryl group, or -H, ' represents a single or double bond,
  • A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
  • R 2 is selected from the group consisting of C1-C10 alkyl optionally substituted with one or more, identical or different, substituents, or a Ca-Cs cycloalkyl optionally substituted with one or more, identical or different, substituents.
  • A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
  • R 2 is selected from the group consisting of C1-C10 alkyl optionally substituted with one or more, identical or different, substituents, or a Ca-Cs cycloalkyl optionally substituted with one or more, identical or different, substituents.
  • R1 represents hydroxy
  • A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring.
  • R1 represents a -H, represents a double bond
  • A represents a substituted aromatic ring.
  • A represents; wherein R 3 is hydroxy
  • R 4 is selected from the group consisting of methoxy, hydroxy and -H
  • R 5 is selected from the group consisting of methoxy, hydroxy and -H.
  • A represents; wherein R 3 is hydroxy, R 4 is methoxy, R 5 is -H.
  • A represents; wherein R 3 is hydroxy, R 4 and R 5 are methoxy.
  • HCAR3 hydroxycarboxylic acid receptor 3
  • AHR aryl hydrocarbon receptor
  • HCAR2 hydroxycarboxylic acid receptor 2
  • compositions for use in the treatment and/or prevention of an IgE-mediated disease in a subject comprising one or more compounds of formula (I).
  • R 2 is selected from the group consisting of C1-C10 alkyl optionally substituted with one or more, identical or different, substituents, or a Ca-Cs cycloalkyl optionally substituted with one or more, identical or different, substituents.
  • compositions for use in the treatment and/or prevention of an IgE-mediated disease in a subject comprising one or more compounds of formula (II).
  • composition for use according to any one of the preceding items wherein the composition comprises or consists of; a. the compound of formula (1 a) formula (1a), or a pharmaceutically acceptable salt thereof, and/or b. the compound of formula (1 b) formula (1 b), or a pharmaceutically acceptable salt thereof.
  • composition for use according to any one of the preceding items wherein the composition comprises or consists of; a. the compound of formula (1 a) formula (1a), or a pharmaceutically acceptable salt thereof, and b. the compound of (1 b) formula (1b), or a pharmaceutically acceptable salt thereof.
  • composition for use according to any one of the preceding items wherein the composition comprises or consists of; a. the compound of (1a) formula (1a), or a pharmaceutically acceptable salt thereof, or b. the compound of (1 b) formula (1b), or a pharmaceutically acceptable salt thereof.
  • the compound or the composition for use according to any one of the preceding items, wherein the compound is 4-hydroxyphenyllactic acid or a pharmaceutically acceptable salt thereof.
  • composition for use according to any one of the preceding items, wherein the composition comprises is 4-hydroxyphenyllactic acid and indole-3-lactic acid.
  • composition for use according to any one of the preceding items, wherein the composition comprises is 4-hydroxyphenyllactic acid or indole-3-lactic acid.
  • the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has an abundance of Aldh+ bacteria of at most 10% of total bacteria, such as at most 9% of total bacteria, such as at most 8% of total bacteria, such as at most 7% of total bacteria, such as at most 6% of total bacteria, such as at most 5% of total bacteria, such as at most 4% of total bacteria, such as at most 3% of total bacteria, such as at most 2% of total bacteria, such as at most 1% of total bacteria.
  • the dysbiotic gut has an abundance of Aldh+ bacteria of at most 10% of total bacteria, such as at most 9% of total bacteria, such as at most 8% of total bacteria, such as at most 7% of total bacteria, such as at most 6% of total bacteria, such as at most 5% of total bacteria, such as at most 4% of total bacteria, such as at most 3% of total bacteria, such as at most 2% of total bacteria, such as at most 1% of total bacteria.
  • the compound or the composition for use according to any one of the preceding items wherein the infant is between 3 weeks and 6 months of age, such as between 3 weeks and 5 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 3 months of age, such as between 3 weeks and 2 months of age, such as between 3 weeks and 1 month of age, such as between 1 month and 6 months of age, such as between 2 month and 6 months of age, such as between 3 month and 6 months of age, such as between 4 month and 6 months of age.
  • the compound or the composition for use according to any one of the preceding items, wherein the infant has at most 3x10 9 Aldh+ bacteria/g feces.
  • aromatic lactic acid such as is 4-hydroxyphenyllactic acid and/or indole-3- lactic acid and/or phenyllactic acid.
  • the compound or the composition for use according to any one of the preceding items wherein the infant has at most 100 nmol/g feces of 4- hydroxyphenyllactic acid in the gut at between 3 weeks and 6 months of age, such as between 2-5 months of age.
  • 3 weeks and 6 months of age such as between 3 weeks and 5 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 3 months of age, such as between 3 weeks and 2 months of age, such as between 3 weeks and 1 month of age, such as between 1 month and 6 months of age, such as
  • allergen-specific IgE production in human B cells and/or plasma cells is decreased systemically to 0.35 kU/L or less, such as to 0.1 kU/L or less.
  • the IgE-mediated disease is an allergic response to an allergen.
  • the allergen is a pollen, a dust mite, a food allergen, a plant allergen, animal dander, insect stings, a fungus, a spore, a mold, latex, or a drug.
  • IgE-mediated disease is allergic asthma, urticaria, angioedema, food allergy, an allergic response, atopic dermatitis, anaphylaxis, cutaneous mastocystosis, allergic rhinitis, allergic rhinoconjuncvititis, nasal polyposis, Kimura's disease, eosinophilic otitis media, eosinophilic gastroenteritis, latex allergy, bronchopulmonary allergic aspergillosis.
  • composition for use according to any of the preceding items, wherein the composition is administered to the subject orally and/or parenterally.
  • composition for use according to any of the preceding items, wherein the composition is administered to the subject via intramuscular, and/or subcutaneous and/or IV administration.
  • composition is a pharmaceutical composition.
  • Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM and/or plasma IgE levels of 0.5 kll/L or below.
  • a method of reducing the risk of IgE-mediated disease in a subject comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kll/L, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM.
  • a method of reducing the risk of IgE-mediated disease in a subject comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kU/L, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM and/or plasma IgE levels or 0.5 kU/L or below.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in an infant comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the infant until a level of at least 5 pM of one or more aromatic lactic acids in plasma is achieved until 6 months of age of the infant.
  • a method of reducing the risk of IgE-mediated disease in an infant comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the infant, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM.
  • a method of reducing the risk of IgE-mediated disease in an infant comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b.
  • Administering a composition comprising one or more compounds of formula (I) or one or more compounds of formula (II), or pharmaceutically acceptable salts thereof, to the infant until a total level of aromatic lactic acids in plasma of at least 5 pM is achieved until 6 months of age.
  • the compound for use or the method according to items 88 to 96 wherein the aromatic lactic acids are selected from the group consisting of 4- hydroxyphenyllactic acid, indole-3-lactic acid and/or phenyllactic acid.
  • a method for the treatment and/or prevention of an IgE-mediated disease comprising administration of a therapeutically effective amount of a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, formula (I) wherein;
  • R 1 represents hydroxy group or -H
  • R 2 represents an alkyl, cycloalkyl, aryl or heteroaryl group, or -H, ' represents a single or double bond,
  • A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, to a subject in need thereof.
  • composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, formula (I) wherein;
  • R 1 represents hydroxy group or -H
  • R 2 represents an alkyl, cycloalkyl, aryl or heteroaryl group, or -H,
  • A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for the manufacture of a medicament for the treatment and/or prevention of an IgE-mediated disease.
  • Example 1 A high gut bacterial load between 1 week and 6 months of age is linked to decreased incidence of IgE-mediated diseases later in life
  • Fecal samples were collected from children taking part in the ALADDIN (Assessment of Lifestyle and Allergic Disease During INfancy) birth cohort (Stenius et al., 2011 ; Nilsson et al., 2019) at eight different timepoints: 3-6 days, 3 weeks, 2 months, 6 months, 12 months, 18 months, 24 months, and 60 months postpartum, and from mothers at 2 months postpartum. All fecal samples were placed at -20 °C within 20 minutes of sampling and transferred on ice for permanent storage at -80 °C.
  • Fecal samples were thawed on ice and small aliquots (100-350 mg, weight noted) were sampled representatively from the middle of the stool. Aliquots were diluted 1:4 in sterile miliQ water (4 °C; 400 pL per 100 mg sample) and homogenized by brief vortexing at 3,000 rpm followed by 1-hour vortexing at 1,000 rpm, 4 °C. Samples were briefly shaken by hand for complete homogenization and divided into two aliquots, one for metabolomics and one for bacterial enumeration, respectively. Homogenized samples for bacterial enumeration were centrifuged at 50g, 4 °C for 15 min.
  • Bacterial pellets were washed twice in PBS (Lonza) with 1% BSA (Sigma-Aldrich) and centrifuged at 8,000g, 4 °C for 5 min. The bacteria were resuspended in PBS with 1% BSA and an aliquot was diluted 150x in PBS with 1% BSA, 1mM EDTA (Sigma- Aldrich), and 0.01% Tween20 (Sigma-Aldrich) before staining with DAPI (Sigma- Aldrich) at room temperature for 15 min. and adding 123count eBeads (eBiosciences). DAPI-stained samples were run on a BD FACSCanto II flow cytometer to determine number of bacteria per pL undiluted sample.
  • Atopic dermatitis was evaluated at 2 years of age using SCORing of Atopic Dermatitis (SCORAD) scores (Stalder ef al., 1992).
  • Genomic DNA for metagenomic sequencing was isolated with the Nucleospin 96-well kit for DNA from soil (Macherey Nagel), with only slight deviations from the manufacturer’s instructions. Fecal samples were thawed on ice and small aliquots (50- 100 mg, weight noted) were sampled representatively from the middle of the stool and transferred to MN Bead Tubes containing ceramic beads. To lyse the samples, 700 pL Buffer SL1 and 150 pL Enhancer SX were added and the samples vortexed for 5 min. at full speed, room temperature (RT).
  • RT room temperature
  • Foam was removed by centrifuging at 11 ,000g, RT for 10 min., and samples were resuspended in 150 pL Buffer SL3, vortexed for 5 seconds, and stored at -80 °C. Later, samples were thawed on ice, vortexed for 5 seconds, centrifuged at 11 ,000g, RT for 10 min., and 800 pL clear supernatant transferred to the MN Nucleospin Inhibitor Removal Plate.
  • Lysate was filtered by centrifugation at 4,700g, RT for 6 min., and the resulting flowthrough in the MN Squarewell Block was mixed with 250 pL Buffer SB before transferring 750 pL onto the MN Nucleospin Soil Binding plates by centrifugation at 4,700g, RT for 5 min.
  • the silica membrane was washed according to manufacturer’s instructions, though with centrifugations at 4,700g, and the binding plate was air-dried for 15 min. on collection tube strips.
  • DNA was eluted by adding 50 pL 80 °C preheated SE buffer directly onto the silica membrane center, incubating for 10 min., and centrifuging at 4,700g, RT for 3 min. The elution was repeated for a final volume of 100 pL and DNA samples were stored in microcentrifuge tubes at -80 °C.
  • End-repair and A-tailing was performed on 20 ng of purified DNA using a 2:2:1 mixture of T4 DNA polymerase, T4 polynucleotide kinase, and rTaq DNA polymerase, before heat-inactivating the enzymes at 75 °C.
  • T4 DNA ligase adaptors with barcodes (Ad1532B) were ligated to DNA fragments at 23 °C and the barcoded fragments were amplified by PCR.
  • 55 ng purified DNA was denatured at 95 °C and ligated at 37 °C with T4 DNA ligase.
  • the resulting 5,139,318 genes were taxonomically annotated to bacteria, fungi, viruses, archaea, and protozoa by aligning to reference genomes from Refseq (2020-01-27) using nucleotide BLAST (v2.6.0;Camacho et al., 2009). For this purpose, only blast hits with sequence identity >45%, coverage >50%, and alignment length >100 bp were included and up to 50 hits fulfilling these requirements were stored per gene. Sequence identity requirements for taxonomic annotation were 95% for subspecies and species, 85% for genus, 75% for family, 65% for order, 55% for class, 50% for phylum, and 45% for superkingdom.
  • MGS metagenomic species
  • co-abundant gene clusters comprising >100 genes were evaluated by summarizing taxonomy across all genes.
  • >75% of genes in a cluster had to be annotated to the same subspecies and ⁇ 10% annotated to another subspecies.
  • Same requirements were used for species annotation but differed for all higher taxonomical levels, for which the requirements were as follows: >60% annotated to the same taxa and ⁇ 10% to another taxa for genus, >50% and ⁇ 20% for family, >40% and ⁇ 20% for order, >30% and ⁇ 20% for class, >25% and ⁇ 20% for phylum, and >20% and ⁇ 15% for superkingdom.
  • MGS multi- abundant gene cluster annotated at species or subspecies level or comprised by >500 genes was considered an MGS.
  • a total of 919 MGS were identified across all samples, of which 8 were annotated at subspecies level, 387 at species level, 39 at genus level, 82 at family level, 245 at order level, 61 at class level, 32 at phylum level, and 55 at superkingdom level.
  • Abundances were calculated by mapping the paired high-quality reads of non-human origin to a subset of the non-redundant gene catalogue, which was subset by using the 100 genes within the co-abundant gene cluster of each MGS that had the highest Pearson correlation coefficient and did not appear in other MGS. Mapping was performed as described in the above section, and a minimum of 10 genes had to be detected for an MGS to be considered present in a sample. Relative abundance of each MGS was defined as the mean gene abundance across the gene subset of the MGS.
  • Bacteria containing the aldh gene (SEQ ID NO: 1) were defined by sequence similarity, and summed to % abundance of aldh+ and aldh- bacteria respectively.
  • the load of aldh+ and aldh- bacteria in individual samples was calculated by multiplying the % abundance with the load of bacteria/g feces.
  • the maximum load in early life was calculated across week 1 to 6 months samples, and compared for children with (yes) or without (no) later development of indicated diseases.
  • aldh aromatic lactate dehydrogenase
  • Example 3 Reduced levels of 4-OH-PLA in fecal samples from infants between 2 and 6 months in children developing IgE-mediated diseases later in life
  • Fecal water aliquots for metabolomics were centrifuged at 16,000g, 4 °C for 5 minutes. Supernatants were transferred to new tubes and centrifuged at 16,000g, 4 °C for 10 minutes. The resulting supernatants were stored at -80 °C in 100 pL aliquots until further sample preparation. Samples were thawed at 4 °C and centrifuged at 16,000g for 10 minutes. Subsequently, 80 pL supernatant was transferred to a new tube in which 20 pL internal standard mix (4 pg/mL) and 240 pL ice-cold acetonitrile were added.
  • Path a denotes the correlation between max early life (0-6m) fecal aldh+ relative abundance and the 2 months metabolite levels (determined by a linear model)
  • path b is the association between the 2 months metabolite levels and food allergen sensitization until 5 years of age (determined by a generalized linear model)
  • path c is the association between max early life (0-6m) fecal aldh+ relative abundance and food allergen sensitization until 5 years (determined by a generalized linear model).
  • Path c’ in Figure 4C corresponds to the direct effect from the mediation analyses, which is the direct effect of max early life (0- 6m) fecal aldh+ relative abundance on food allergen sensitization until 5 years when the metabolite is considered as a mediator.
  • path c analyses the direct effect of the aldh+ bifidobacteria on food allergen sensitization
  • path c’ does the same but with consideration of whether the effect is driven by the mediator. Since no significant effect was observed (c’ was not significant) of aldh+ bifidobacteria on food allergen sensitization when considering the 2 months 4-OH-PLA levels, and the mediation analysis (Figure 4A) demonstrated a significant causal mediation effect, a complete mediation through 4-OH-PLA was observed, while PLA did not mediate the effect.
  • Example 3a High fecal levels of 4-OH-PLA in infants at 2-6 months of age decreased the risk of later development of IgE-mediated diseases.
  • 4-OH-PLA is the only one of the three gut bacteria-derived aromatic lactic acids with a causal mediating effect on the reduction of IgE-mediated diseases in vivo.
  • Total ALA levels The summed 2 months fecal levels of ALAs ranged from about 1-600 nmol/g feces (Figure 5a), while the maximum summed levels of ALAs in infant fecal samples between 2 and 6 months of age (Figure 5b) ranged from about 40 to 680 nmol/g feces.
  • 4-OH-PLA Increased levels of 4-OH-PLA were linked to reduced risk of development of IgE-mediated atopic diseases in children ( Figure 3 and Figure 4). Children that did not develop food allergen sensitization during the first 5 years of life displayed mean fecal levels of 4-OH-PLA above 61 nmol/g feces at 2 months of age, whereas children that did develop food allergen sensitization during the first 5 years of age displayed mean fecal levels of 4-OH-PLA below 37 nmol/g feces at 2 months of age (Figure 5a).
  • Example 4 Direct and consistent inhibitory effect of both ILA and 4-OH-PLA, but not PLA, on IgE production in human plasma B cells
  • PBMCs Peripheral blood mononuclear cells
  • FBS heat-inactivated fetal bovine serum
  • PBMCs were used directly or gently thawed on the day of experiment and plated at 250,000 PBMCs/well in preheated culture medium (RPMI 1640 (gibco) with 10% heat-inactivated FBS (Lonza), 1% penicillin/streptomycin (Lonza), 2 mM L-glutamine (Lonza), and 50 pM 2-ME (gibco)).
  • IDL Interleukin-1 (Sigma- Aldrich I5508), 4-OH-PLA (Sigma-Aldrich H3253), D-PLA (Sigma-Aldrich 376906), or L- PLA (Sigma-Aldrich 113069)) dissolved in a maximum of 0.1% DMSO in culture medium were added to the PBMCs for final concentrations of 5 pM, 50 pM, 200 pM, or 1000 pM. A vehicle control (0.1% DMSO) was also included.
  • B cells in the PBMC suspension were then activated for IgE production by adding an activation solution containing rhlL-4 (50 ng/mL, Miltenyi Biotec), CD40L (50 ng/mL, R&D systems), and cross-linking HA-Tag antibody (100 ng/mL or 500 ng/mL, R&D systems) in culture medium, and incubated in a humidified 37 °C, 5% CO2 incubator for a total of 10 days. At day 4 and 7, the medium was replenished by removing 50% of the supernatant and adding fresh culture medium with modifier followed by fresh culture medium with activation solution for the original assay concentration and volume. Cell-free supernatants were harvested and immediately stored at -80 °C until analysis, and cells were subjected to viability testing.
  • rhlL-4 50 ng/mL, Miltenyi Biotec
  • CD40L 50 ng/mL, R&D systems
  • cross-linking HA-Tag antibody 100 ng/
  • IgE production was quantified by IgE ELISA (Invitrogen) performed according to the manufacturer’s instructions.
  • IgE production in human plasma B cells is directly inhibited by ILA in a dose-dependent manner (Figure 8).
  • ILA suppresses IgE production on average 58- 92% in the concentration range between 5 to 1000 pM.
  • 4-OH-PLA has the biggest effect on IgE inhibition between 5 and 50 pM, with average suppressive effects of 60% in said range, while at concentrations of 200 and 1000 pM, a reduced effect was observed, with an average IgE inhibition of 26-37%.
  • Example 5 ILA and 4-OH-PLA, but not PLA, have an anti-inflammatory effect in LPS- primed human monocytes.
  • PBMCs peripheral blood mononuclear cells
  • PBMCs peripheral blood mononuclear cells
  • monocytes monocytes were isolated from the PBMCs for a purity >92% using a Pan Monocyte Isolation kit (Miltenyi Biotec). The purity was determined by flow cytometry (MACSQuant 16) on cells stained with 1:25 dilutions of CD14-PE-Cy7 (eBiosciences) and CD16-FITC (Biolegend).
  • Monocytes were incubated in culture medium (RPMI 1640 (Lonza) with 10% heat-inactivated fetal bovine serum (Lonza), 1% penicillin/streptomycin (Lonza), 2 mM L-glutamine (Lonza), and 50 pM 2-ME (Sigma- Aldrich)) in a humidified 37 °C, 5% CO2 incubator.
  • culture medium RPMI 1640 (Lonza) with 10% heat-inactivated fetal bovine serum (Lonza), 1% penicillin/streptomycin (Lonza), 2 mM L-glutamine (Lonza), and 50 pM 2-ME (Sigma- Aldrich)
  • IDLPA Interleukin-Aldrich I5508
  • 4-OH-PLA Sigma-Aldrich H3253
  • D-PLA Sigma-Aldrich 376906
  • L-PLA Sigma-Aldrich 113069
  • DMSO dimethyl methoxysulfoxide
  • Monocytes were then stimulated with lipopolysaccharide (LPS, TLR4 ligand, E. coli 026: B6, Sigma L2654) and IFN-y (RD285-IF-100) at final concentrations of 100 ng/mL of both, and incubated for 18 hours.
  • LPS lipopolysaccharide
  • RD285-IF-100 IFN-y
  • ILA, 4-OH-PLA and PLA The effect of ILA, 4-OH-PLA and PLA on the production of the pro-inflammatory cytokine; I L-12p70, in primary LPS-primed human monocytes is demonstrated in Figure 10.
  • ILA and 4-OH-PLA inhibited I L-12p70 production in primary LPS-primed human monocytes, whereas D- and L-PLA had no significant effect.
  • Ehrlich AM Pacheco AR, Henrick BM, et al (2020) lndole-3-lactic acid associated with Bifidobacterium- dominated microbiota significantly decreases inflammation in intestinal epithelial cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Pulmonology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to the use of compositions comprising 4-hydroxyphenyllactic acid for the prevention and/or treatment of IgE-mediated diseases.

Description

-HYDROXYPHENYLLACTIC ACID FOR USE IN TREATING AND REDUCING THE RISK OF IGE-MEDIATED DISEASES
Technical field
The present invention relates to the use of compositions comprising 4- hydroxyphenyllactic acid for the prevention and/or treatment of IgE-mediated diseases.
Background
The gut microbiota can impact host physiology in various ways via the production of metabolites. This can result in biochemical, immune and metabolic changes in the gut. Indeed, depletion of specific microbes, such as Bifidobacterium, in early life has been associated with increased risk of development of IgE-mediated diseases, such as allergy and asthma, in childhood, and suggested to compromise immune function and susceptibility to infectious disease. Consequently, there is a significant interest in pinpointing microbial elements that can promote a more robust imprinting of the immune system, potentially preventing the onset of allergies, autoimmunity, and other conditions related to immune function (Renz & Skevaki, 2020).
The depletion of Bifidobacterium in early life has been linked to an elevated risk of developing autoimmune conditions, as evidenced by a birth cohort study in Finland (Vatanen et al., 2016) and the occurrence of atopic wheeze in a rural Ecuadorian cohort (Arrieta et al., 2017). Additionally, observational studies have noted a correlation between the absence of Bifidobacterium in infants and early-life enteric inflammation, although the underlying mechanisms remain unclear (Henrick et al., 2019; Rhoads et al., 2018).
Treatment of IgE-mediated diseases, such as allergy and asthma, face several limitations. Many existing therapies, such as antihistamines, corticosteroids, bronchodilators and immunomodulators primarily focus on alleviating symptoms rather than addressing the underlying causes of IgE-mediated diseases. In addition, currently available therapeutics for IgE-mediated diseases are associated with highly variable patient responses, limited efficacy in severe cases and in some cases, such as with systemic corticosteroids, may exhibit immunosuppressive effects that can increase susceptibility to infections and other complications. Therefore, there is an unmet need for efficacious therapeutics for IgE-mediated diseases which target their fundamental aetiology. Summary
As outlined above, the depletion of Bifidobacterium in the gut in early life has been linked to an elevated risk of development of IgE-mediated diseases, such as allergy and asthma, in childhood. Further, there is an unmet need for efficacious therapeutics for IgE-mediated diseases which target their fundamental aetiology. The present invention is directed to compositions for use to prevent and/or treat IgE-mediated diseases.
Aldh+ Bifidobacterium species convert aromatic amino acids (tryptophan, phenylalanine and tyrosine) into their respective aromatic lactic acids (indolelactic acid, phenyllactic acid and 4-hydroxyphenyllactic acid) via the aromatic lactate dehydrogenase (ALDH) enzyme (Laursen et al., 2021). The inventors have identified that a deficiency in specific Bifidobacterium-derived metabolites within the gut at infancy is associated with an increased risk of the development of IgE-mediated disease later in life.
It has for long been recognized that there is a stronger TH2 driven environment in early life in later atopic vs. non-atopic children (Prescott et al, Lancet, 1999), and/or in subjects with type 2-promoting genetics. This associates with a higher propensity for programming of the IgE-driving events that underlie IgE-mediated diseases, such as allergic asthma, food allergy and allergic rhinitis. IgE is produced in plasma B cells following cross-linking of B cell receptors (BCR) by allergens and stimulation via IL-4. The molecular events that protect against IgE production in early life and in adults have been unknown so far. The present invention covers the identification of compositions that inhibit the BCR-mediated stimulation of IgE production.
Surprisingly, the inventors have found that the specific Bifidobacterium-derived metabolites ILA and 4-OH-PLA both decrease the production of IgE in B cells in vitro at physiological concentrations, whereas others do not (D-PLA and L-PLA), but that only high levels of 4-OH-PLA (and not ILA) decreases disease incidence in vivo. Therefore, supplementation of 4-OH-PLA, but not the other aromatic lactic acids, can be used for the prevention and/or treatment of IgE mediated diseases. In one aspect, the present invention is directed to compositions comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof, formula (II), wherein;
A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
In another aspect, the present invention is directed to a compound of formula (II) or a pharmaceutically acceptable salt thereof, formula (II), wherein;
A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in a method of reducing the risk of IgE-mediated disease in a subject, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or a systemic allergen-specific IgE level above 0.35 kll/L, and, b. Administering a composition comprising one or more compounds of formula (II) or a pharmaceutically acceptable salt thereof to the subject to achieve a total level of aromatic lactic acids in plasma of at least 5 pM.
The invention is further described in the claims attached hereto. Description of Drawings
Figure 1. A high gut bacterial load between week 1 and 6 months of age is linked to decreased incidence of IgE-mediated diseases later in life. (A) Bacterial load in fecal samples of children from 3-6 days (1w), 3 weeks, 2 months, 6 months, 12 months, 18 months, 24 months (2 years), and 60 months (5 years) postpartum and in fecal samples from their mothers 2 months postpartum (Mother). Dots represent individual samples, and the line represents median abundance. (B-E) Mean bacterial load in early life fecal samples (1 week to 6 months) of children which are not (No)/are (Yes), (B) sensitized to food allergens < 5 years postpartum (37/18), (C) diagnosed with atopic dermatitis as defined by SCORAD at 2 years of age (51/4), (D) repeatedly prescribed asthma medication < 14 years postpartum (45/11) and (E) repeatedly prescribed allergic rhinitis medication < 14 years postpartum (45/11). Dots represent individual samples. Statistics: Welch two-sample t-test.
Figure 2. Bacteria that possess the gene aromatic lactate dehydrogenase (aldh), in subjects between week 1 and 6 months of age, reduce the risk of development of IgE-mediated diseases later in life. (A) Load of aromatic lactic acid (ALA)- producing bifidobacteria (aldh+) and bifidobacteria which do not possess the gene required for ALA production (aldh-) in fecal samples of children and their mothers. Dots represent individual samples and the lines represent median load. Median % aldh+ describes the percent load of aldh+ bifidobacteria among all bifidobacteria at each timepoint. (B-E) Maximum load of bifidobacterial species in early life (from 1 week to 6 months) in fecal samples from children which are not (No)/are (Yes) (B) sensitized to food allergens < 5 years postpartum (37/18), (C) diagnosed with atopic dermatitis as defined by SCORAD at 2 years of age (51/4), (D) repeatedly prescribed asthma medication < 14 years postpartum (45/11) and (E) repeatedly prescribed allergic rhinitis medication < 14 years postpartum (45/11). Dots represent individual samples.
Statistics: Two-sided wilcoxon rank sum test.
Figure 3. High levels of 4-OH-PLA at 2 months and between 2 and 6 months postpartum are associated with decreased incidence of IgE-mediated diseases later in life. (A-B) Fecal levels of 4-OH-PLA at 2 months in children which are not (No)/are (Yes) (A) sensitized to food allergens < 5 years postpartum (81/59) and (B) diagnosed with atopic dermatitis as defined by SCORAD at 2 years of age (126/13). (C-D) Maximum fecal levels of 4-OH-PLA between 2 and 6 months in children which are not (No)/are (Yes) (C) repeatedly prescribed asthma medication < 14 years postpartum (45/11) and (D) repeatedly prescribed allergic rhinitis medication < 14 years postpartum (45/11). (E-F) Fecal levels of PLA at 2 months in children which are not (No)/are (Yes) (E) sensitized to food allergens < 5 years postpartum (79/59) and (F) diagnosed with atopic dermatitis at 2 years of age (125/12). (G-H) Maximum fecal levels of PLA between 2 and 6 months in children which are not (No)/are (Yes) (G) repeatedly prescribed asthma medication < 14 years postpartum (45/11) and (H) repeatedly prescribed allergic rhinitis medication < 14 years postpartum (45/11). (I-J) Fecal levels of ILA at 2 months in children which are not (No)/are (Yes) (I) sensitized to food allergens < 5 years postpartum (77/59) and (J) diagnosed with atopic dermatitis at 2 years of age (123/12). (K-L) Maximum fecal levels of ILA between 2 and 6 months in children which are not (No)/are (Yes) (K) repeatedly prescribed asthma medication < 14 years postpartum (44/11) and (L) repeatedly prescribed allergic rhinitis medication < 14 years postpartum (44/11). Dots represent individual samples. Statistics: Welch two- sample t-test.
Figure 4. The metabolite 4-OH-PLA completely mediates the association of aldh+ bifidobacteria with decreased incidence of food allergen sensitization, while PLA does not. (A-B) Forest plots displaying causal mediation analyses quantifying the contribution of (A) 2 months fecal 4-OH-PLA levels (showing statistically significant causal mediation effect, P = 0.002) and (B) 2 months fecal PLA levels (with no statistically significant causal mediation effect, P = 0.122) to the association of max early life (0-6m) aldh+ fecal relative abundance with food allergen sensitization until 5 years of age. Dots represent the point estimate and horizontal lines the 95% confidence interval. Significance was tested by bootstrapping with 1000 Monte Carlo draws. The estimate corresponds to the change in log odds of food allergen sensitization until 5 years of age per one-unit increase. Accordingly, a 100 nmol/g feces (100-unit) increase in 2 months faecal 4-OH-PLA levels is estimated to significantly decrease the odds of developing food allergen sensitization during the first 5 years of life by 15% (C) Total effect and mediation model, including tests of significant associations proceeding the mediation analyses. Association between max early life (0-6m) fecal aldh+ relative abundance and food allergen sensitization until 5 years of age (path c) and 2m fecal 4-OH-PLA or PLA levels and food allergen sensitization (path b) were tested with a generalized linear model. The 2m fecal 4-OH-PLA or PLA levels were predicted by max early life (0-6m) fecal aldh+ relative abundance (path a) using a linear model. Path c corresponds to the total effect in the mediation analyses.
Figure 5. Levels of individual aromatic lactic acids (ALAs) and total ALA levels in fecal samples at 2 months of age in children with later development of food allergen sensitization (until 5 years of age) or diagnosis of atopic dermatitis at 2 years and at maximum 2-6 months of age in children with repeated prescriptions of asthma or allergic rhinitis medication until 14 years of age. (A) Levels of the three ALAs and their sum in feces at 2 months of age in children that later are not (No)/are (Yes) i) sensitized to food allergens < 5 years postpartum and ii) diagnosed with atopic dermatitis as determined by SCORAD. (B) Maximum levels of the three ALAs and their sum in feces between 2 and 6 months of age in children that later are not (No)/are (Yes) i) repeatedly prescribed asthma medication < 14 years postpartum and ii) repeatedly prescribed allergic rhinitis medication < 14 years postpartum. Vertical lines on the x-axes represent individual samples and the vertical line on each density curve represents the mean.
Figure 6. ILA levels detected in plasma at 6 months of age correlate with relative abundance of fecal aldh+ bacteria and with fecal ILA levels at 6 months of age.
(A) Correlation between the relative abundance of aldh+ bifidobacteria at 6 months and plasma ILA at 6 months (n = 40). ILA levels in plasma were below 11 pM. 1 pM plasma ILA corresponds to around 30% of fecal aldh+ bacteria at 6 months of age. (B) Correlation between fecal ILA and plasma ILA at 6 months (n = 40). 1 pM plasma ILA corresponds to ca. 40 nmol ILA/g feces. Dots represent individual samples. Statistics: Spearman rank correlation. SCC: Spearman rank correlation coefficient. Lines represent the linear regression line; grey area represents 95% confidence interval.
Figure 7. Both ILA and 4-OH-PLA decrease IgE production in vitro in blood- derived human plasma B cells. Freshly isolated human blood-derived peripheral blood mononuclear cells were stimulated for 10 days with 50 ng/mL CD40L, 500 ng/mL cross-linking HA antibody and 50 ng/mL rhlL-4 to induce IgE production in plasma B cells. Co-incubation with 200 pM ILA or 4-OH-PLA resulted in at least 50% inhibition of IgE production. Data represent mean +/- SEM of assay replicates in two different subjects. Figure 8. Dose-dependent inhibition of IgE production in vitro in blood-derived human plasma B cells by both ILA and 4-OH-PLA. Cryopreserved human blood- derived peripheral blood mononuclear cells were thawed and stimulated for 10 days with 50 ng/mL CD40L, 100 ng/mL cross-linking HA antibody and 50 ng/mL rhlL-4 to induce IgE production in plasma B cells. Co-incubation with doses of 5, 50, 200 and 1000 pM ILA or 4-OH-PLA resulted in varying in vitro IgE inhibition patterns.
Concentrations up to 11 pM are detectable in human blood. 4-OH-PLA showed highest effect at physiological doses of 5-50 pM, while ILA was even more effective at the very higher doses. Cell viability was unaltered at these compound concentrations. Data represent mean +/- SEM of replicates from 6 subjects.
Figure 9. ILA and 4-OH-PLA consistently inhibit in vitro IgE production in primary human plasma B cells, while D- and L-PLA exhibit inconsistent effects across subjects.
Human blood-derived peripheral blood mononuclear cells were stimulated for 10 days with 50 ng/mL CD40L, 100 ng/mL cross-linking HA antibody and 50 ng/mL rhlL-4 to induce IgE production in plasma B cells. Co-incubation with high concentrations of 200 pM ILA or 200 pM 4-OH-PLA resulted in consistent in vitro inhibition of IgE production, while 200 pM D-PLA and L-PLA showed inconsistent effects across subjects, even at this high concentration. Cell viability was unaltered at these compound concentrations. Data represent mean +/- SEM of replicates from 6 subjects.
Figure 10. Both ILA and 4-OH-PLA inhibit in vitro IL-12p70 production in primary human monocytes, while D- and L-PLA show no effect. Human blood derived monocytes were isolated and stimulated for 20 hours with 100 ng/mL E. coli LPS and 100 ng/mL rhIFN-g. Co-incubation with high concentrations of 200 pM ILA, 200 pM D- PLA, 200 pM L-PLA or 200 pM 4-OH-PLA resulted in in vitro inhibition of I L-12p70 production for ILA and 4-OH-PLA, while D-PLA and L-PLA showed no reducing effect. Cell viability was unaltered at these compound concentrations. Data represent mean +/- SEM of replicates from 5 subjects. Statistics: Paired t-test, ns > 0.05, * < 0.05 and ** < 0.01.
Detailed description
Definitions As used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise.
As used herein, the expression "and/or" in connection with two or more recited objects includes individually each of the recited objects and the various combinations of two or more of the recited objects, unless otherwise understood from the context and use. As used herein, unless specifically indicated otherwise, the word "or" is used in the inclusive sense of "and/or" and not the exclusive sense of "either/or."
The use of the term "include," "includes," "including," "have," "has," "having," "contain," "contains," or "containing," including grammatical equivalents thereof, should be understood generally as open-ended and non-limiting, for example, not excluding additional unrecited elements or steps, unless otherwise specifically stated or understood from the context.
Where the use of the term "about" is before a quantitative value, the present disclosure also includes the specific quantitative value itself, unless specifically stated otherwise. As used herein, the term "about" refers to a plus or minus 10 percent variation from the nominal value unless otherwise indicated or inferred.
As used herein, the terms "administering" and "administration" refer to any method of providing an agent to the subject (e.g., an IgE-reducing agent). Such methods are known to those skilled in the art, and include, but are not limited to, oral administration, transdermal administration, administration by inhalation, nasal administration, topical administration, intravaginal administration, ophthalmic administration, intra-aural administration, intracerebral administration, administration to spinal cord, administration to intracerebral fluid, rectal administration, and parenteral administration, including injectable such as intravenous administration, intra-arterial administration, intramuscular administration, and subcutaneous administration. Administration can be continuous or intermittent. In some instances an IgE-reducing agent can be administered therapeutically. In other instances, an IgE-reducing agent can be administered prophylactically, such as administered for prevention of a disease or condition in a subject, or for improvement of one or more immune cell (e.g., basophil) functions in a subject (e.g., in the lung of a subject). As used herein, the terms "effective amount" or "amount effective" or "therapeutically effective amount" refer to an amount that is sufficient to achieve the desired result (e.g., therapeutic benefit) or to have an effect on an undesired condition. For example, a "therapeutically effective amount" of a composition of the present invention may refer to an amount that is sufficient to achieve the desired result or to have an effect on a disease in a subject. Alternatively, or in addition, a "therapeutically effective amount" of a composition of the present invention may refer to an amount of the compositions of the present invention that is sufficient to reduce the level and/or activity of IgE in a subject (e.g., in a biological sample obtained from the subject), or to switch one or more functions of an immune cell (e.g., B cells) in a subject to whom a composition of the present invention is administered. The specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including: the disorder being treated and the severity of the disorder; the age, body weight, general health, sex, diet, ethnic group and/or geographical location of the subject; the time of administration; the route of administration; the rate of excretion of the composition of the present invention employed; the duration of the treatment; drugs used in combination or coincidental with the specific composition of the present invention employed and like factors known in the medical arts. If desired, the effective daily dose can be divided into multiple doses for purposes of administration. Consequently, single dose compositions can contain such amounts or submultiples thereof to make up the daily dose. The dosage can be adjusted by the individual physician in the event of any contraindications. Dosage can vary, and can be administered in one or more dose administrations daily, for one or several days. In some instances, a composition of the present invention can be administered in a prophylactically effective amount.
As used herein with respect to a given parameter, the term "decreased level" or “decreased concentration” or “decreased” refers to a level that is detectably lower (e.g., by about 5-10 percent, 10-20 percent, 20-30 percent, 30-40 percent, 40-50 percent, 50- 60 percent, 60-70 percent, 70-80 percent, 80-90 percent, 85-95 percent, or more; such as, by about 5 percent, 10 percent, 15 percent, 20 percent, 25 percent, 30 percent, 35 percent, 40 percent, 45 percent, 50 percent, 55 percent, 60 percent, 65 percent, 70 percent, 75 percent, 80 percent, 85 percent, 90 percent, 95 percent, 97 percent, 99 percent, or more) compared to a reference level. For example, as used herein, decreased IgE production, in a subject may refer to detectably lower (e.g., by about 5- 10 percent, 10-20 percent, 20-30 percent, 30-40 percent, 40-50 percent, 50-60 percent, 60- 70 percent, 70-80 percent, 80-90 percent, or 85-95 percent, or more; such as, by about 5 percent, 10 percent, 15 percent, 20 percent, 25 percent, 30 percent, 35 percent, 40 percent, 45 percent, 50 percent, 55 percent, 60 percent, 65 percent, 70 percent, 75 percent, 80 percent, 85 percent, 90 percent, 95 percent, 97 percent, 99 percent, or more) level of IgE compared to a reference level of IgE.
As used herein, the term “treatment and/or prevention” is to be understood as the treatment and/or prevention and/or reduction and/or alleviation of the indicated pathology, pathophysiological process and/or disease, and/or the treatment and/or prevention and/or reduction and/or alleviation of the risk factors for the indicated pathology, pathophysiological process and/or disease.
As used herein, the term “HCAR2” is to be understood as Hydroxycarboxylic acid receptor 2, also known as GPR109A, niacin receptor 1 (NIACR1), HCA2, HM74a, HM74b, NIACR1, PUMAG, and Puma-g.
As used herein, the term “HCAR3” is to be understood as Hydroxycarboxylic acid receptor 3, also known as niacin receptor 2 (NIACR2) and GPR109B.
As used herein, the term “AhR” is to be understood as aryl hydrocarbon receptor, also known as AHR, ahr, ahR, AH receptor, or dioxin receptor.
As used herein, the term “bifidobacteria" is to be understood as microorganisms belonging to the genus Bifidobacterium. Examples include but are not limited to Bifidobacterium adolescentis, B. angulatum, B. animalis subsp. animalis, B. animalis subsp. lactis, B. asteroides, B. bifidum, B. bourn, B. breve, B. catenulatum, B. choerinum, B. coryneforme, B. cuniculi, B. denticolens, B. dentium, B. gallium, B. gallinarum, B. globosum; B. indicum, B. infantis, B. inopinatum, B. lactis, B. longum, B. magnum, B. merycicum, B. minimum, B. parvulorum, B. pseudocatenulatum, B. pseudoIongum subsp. globosum, B. pseudoIongum subsp. pseudoIongum, B. pullorum, B. ruminale, B. ruminantium, B. saeculare, B. scardovii, B. subtile, B. suis, B. thermacidophilum, and B. thermophilum.
As used herein, the term “Aldh+ bifidobacteria" is to be understood as any microorganism belonging to the genus Bifidobacterium which contains the gene encoding the enzyme aromatic lactate dehydrogenase (Aldh), as defined in SEQ ID NO: 1.
“Aromatic lactic acids” or “ALAs” are metabolites synthesized by certain bacteria, for example by microorganisms belonging to the genus Bifidobacterium, from dietary amino acids. Examples of ALAs are indole-3-lactic acid (ILA), 4-hydroxyphenyllactic acid (4-OH-PLA), D-phenyllactic acid (D-PLA) and L- phenyllactic acid (L-PLA).
As used herein, the term “gut microbiota” is to be understood as all the bacteria and other microorganisms within the gut.
As used herein, "gut microbiota dysbiosis" refers to an imbalance in the relative abundance or presence of microbes (e.g., beneficial and/or pathogenic microbes) in the gut of a subject that can result in a variety of symptoms including, for example, one or more of bloating, flatus, spasms, inflammation with loss of intestinal permeability, dysplasia of a mucosal surface, insufficient reclamation of nutrients for buffering capacity or increasing risk for development of autoimmune or IgE-mediated diseases. Dysbiosis can include a loss of beneficial microbes and/or an expansion of pathogenic microbes (e.g., pathobionts). Dysbiosis is believed to trigger pro-inflammatory effects and immune dysregulation associated with various disease states.
As used herein, the term "pharmaceutical composition" refers to the combination of an active agent (e.g., a composition of the present invention) with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo. As used herein, the term "pharmaceutical composition" can be a formulation containing a composition of the present invention and/or an agent for reducing IgE production of the present disclosure in a form suitable for administration to a subject. In one embodiment, the pharmaceutical composition is in bulk or in unit dosage form. The unit dosage form is any of a variety of forms, including, for example, a capsule, an intravenous bag, a tablet, a single pump on an aerosol inhaler or a vial. The quantity of active ingredient (e.g., a formulation of a disclosed agent for reducing IgE production alone or together with other immune modulating agents, as well as salt, hydrate, solvate or isomer thereof) in a unit dose of composition is an effective amount and can vary depending upon the particular treatment involved. One skilled in the art will appreciate that it is sometimes necessary to make routine variations to the dosage depending on the age and condition of the subject. The dosage will also depend on the route of administration. A variety of routes are contemplated, including, for example, oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like. Dosage forms for the topical or transdermal administration of an IgE-reducing agent alone or together with other immune modulating agents include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. In one embodiment, an IgE-reducing agent is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that are required.
In some embodiments, an IgE-reducing agent is one or more compounds according to Formula I or Formula II as disclosed and defined herein.
The term "pharmaceutically acceptable carrier" as used herein refers to buffers, carriers, and excipients suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable carriers include any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions (e.g., such as an oil/water or water/oil emulsions), and various types of wetting agents. Pharmaceutically acceptable carriers include buffers, solvents, dispersion media, coatings, isotonic and absorption delaying agents, and the like, that are compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is known in the art.
The terms "pharmaceutically effective amount," "pharmacologically effective amount," "physiologically effective amount," or "effective amount" of the compositions of the present invention are used interchangeably and refer to the amount of a bioactive agent or combination of bioactive agents present in one or more pharmaceutical compositions as described herein that is needed to provide a desired level of active agent or agents in the bloodstream or at the site of action in an subject (e.g., the hepatic system, the renal system, the circulatory system, the lungs, the gastrointestinal system, the colorectal system, the vagina etc.) to be treated to give an anticipated physiological response when such composition is administered. The term “immune modulating agent” as used herein refers to nutrients that have been identified as critical for the growth and function of immune cells, for example vitamins and minerals. In one embodiment, the immune modulating agent may be a vitamin such as Vitamin A, C, D, B, E, p-carotene and others, and/or minerals such as zinc, magnesium, selenium, iron and others.
The term "Allergen" refers to a substance that may elicit an immune reaction (hypersensitive response such as antibody formation) in a subject. Common examples include, but are not limited to: grasses, pollen, milk, egg, seafood (such as shrimp), nuts, bee venom, drugs (such as penicillin), and natural rubber latex proteins.
The term "IgE" or “immunoglobulin E” refers to an immunoglobulin of class E which may be involved in a hypersensitivity response.
The term "sample," as used herein, refers to biological material containing analytes to be assayed. Biological material may be obtained from a subject and may include, for example, tissue, cells, feces, or bodily fluid. In exemplary embodiments, the sample is a bodily fluid. "Sample" can refer to any fluid that may be obtained from a subject, including, but not limited to saliva, blood, plasma, serum, feces, urine, interstitial fluid, lymph, gastric juices, bile, sweat, and spinal and brain fluids, including fluids that are processed or present in their natural form. In some embodiments, a “sample” from a subject is one that may contain circulating IgE. In other embodiments, a “sample” from a subject is one that is suspected to contain suboptimal levels of ALAs.
The term “alkyl” as used herein refers to a linear or branched hydrocarbon moiety.
As used herein the term “cycloalkyl” refers to a monocylcic or polycyclic system. The term “cycloalkyl” also used herein can optionally contain one or more unsaturations or substituents.
The term "heteroaromatic" or “heteroaryl” as used herein, alone or in combination, refers to an aromatic ring containing from 5 to 6 ring atoms where at least one of the ring atoms are heteroatom(s). By "heteroatom" is intended to mean sulfur, oxygen or nitrogen.
The term "aromatic" or “aryl” refers to a cyclic or polycyclic moiety having a conjugated unsaturated (4r|+2)TT electron system (where n is a positive integer), sometimes referred to as a delocalized TT electron system.
The compounds described herein may contain one or more stereocenters. When referring to a chiral carbon or position, the term “stereochemistry configuration” as used herein refers to the absolute configuration of that center, which can be R or S, as determined by the Cahn-lngold-Prelog priority rules.
Compounds and compositions
In one aspect, the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, formula (I) wherein;
R1 represents hydroxy group or -H,
R2 represents an alkyl, cycloalkyl, aryl or heteroaryl group, or -H,
* represents a single or double bond,
A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
In some embodiments, R2 is selected from the group consisting of C1-C10 alkyl optionally substituted with one or more, identical or different, substituents, or a Ca-Cs cycloalkyl optionally substituted with one or more, identical or different, substituents. In one aspect, the present invention relates to a compound of formula (II) or a pharmaceutically acceptable salt thereof, formula (II), wherein; A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in the treatment and/or prevention of an IgE-mediated disease in a subject. In some embodiments, the compound is not phenyllactic acid.
In some embodiments, R1 represents hydroxy,
- represents a single bond,
A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring.
In some embodiments, A represents an unsubstituted indole or; wherein A represents; _
, wherein R is hydroxy.
In some embodiments, A represents; , wherein R is hydroxy.
In some embodiments, A represents an unsubstituted indole.
In some embodiments, R1 represents a -H, represents a double bond,
A represents a substituted aromatic ring.
In some embodiments, A represents; wherein R3 is hydroxy,
R4 is methoxy, hydroxy or -H, R5 is methoxy, hydroxy or -H.
In some embodiments, A represents; wherein R3is hydroxy,
R4is methoxy or-H, R5is methoxy or-H.
In some embodiments, A represents; wherein R3is hydroxy, R4 is methoxy, R5is-H.
In some embodiments, A represents; wherein R3is hydroxy,
R4 and R5 are methoxy or hydroxy.
In some embodiments, A represents; wherein R3is hydroxy, R4 and R5 are methoxy. In some embodiments, the compound binds hydroxycarboxylic acid receptor 3 (HCAR3), aryl hydrocarbon receptor (AHR), and/or hydroxycarboxylic acid receptor 2 (HCAR2). In some embodiments, the compound is an agonist of HCAR3, AHR and/or HCAR2receptors.
In some embodiments, the compound has the structure of formula (1a) formula (1a), or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound has the structure of formula (1b) formula (1b), or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound does not have the structure of formula (1c) formula (1c).
In some embodiments, the compound has the structure of formula (1d) formula (1 d), or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound has the structure of formula (1e) formula (1e), or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound has the structure of formula (1f) formula (1f), or a pharmaceutically acceptable salt thereof.
In one aspect, the present invention relates to a composition comprising or consisting of one or more compounds of formula (I) or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
In one aspect, the present invention relates to a composition comprising or consisting of one or more compounds of formula (II) or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
In some embodiments, the composition comprises, or consists of; a. the compound of formula (1 a) formula (1a), or a pharmaceutically acceptable salt thereof, and/or b. the compound of formula (1 b) formula (1b), or a pharmaceutically acceptable salt thereof.
In some embodiments, the composition comprises, or consists of; a. the compound of formula (1 a) formula (1a), or a pharmaceutically acceptable salt thereof, and b. the compound of (1 b) formula (1b), or a pharmaceutically acceptable salt thereof.
In some embodiments, the composition comprises, or consists of; a. the compound of (1a) formula (1a), or a pharmaceutically acceptable salt thereof, or b. the compound of (1 b) formula (1 b), or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is 4-hydroxyphenyllactic acid or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is indole-3-lactic acid or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is not phenyllactic acid. In some embodiments, the composition comprises 4-hydroxyphenyllactic acid and indole-3-lactic acid. In some embodiments, the composition comprises is 4-hydroxyphenyllactic acid or indole-3- lactic acid.
In some embodiments, the compound is p-coumaric acid or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is Sinapinic acid or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is Ferulic acid or a pharmaceutically acceptable salt thereof.
Patient group
According to the compounds and compositions for use disclosed in the section ‘Compounds and compositions’, in some embodiments, the subject is a human. In some embodiments, the subject is a non-infant. In some embodiments, the subject is an infant. In some embodiments, the subject is a pre-term infant or a term infant. In some embodiments, the infant is an infant born by C-section. In some embodiments, the infant is born from a first-time mother. In some embodiments, the infant has one or both parents affected by an IgE-mediated disease.
In some embodiments, the infant has a dysbiotic gut microbiota. In some embodiments, the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a low abundance of Aldh+ bacteria. In some embodiments, the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a low abundance of Aldh+ bacteria, wherein said Aldh+ bacteria comprises an Aldh gene encoding the enzyme aromatic lactate dehydrogenase, preferably an Aldh of SEQ ID NO: 1. In some embodiments, the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has an abundance of Aldh+ bacteria of at most 10% of total bacteria, such as at most 9% of total bacteria, such as at most 8% of total bacteria, such as at most 7% of total bacteria, such as at most 6% of total bacteria, such as at most 5% of total bacteria, such as at most 4% of total bacteria, such as at most 3% of total bacteria, such as at most 2% of total bacteria, such as at most 1% of total bacteria.
In some embodiments, the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has an abundance of Aldh+ bacteria of at most 5% of total bacteria. In some embodiments, the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a low load of Aldh+ bacteria. In some embodiments, the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a load of Aldh+ bacteria of at most 3x109 Aldh+ bacteria/g feces. In some embodiments, the infant is between 3 weeks and 6 months of age, such as between 3 weeks and 5 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 3 months of age, such as between 3 weeks and 2 months of age, such as between 3 weeks and 1 month of age, such as between 1 month and 6 months of age, such as between 2 month and 6 months of age, such as between 3 month and 6 months of age, such as between 4 month and 6 months of age.
In some embodiments, the infant has at most 3x109 Aldh+ bacteria/g feces. In some embodiments, the infant has at most 100 nmol/g feces of at least one aromatic lactic acid. In some embodiments, the infant has at most 100 nmol/g feces of at least one aromatic lactic acid, corresponding to a plasma concentration of at most 3 pM.
The skilled person will be knowledgeable regarding methods for determining concentrations of aromatic lactic acids and/or tryptophan metabolites in faecal and/or plasma samples, non-limiting examples include Liquid chromatography-mass spectrometry utilising appropriate internal standards. Accordingly, in some embodiments, the infant has at most 100 nmol/g feces of at least one aromatic lactic acid, wherein the concentration of the at least one aromatic lactic acid is determined by LC-MS.
In some embodiments, the infant has at most 100 nmol/g feces of at least one aromatic lactic acid, such as is 4-hydroxyphenyllactic acid and/or indole-3-lactic acid and/or phenyllactic acid. In some embodiments, the infant has at most 100 nmol/g feces of the compound of formula (1a), formula (1a), in the gut. In some embodiments, the infant has at most 100 nmol/g feces of 4- hydroxyphenyllactic acid in the gut. In some embodiments, the infant has at most 100 nmol/g feces of 4-hydroxyphenyllactic acid in the gut at between 3 weeks and 6 months of age, such as between 2-5 months of age.
In some embodiments, the infant has less than 100 nmol/g feces of the compound of formula 1b, formula (1b), in the gut.
In some embodiments, the infant has less than 100 nmol/g feces of indole-3-lactic acid in the gut.
In some embodiments, the infant has less than 100 nmol/g feces of the compound of formula (1c), formula (1c), in the gut.
In some embodiments, the infant has less than 100 nmol /g feces of phenyllactic acid in the gut.
In some embodiments, the infant has less than 100 nmol/g feces of the compound or indole-3-lactic acid in the gut between 3 weeks and 6 months of age, such as between 3 weeks and 5 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 3 months of age, such as between 3 weeks and 2 months of age, such as between 3 weeks and 1 month of age, such as between 1 month and 6 months of age, such as between 2 month and 6 months of age, such as between 3 month and 6 months of age, such as between 4 month and 6 months of age.
In some embodiments, the compound is administered in an amount which is effective to decrease IgE production. effect
In some embodiments, the compound decreases IgE production in human B cells and/or plasma cells. In some embodiments, the compound decreases IgE production in human B cells and/or plasma cells to at most 25 ng/mL.
In some embodiments, the compound decreases IgE production in human B cells and/or plasma B cells, wherein IgE production is determined by human B cell assays either alone or together with other cells.
In some embodiments, the compound decreases IgE production in human B cells.
In some embodiments, the compound decreases IgE production in human B cells to at most 25 ng/mL.
In some embodiments, the compound decreases IgE production in plasma B cells.
In some embodiments, the compound decreases IgE production in plasma B cells to at most 25 ng/mL.
In some embodiments, the compound decreases systemic IgE production to below 0.50 kU/L plasma IgE. In some embodiments, the compound decreases systemic IgE production to below 0.50 kU/L plasma IgE, such as to 0.35 or below kU/L, such as to 0.1 kU/L or below. In some embodiments, the compound decreases systemic IgE production to 0.50 kU/L plasma allergen-specific IgE or less.
In some embodiments, the compound decreases systemic IgE production to 0.50 kU/L plasma allergen-specific IgE or below, such as to 0.35 kU/L or below, such as to about 0.1 kU/L. In some embodiments, IgE production in human B cells and/or plasma B cells is decreased systemically to about 0.1 kU/L, such as to 0.35 kU/L or below.
Indications
In some embodiments, the IgE-mediated disease is an allergic response to an allergen.
In some embodiments, the allergen is a pollen, a dust mite, a food allergen, a plant allergen, animal dander, insect stings, a fungus, a spore, a mold, latex, or a drug.
In some embodiments, the IgE-mediated disease is allergic asthma, urticaria, angioedema, food allergy, an allergic response, atopic dermatitis, anaphylaxis, cutaneous mastocystosis, allergic rhinitis, allergic rhinoconjunctivitis, nasal polyposis, Kimura's disease, eosinophilic otitis media, eosinophilic gastroenteritis, latex allergy, bronchopulmonary allergic aspergillosis.
In some embodiments, the IgE-mediated disease is acute or chronic.
Administration
In some embodiments, the compound or composition is administered to the subject prophylactically. In some embodiments, the compound or composition is administered to the subject orally and/or parenterally. In some embodiments, the compound or composition is administered to the subject orally. In some embodiments, the compound or composition is administered to the subject parenterally. In some embodiments, the compound or composition is administered to the subject via intramuscular, and/or subcutaneous and/or IV administration. In some embodiments, the compound or composition is administered with at least one or more therapeutic agent(s). In some embodiments, the at least one or more therapeutic agent(s) is the standard of care treatment for IgE-mediated disease. In some embodiments, the at least one or more therapeutic agent(s) is administered simultaneously, sequentially or separately.
In some embodiments, the composition is a pharmaceutical composition. In some embodiments, a therapeutically effective amount of the composition is administered to the subject. In some embodiments, a therapeutically effective amount of the compound or composition is administered to the infant.
Method to reduce the risk of IgE-mediated disease
In one aspect, the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in a subject, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kll/L, and, b. Administering a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM.
In one aspect, the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in a subject, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kll/L, and, b. Administering a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM and/or plasma IgE levels of 0.5 kll/L or below.
In one aspect, the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in an infant, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the infant, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM.
In one aspect, the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in an infant, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the infant until a level of at least 5 pM of one or more aromatic lactic acids in plasma is achieved until 6 months of age of the infant.
In one aspect, the present invention relates to a method of reducing the risk of IgE- mediated disease in a subject, comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kll/L, and, b. Administering a compound of formula (I) to the subject thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM and/or individual allergen-specific plasma IgE levels below 0.5 kll/L.
In one aspect, the present invention relates to a method of reducing the risk of IgE- mediated disease in a subject, comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut, and, b. Administering a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the subject to achieve a total level of aromatic lactic acids in plasma of at least 5 pM and/or until plasma IgE levels are at most 0.5 kll/L.
In one aspect, the present invention relates to a method of reducing the risk of IgE- mediated disease in an infant, comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the infant thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM. In one aspect, the present invention relates to method of reducing the risk of IgE- mediated disease in an infant, comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the infant until a total level of aromatic lactic acids in plasma of at least 5 pM is achieved until 6 months of age.
In some embodiments, a reduced total level of aromatic lactic acids in the infant gut corresponds to, and is measured by, a level of one or more of the ALAs described herein of 100 nmol/g feces or less, as defined herein.
Currently used assays for measuring allergen-specific plasma IgE level use an allergen-specific plasma IgE level above 0.35 kll/L as cutoff, wherein the assay is positive if the allergen-specific plasma IgE level is above 0.35 kll/L. However, a subject presenting symptoms of IgE-mediated disease such as allergic asthma, urticaria, angioedema, food allergy, an allergic response, atopic dermatitis, anaphylaxis, cutaneous mastocystosis, allergic rhinitis, allergic rhinoconjunctivitis, nasal polyposis, Kimura's disease, eosinophilic otitis media, eosinophilic gastroenteritis, latex allergy, bronchopulmonary allergic aspergillosis, typically have allergen-specific IgE levels of 1 kll/L and above.
Thus, in some embodiments, step a. comprises identifying an allergen-specific plasma IgE level of 0.5 kU/L or higher, such as of 1 kU/L or higher, such as of 10 kU/L or higher.
Reducing an allergen-specific plasma IgE level to 0.5 kU/L or less corresponds to reaching an allergen-specific plasma IgE level at which a subject does not feel, experience or show any allergic symptoms.
In some embodiments, the administered compound is a compound of formula (la) or formula (lb), or a combination thereof. In some embodiments, the aromatic lactic acids are selected from the group consisting of 4-hydroxyphenyllactic acid, indole-3-lactic acid and/or phenyllactic acid.
In one aspect, the present invention relates to a method for the treatment and/or prevention of an IgE-mediated disease comprising administration of a therapeutically effective amount of a composition comprising or consisting of a compound of formula (I) or a pharmaceutically acceptable salt thereof, formula (I) wherein;
R1 represents hydroxy group or -H,
R2 represents an alkyl, cycloalkyl, aryl or heteroaryl group, or -H,
' represents a single or double bond,
A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, to a subject in need thereof.
In one aspect, the present invention relates to use of a composition comprising or consisting of a compound of formula (I) or a pharmaceutically acceptable salt thereof, formula (I) wherein;
R1 represents hydroxy group or -H,
R2 represents an alkyl, cycloalkyl, aryl or heteroaryl group, or -H, represents a single or double bond, A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for the manufacture of a medicament for the treatment and/or prevention of an IgE- mediated disease.
In one embodiment, a therapeutically effective amount of the compound is administered to the subject. In one embodiment, a therapeutically effective amount of the compound is administered to the infant.
Items
1 . A compound of formula (I) or a pharmaceutically acceptable salt thereof, formula (I) wherein;
R1 represents hydroxy group or -H,
R2 represents an alkyl, cycloalkyl, aryl or heteroaryl group, or -H, ' represents a single or double bond,
A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
2. The compound for use according to item 1 , wherein R2 is selected from the group consisting of C1-C10 alkyl optionally substituted with one or more, identical or different, substituents, or a Ca-Cs cycloalkyl optionally substituted with one or more, identical or different, substituents.
3. A compound of formula (II) or a pharmaceutically acceptable salt thereof, formula (II), wherein;
A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
4. The compound for use according to item 3, wherein R2 is selected from the group consisting of C1-C10 alkyl optionally substituted with one or more, identical or different, substituents, or a Ca-Cs cycloalkyl optionally substituted with one or more, identical or different, substituents.
5. The compound for use according to item 1 , wherein the compound is not phenyllactic acid.
6. The compound for use according to item 3, wherein the compound is not phenyllactic acid.
7. The compound for use according to item 1 , wherein;
R1 represents hydroxy,
- represents a single bond,
A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring.
8. The compound for use according to any of the preceding items, wherein A represents an unsubstituted indole or; wherein A represents; The compound for use according to any of the preceding items, wherein A represents; , wherein R2 is hydroxy. The compound for use according to any of the preceding items, wherein A represents an unsubstituted indole. The compound for use according to any of the preceding items, wherein;
R1 represents a -H, represents a double bond,
A represents a substituted aromatic ring. The compound for use according to any of the preceding items, wherein, A represents; wherein R3 is hydroxy,
R4 is selected from the group consisting of methoxy, hydroxy and -H, R5 is selected from the group consisting of methoxy, hydroxy and -H. The compound for use according to any of the preceding items, wherein, A represents; wherein R3 is hydroxy, R4 is methoxy, R5 is -H. 14. The compound for use according to any of the preceding items, wherein, A represents; wherein R3 is hydroxy, R4 and R5 are methoxy.
15. The compound for use according to any of the preceding items, wherein the compound binds hydroxycarboxylic acid receptor 3 (HCAR3), aryl hydrocarbon receptor (AHR) and/or hydroxycarboxylic acid receptor 2 (HCAR2).
16. The compound for use according to any of the preceding items, wherein the compound is an agonist of HCAR3, AHR and/or HCAR2 receptors.
17. The compound for use according to any of the preceding items, wherein the compound has the structure of formula (1a) formula (1a), or a pharmaceutically acceptable salt thereof.
18. The compound for use according to any of the preceding items, wherein the compound has the structure of formula (1b) formula (1b), or a pharmaceutically acceptable salt thereof. 19. The compound for use according to any of the preceding items, wherein the compound does not have the structure of formula (1c) formula (1c).
20. The compound for use according to any of the preceding items, wherein the compound has the structure of formula (1d) formula (1 d), or a pharmaceutically acceptable salt thereof.
21. The compound for use according to any of the preceding items, wherein the compound has the structure of formula (1e) or a pharmaceutically acceptable salt thereof.
22. The compound for use according to any of the preceding items, wherein the compound has the structure of formula (1f) or a pharmaceutically acceptable salt thereof.
23. A composition for use in the treatment and/or prevention of an IgE-mediated disease in a subject, the composition comprising one or more compounds of formula (I). 24. The composition for use according to item 18, wherein R2 is selected from the group consisting of C1-C10 alkyl optionally substituted with one or more, identical or different, substituents, or a Ca-Cs cycloalkyl optionally substituted with one or more, identical or different, substituents.
25. A composition for use in the treatment and/or prevention of an IgE-mediated disease in a subject, the composition comprising one or more compounds of formula (II).
26. The composition for use according to any one of the preceding items, wherein the composition comprises or consists of; a. the compound of formula (1 a) formula (1a), or a pharmaceutically acceptable salt thereof, and/or b. the compound of formula (1 b) formula (1 b), or a pharmaceutically acceptable salt thereof.
27. The composition for use according to any one of the preceding items, wherein the composition comprises or consists of; a. the compound of formula (1 a) formula (1a), or a pharmaceutically acceptable salt thereof, and b. the compound of (1 b) formula (1b), or a pharmaceutically acceptable salt thereof. The composition for use according to any one of the preceding items, wherein the composition comprises or consists of; a. the compound of (1a) formula (1a), or a pharmaceutically acceptable salt thereof, or b. the compound of (1 b) formula (1b), or a pharmaceutically acceptable salt thereof. The compound or the composition for use according to any one of the preceding items, wherein the compound is 4-hydroxyphenyllactic acid or a pharmaceutically acceptable salt thereof. The compound or the composition for use according to any one of the preceding items, wherein the compound is indole-3-lactic acid or a pharmaceutically acceptable salt thereof. 31 . The compound or the composition for use according to any one of the preceding items, wherein the compound is not phenyllactic acid.
32. The compound or the composition for use according to any one of the preceding items, wherein the composition comprises is 4-hydroxyphenyllactic acid and indole-3-lactic acid.
33. The compound or the composition for use according to any one of the preceding items, wherein the composition comprises is 4-hydroxyphenyllactic acid or indole-3-lactic acid.
34. The compound or the composition for use according to any one of the preceding items, wherein the subject is a human.
35. The compound or the composition for use according to any one of the preceding items, wherein the subject is a non-infant.
36. The compound or the composition for use according to any one of the preceding items, wherein the subject is an infant.
37. The compound or the composition for use according to any one of the preceding items, wherein the subject is a pre-term infant or a term infant.
38. The compound or the composition for use according to any one of the preceding items, wherein the infant is an infant born by C-section.
39. The compound or the composition for use according to any one of the preceding items, wherein the infant is born from a first-time mother.
40. The compound or the composition for use according to any one of the preceding items, wherein the infant has one or both parents affected by an IgE- mediated disease.
41 . The compound or the composition for use according to any one of the preceding items, wherein the infant has a dysbiotic gut microbiota. 42. The compound or the composition for use according to any one of the preceding items, wherein the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a low abundance of Aldh+ bacteria.
43. The compound or the composition for use according to any one of the preceding items, wherein the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a low abundance of Aldh+ bacteria, wherein said Aldh+ bacteria comprises an Aldh gene encoding the enzyme aromatic lactate dehydrogenase, preferably an Aldh of SEQ ID NO: 1.
44. The compound or the composition for use according to any one of the preceding items, wherein the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has an abundance of Aldh+ bacteria of at most 10% of total bacteria, such as at most 9% of total bacteria, such as at most 8% of total bacteria, such as at most 7% of total bacteria, such as at most 6% of total bacteria, such as at most 5% of total bacteria, such as at most 4% of total bacteria, such as at most 3% of total bacteria, such as at most 2% of total bacteria, such as at most 1% of total bacteria.
45. The compound or the composition for use according to any one of the preceding items, wherein the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has an abundance of Aldh+ bacteria of at most 5% of total bacteria.
46. The compound or the composition for use according to any one of the preceding items, wherein the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a low load of Aldh+ bacteria.
47. The compound or the composition for use according to any one of the preceding items, wherein the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a load of Aldh+ bacteria of at most 3x109 Aldh+ bacteria/g feces. The compound or the composition for use according to any one of the preceding items, wherein the infant is between 3 weeks and 6 months of age, such as between 3 weeks and 5 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 3 months of age, such as between 3 weeks and 2 months of age, such as between 3 weeks and 1 month of age, such as between 1 month and 6 months of age, such as between 2 month and 6 months of age, such as between 3 month and 6 months of age, such as between 4 month and 6 months of age. The compound or the composition for use according to any one of the preceding items, wherein the infant has at most 3x109 Aldh+ bacteria/g feces. The compound or the composition for use according to any one of the preceding items, wherein the infant has at most 100 nmol/g feces of at least one aromatic lactic acid. The compound or the composition for use according to any one of the preceding items, wherein the infant has at most 100 nmol/g feces of at least one aromatic lactic acid, corresponding to a plasma concentration of at most 3 pM. The compound or the composition for use according to any one of the preceding items, wherein the infant has at most 100 nmol/g feces of at least one aromatic lactic acid, wherein the concentration of the at least one aromatic lactic acid is determined by LC-MS. The compound or the composition for use according to any one of the preceding items, wherein the infant has at most 100 nmol/g feces of at least one aromatic lactic acid, such as is 4-hydroxyphenyllactic acid and/or indole-3- lactic acid and/or phenyllactic acid. The compound or the composition for use according to any one of the preceding items, wherein the infant has at most 100 nmol/g feces of the compound of formula (1a), formula (1a), in the gut. The compound or the composition for use according to any one of the preceding items, wherein the infant has at most 100 nmol/g feces of 4- hydroxyphenyllactic acid in the gut. The compound or the composition for use according to any one of the preceding items, wherein the infant has at most 100 nmol/g feces of 4- hydroxyphenyllactic acid in the gut at between 3 weeks and 6 months of age, such as between 2-5 months of age. The compound or the composition for use according to any one of the preceding items, wherein the infant has less than 100 nmol/g feces of the compound of formula 1b, formula (1b), in the gut. The compound or the composition for use according to any one of the preceding items, wherein the infant has less than 100 nmol/g feces of indole-3- lactic acid in the gut. The compound or the composition for use according to any one of the preceding items, wherein the infant has less than 100nmol/g feces of the compound of formula (1c), formula (1c), in the gut. 60. The compound or the composition for use according to any one of the preceding items, wherein the infant has less than 100nmol /g feces of phenyllactic acid in the gut.
61 . The compound or the composition for use according to any one of the preceding items, wherein the infant has less than 100nmol/g feces of the compound or indole-3-lactic acid in the gut between 3 weeks and 6 months of age, such as between 3 weeks and 5 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 3 months of age, such as between 3 weeks and 2 months of age, such as between 3 weeks and 1 month of age, such as between 1 month and 6 months of age, such as between 2 month and 6 months of age, such as between 3 month and 6 months of age, such as between 4 month and 6 months of age.
62. The compound or the composition for use according to any of the preceding items, wherein the compound is administered in an amount which is effective to decrease IgE production to allergen-specific plasma IgE levels below 0.5 kll/L.
63. The compound or the composition for use according to any of the preceding items, wherein the compound decreases in vitro IgE production in human B cells and/or plasma cells.
64. The compound or the composition for use according to any of the preceding items, wherein the compound decreases in vitro IgE production in human B cells and/or plasma cells to at most 25 ng/mL.
65. The compound or the composition for use according to any of the preceding items, wherein the compound decreases in vitro IgE production in human B cells and/or plasma cells, wherein IgE production is determined by human B cell assays.
66. The compound or the composition for use according to any of the preceding items, wherein the compound decreases IgE production in human B cells. 67. The compound or the composition for use according to any of the preceding items, wherein the compound decreases in vitro IgE production in human B cells to at most 25 ng/mL.
68. The compound or the composition for use according to any of the preceding items, wherein the compound decreases in vitro IgE production in plasma cells.
69. The compound or the composition for use according to any of the preceding items, wherein the compound decreases in vitro IgE production in plasma cells to at most 25 ng/mL.
70. The compound or the composition for use according to any of the preceding items, wherein the compound decreases systemic allergen-specific IgE production to 0.50 kll/L plasma IgE or less.
71. The compound or the composition for use according to any of the preceding items, wherein the compound decreases systemic allergen-specific IgE production to 0.50 kll/L plasma IgE or less, such as to 0.35 kU/L or less, such as 0.1kU/L or less.
72. The compound or the composition for use according to any of the preceding items, wherein the compound the compound decreases systemic IgE production of at most 0.50 kU/L plasma allergen-specific IgE.
73. The compound or the composition for use according to any of the preceding items, wherein the compound decreases systemic IgE production to 0.50 kU/L or less plasma allergen-specific IgE, such as to 0.35 kU/L or less, such as to 0.1kU/L or less.
74. The compound or the composition for use according to any of the preceding items, wherein allergen-specific IgE production in human B cells and/or plasma cells is decreased systemically to 0.35 kU/L or less, such as to 0.1 kU/L or less.
75. The compound or the composition for use according to any of the preceding items, wherein the IgE-mediated disease is an allergic response to an allergen. 76. The compound or the composition for use according to item 73, wherein the allergen is a pollen, a dust mite, a food allergen, a plant allergen, animal dander, insect stings, a fungus, a spore, a mold, latex, or a drug.
77. The compound or the composition for use according to any of the preceding items, wherein the IgE-mediated disease is allergic asthma, urticaria, angioedema, food allergy, an allergic response, atopic dermatitis, anaphylaxis, cutaneous mastocystosis, allergic rhinitis, allergic rhinoconjuncvititis, nasal polyposis, Kimura's disease, eosinophilic otitis media, eosinophilic gastroenteritis, latex allergy, bronchopulmonary allergic aspergillosis.
78. The compound or the composition for use according to any of the preceding items, wherein the IgE-mediated disease is acute or chronic.
79. The compound or the composition for use according to any of the preceding items, wherein the composition is administered to the subject prophylactically.
80. The compound or the composition for use according to any of the preceding items, wherein the composition is administered to the subject orally and/or parenterally.
81. The compound or the composition for use according to any of the preceding items, wherein the composition is administered to the subject orally.
82. The compound or the composition for use according to any of the preceding items, wherein the composition is administered to the subject parenterally.
83. The compound or the composition for use according to any of the preceding items, wherein the composition is administered to the subject via intramuscular, and/or subcutaneous and/or IV administration.
84. The compound or the composition for use according to any of the preceding items, wherein the composition is administered with at least one or more therapeutic agent(s). 85. The compound or the composition for use according to item 82, wherein the at least one or more therapeutic agent(s) is the standard of care treatment for IgE- mediated disease.
86. The compound or the composition for use according to items 82 or 83, wherein the at least one or more therapeutic agent(s) is administered simultaneously, sequentially or separately.
87. The compound or the composition for use according to any of the preceding items, wherein the composition is a pharmaceutical composition.
88. The compound or the composition for use according to any of the preceding items, wherein a therapeutically effective amount of the composition is administered to the subject.
89. The compound or the composition for use according to any of the preceding items, wherein a therapeutically effective amount of the composition is administered to the infant.
90. A compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in a subject, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kll/L, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM.
91. A compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in a subject, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kll/L, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM and/or plasma IgE levels of 0.5 kll/L or below.
92. A method of reducing the risk of IgE-mediated disease in a subject, comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kll/L, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM.
93. A method of reducing the risk of IgE-mediated disease in a subject, comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kU/L, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM and/or plasma IgE levels or 0.5 kU/L or below.
94. A compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in an infant, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the infant, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM. A compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in an infant, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the infant until a level of at least 5 pM of one or more aromatic lactic acids in plasma is achieved until 6 months of age of the infant. A method of reducing the risk of IgE-mediated disease in an infant, comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (II) or a pharmaceutically acceptable salt thereof to the infant, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM. A method of reducing the risk of IgE-mediated disease in an infant, comprising: a. Identifying a reduced total level of aromatic lactic acids in the infant gut, wherein the infant is between 3 weeks and 6 months of age, and, b. Administering a composition comprising one or more compounds of formula (I) or one or more compounds of formula (II), or pharmaceutically acceptable salts thereof, to the infant until a total level of aromatic lactic acids in plasma of at least 5 pM is achieved until 6 months of age. The compound for use or the method according to any one of items 88 to 95, wherein the administered compound is a compound of formula (la) or formula (lb), or a combination thereof. 99. The compound for use or the method according to items 88 to 96, wherein the aromatic lactic acids are selected from the group consisting of 4- hydroxyphenyllactic acid, indole-3-lactic acid and/or phenyllactic acid.
100. A method for the treatment and/or prevention of an IgE-mediated disease comprising administration of a therapeutically effective amount of a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, formula (I) wherein;
R1 represents hydroxy group or -H,
R2 represents an alkyl, cycloalkyl, aryl or heteroaryl group, or -H, ' represents a single or double bond,
A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, to a subject in need thereof.
101. Use of a composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, formula (I) wherein;
R1 represents hydroxy group or -H,
R2 represents an alkyl, cycloalkyl, aryl or heteroaryl group, or -H,
' represents a single or double bond, A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for the manufacture of a medicament for the treatment and/or prevention of an IgE-mediated disease.
102. The method according to any of the preceding items, wherein a therapeutically effective amount of the compound is administered to the subject.
103. The method according to any of the preceding items, wherein a therapeutically effective amount of the compound is administered to the infant.
Examples
Example 1: A high gut bacterial load between 1 week and 6 months of age is linked to decreased incidence of IgE-mediated diseases later in life
Materials and Methods
Human study population and fecal sampling
Fecal samples were collected from children taking part in the ALADDIN (Assessment of Lifestyle and Allergic Disease During INfancy) birth cohort (Stenius et al., 2011 ; Nilsson et al., 2019) at eight different timepoints: 3-6 days, 3 weeks, 2 months, 6 months, 12 months, 18 months, 24 months, and 60 months postpartum, and from mothers at 2 months postpartum. All fecal samples were placed at -20 °C within 20 minutes of sampling and transferred on ice for permanent storage at -80 °C.
Isolation of fecal water and bacteria
Fecal samples were thawed on ice and small aliquots (100-350 mg, weight noted) were sampled representatively from the middle of the stool. Aliquots were diluted 1:4 in sterile miliQ water (4 °C; 400 pL per 100 mg sample) and homogenized by brief vortexing at 3,000 rpm followed by 1-hour vortexing at 1,000 rpm, 4 °C. Samples were briefly shaken by hand for complete homogenization and divided into two aliquots, one for metabolomics and one for bacterial enumeration, respectively. Homogenized samples for bacterial enumeration were centrifuged at 50g, 4 °C for 15 min. to pellet large particles and fecal water supernatant was transferred to new microcentrifuge tubes. Supernatants were diluted 2.5x in sterile miliQ water (4 °C) for a final 1 :10 dilution (corresponding to 1 mL per 100 mg sample) and centrifuged at 8,000g, 4 °C for 5 min. to pellet bacteria.
Bacterial count
Bacterial pellets were washed twice in PBS (Lonza) with 1% BSA (Sigma-Aldrich) and centrifuged at 8,000g, 4 °C for 5 min. The bacteria were resuspended in PBS with 1% BSA and an aliquot was diluted 150x in PBS with 1% BSA, 1mM EDTA (Sigma- Aldrich), and 0.01% Tween20 (Sigma-Aldrich) before staining with DAPI (Sigma- Aldrich) at room temperature for 15 min. and adding 123count eBeads (eBiosciences). DAPI-stained samples were run on a BD FACSCanto II flow cytometer to determine number of bacteria per pL undiluted sample.
Blood sampling and determining food allergen sensitization
Blood samples were collected in heparin tubes and plasma was stored at -20 °C until tested for IgE concentration using ImmunoCAP (Thermo Fisher Scientific). At 5 years of age, a food mix (fx5, Thermo Fisher Scientific) was used to initially determine food allergen sensitization, and if fx5 was positive, the individual allergens (cow’s milk, hen’s egg, peanut, codfish, wheat flour, and soy-bean) were analyzed separately. Food allergen-specific IgE levels > 0.35 kllA/L were categorized as food allergen sensitization, corresponding to the recommended cutoff (Ballardini et al., 2006).
Other clinical outcomes
Children were considered to receive asthma medication if they were prescribed the medication at least twice within the given time period. Likewise, children were considered to receive medication for allergic rhinitis if they had it prescribed at least twice within the given time period. Atopic dermatitis was evaluated at 2 years of age using SCORing of Atopic Dermatitis (SCORAD) scores (Stalder ef al., 1992).
Results
Higher bacterial load in early life (1 week to 6 months) fecal samples was found in children without food allergen sensitization < 5 years postpartum (Figure 1b), diagnosis of atopic dermatitis at 2 years postpartum (Figure 1c), repeated prescriptions of asthma medication < 14 years postpartum (Figure 1d) and repeated prescriptions of allergic rhinitis medication < 14 years postpartum (Figure 1e). Example 2: Bacteria that possess the gene aromatic lactate dehydrogenase (aldh), in subjects between week 1 and 6 months of age, reduce the risk of development of IgE- mediated diseases later in life.
Materials and Methods
Extraction of bacterial DNA from fecal samples
Genomic DNA for metagenomic sequencing was isolated with the Nucleospin 96-well kit for DNA from soil (Macherey Nagel), with only slight deviations from the manufacturer’s instructions. Fecal samples were thawed on ice and small aliquots (50- 100 mg, weight noted) were sampled representatively from the middle of the stool and transferred to MN Bead Tubes containing ceramic beads. To lyse the samples, 700 pL Buffer SL1 and 150 pL Enhancer SX were added and the samples vortexed for 5 min. at full speed, room temperature (RT). Foam was removed by centrifuging at 11 ,000g, RT for 10 min., and samples were resuspended in 150 pL Buffer SL3, vortexed for 5 seconds, and stored at -80 °C. Later, samples were thawed on ice, vortexed for 5 seconds, centrifuged at 11 ,000g, RT for 10 min., and 800 pL clear supernatant transferred to the MN Nucleospin Inhibitor Removal Plate. Lysate was filtered by centrifugation at 4,700g, RT for 6 min., and the resulting flowthrough in the MN Squarewell Block was mixed with 250 pL Buffer SB before transferring 750 pL onto the MN Nucleospin Soil Binding plates by centrifugation at 4,700g, RT for 5 min. The silica membrane was washed according to manufacturer’s instructions, though with centrifugations at 4,700g, and the binding plate was air-dried for 15 min. on collection tube strips. DNA was eluted by adding 50 pL 80 °C preheated SE buffer directly onto the silica membrane center, incubating for 10 min., and centrifuging at 4,700g, RT for 3 min. The elution was repeated for a final volume of 100 pL and DNA samples were stored in microcentrifuge tubes at -80 °C.
Shotgun metagenomic sequencing
Library preparation and 100PE (paired-end 100 bp) shotgun sequencing of genomic DNA from each fecal sample were performed using the BGISEQ-500 platform (Fang et al., 2018). Briefly, five hundred ng DNA was fragmented sonically using Covaris E220 (Covaris) to obtain 300-700 bp fragments, and all DNA was purified with the Axygen™ AxyPrep™ Mag PCR Clean-Up Kit, adding beads to each sample in a 1 :1 ratio (v/v) and eluting in 45 pL TE buffer. End-repair and A-tailing was performed on 20 ng of purified DNA using a 2:2:1 mixture of T4 DNA polymerase, T4 polynucleotide kinase, and rTaq DNA polymerase, before heat-inactivating the enzymes at 75 °C. Using T4 DNA ligase, adaptors with barcodes (Ad1532B) were ligated to DNA fragments at 23 °C and the barcoded fragments were amplified by PCR. To generate a circular singlestrand DNA library, 55 ng purified DNA was denatured at 95 °C and ligated at 37 °C with T4 DNA ligase. Libraries were pooled equimolarly in groups of eight to form DNA Nanoballs, which were each loaded into one lane for sequencing. Low quality reads were detected and filtered from the raw reads by an overall accuracy (OA)-based algorithm using default parameter (OAfragment=0.8), and human-derived reads were detected and filtered from the remaining reads by aligning to the GRCh19 (hg19) human genome assembly (Li et al., 2014)
Establishment of non-redundant gene catalog
Contigs were assembled de novo from paired reads of high quality and non-host origin using MEGAHIT (v1.1.1) (Li et al., 2015), procaryotic genes identified with Prodigal (Hyatt et al., 2010), and a de novo non-redundant gene catalogue created by clustering the complete genes using easy-linclust from MMseqs2 as detailed in Steinegger et al., 2017. Gene clustering was achieved using the “0” algorithm and a requirement of 95% sequence identity and 80% alignment coverage of query and target. Potential duplicate genes were reduced by filtering out genes found in less than three samples, and the remaining genes were clustered again with the same requirements for identity and coverage using the “2” algorithm. The resulting 5,139,318 genes were taxonomically annotated to bacteria, fungi, viruses, archaea, and protozoa by aligning to reference genomes from Refseq (2020-01-27) using nucleotide BLAST (v2.6.0;Camacho et al., 2009). For this purpose, only blast hits with sequence identity >45%, coverage >50%, and alignment length >100 bp were included and up to 50 hits fulfilling these requirements were stored per gene. Sequence identity requirements for taxonomic annotation were 95% for subspecies and species, 85% for genus, 75% for family, 65% for order, 55% for class, 50% for phylum, and 45% for superkingdom.
Identification of metagenomic species
To identify metagenomic species (MGS) in each sample, the paired high-quality reads of non-human origin were mapped individually to the non-redundant gene catalogue using the “bwa mem algorithm” from BWA (v0.7.16a; Li & Durbin., 2009; Li., 2014). Mapping criteria for the reads were sequence identity >95%, alignment length >80 bp, and mapping quality >20. MGS were then identified by canopy-based clustering (Nielsen et al., 2014), using a scaled gene count of 1 million genes per sample, where co-abundant genes were clustered based on a Pearson Correlation coefficient cut-off >0.9 and a canopy profile constituted by the 3rd quartile of the scaled gene count. For taxonomic annotation, co-abundant gene clusters comprising >100 genes were evaluated by summarizing taxonomy across all genes. As requirements for subspecies annotation, >75% of genes in a cluster had to be annotated to the same subspecies and <10% annotated to another subspecies. Same requirements were used for species annotation but differed for all higher taxonomical levels, for which the requirements were as follows: >60% annotated to the same taxa and <10% to another taxa for genus, >50% and <20% for family, >40% and <20% for order, >30% and <20% for class, >25% and <20% for phylum, and >20% and <15% for superkingdom. Each co- abundant gene cluster annotated at species or subspecies level or comprised by >500 genes was considered an MGS. A total of 919 MGS were identified across all samples, of which 8 were annotated at subspecies level, 387 at species level, 39 at genus level, 82 at family level, 245 at order level, 61 at class level, 32 at phylum level, and 55 at superkingdom level.
Calculation of MGS abundance
Abundances were calculated by mapping the paired high-quality reads of non-human origin to a subset of the non-redundant gene catalogue, which was subset by using the 100 genes within the co-abundant gene cluster of each MGS that had the highest Pearson correlation coefficient and did not appear in other MGS. Mapping was performed as described in the above section, and a minimum of 10 genes had to be detected for an MGS to be considered present in a sample. Relative abundance of each MGS was defined as the mean gene abundance across the gene subset of the MGS.
Identification of the load of bacteria expressing the gene aromatic lactate dehydrogenase (aldh)
Bacteria containing the aldh gene (SEQ ID NO: 1) were defined by sequence similarity, and summed to % abundance of aldh+ and aldh- bacteria respectively. The load of aldh+ and aldh- bacteria in individual samples was calculated by multiplying the % abundance with the load of bacteria/g feces. The maximum load in early life was calculated across week 1 to 6 months samples, and compared for children with (yes) or without (no) later development of indicated diseases.
Results
A higher load of bacteria possessing the gene aromatic lactate dehydrogenase (aldh) (SEQ ID NO: 1) was found in infants without food allergen sensitization and prescription of asthma medication later in life. Aldh+ bacterial load in early life fecal samples (1 week to 6 months) was higher in children without food allergen sensitization < 5 years postpartum (Figure 2b) and repeated prescriptions of asthma medication < 14 years postpartum (Figure 2d). A similar trend was seen for children with repeated prescriptions of allergic rhinitis medication < 14 years postpartum (Figure 2e), but not for diagnosis of atopic dermatitis at 2 years (Figure 2c). aldh- bacterial load did not associate with any of the outcomes.
Example 3: Reduced levels of 4-OH-PLA in fecal samples from infants between 2 and 6 months in children developing IgE-mediated diseases later in life
Materials and Methods
Detection of ILA, PLA and 4-OH-PLA in fecal and blood samples
Fecal water aliquots for metabolomics were centrifuged at 16,000g, 4 °C for 5 minutes. Supernatants were transferred to new tubes and centrifuged at 16,000g, 4 °C for 10 minutes. The resulting supernatants were stored at -80 °C in 100 pL aliquots until further sample preparation. Samples were thawed at 4 °C and centrifuged at 16,000g for 10 minutes. Subsequently, 80 pL supernatant was transferred to a new tube in which 20 pL internal standard mix (4 pg/mL) and 240 pL ice-cold acetonitrile were added. Samples were vortexed, incubated at -20 °C for 10 minutes, and then centrifuged at 16,000g, 4°C for 10 minutes. For each sample, 320 pL supernatant was transferred to a new tube and dried by nitrogen evaporation. Residues were redissolved in 80 pL sterile Milli-Q water for a final sample dilution of 1 :4 and internal standard concentration of 1 pg/mL, vortexed, and centrifuged at 16,000g, 4 °C for 5 minutes. Final supernatants were transferred to liquid chromatography vials with inserts and stored at -20 °C until analysis. 10 pL from 56 random samples (seven from each of the eight time points) were pooled for QC. Subsequently, targeted UPLC-MS was employed to quantify the aromatic lactic acids in the fecal water samples using isotopelabelled aromatic amino acids (Tyrosine-d4, Phenylalanine-d5, Tryptophan-d5, and Indoleacetic acid-d2) as internal standards as previously described (Laursen et al., 2021).
Mediation analysis
Causal mediation analyses were conducted to determine whether 4-OH-PLA or PLA mediate the association between aldh+ bifidobacteria and food allergen sensitization until 5 years of age. Figures 4A and 4B present the mediation analyses. Path a, b, and c in Figure 4C represent analyses preceding the mediation analyses, which must all be significant for a mediation analysis to be valid. Path a denotes the correlation between max early life (0-6m) fecal aldh+ relative abundance and the 2 months metabolite levels (determined by a linear model), path b is the association between the 2 months metabolite levels and food allergen sensitization until 5 years of age (determined by a generalized linear model), and path c is the association between max early life (0-6m) fecal aldh+ relative abundance and food allergen sensitization until 5 years (determined by a generalized linear model). Path c’ in Figure 4C corresponds to the direct effect from the mediation analyses, which is the direct effect of max early life (0- 6m) fecal aldh+ relative abundance on food allergen sensitization until 5 years when the metabolite is considered as a mediator. Thus, while path c analyses the direct effect of the aldh+ bifidobacteria on food allergen sensitization, path c’ does the same but with consideration of whether the effect is driven by the mediator. Since no significant effect was observed (c’ was not significant) of aldh+ bifidobacteria on food allergen sensitization when considering the 2 months 4-OH-PLA levels, and the mediation analysis (Figure 4A) demonstrated a significant causal mediation effect, a complete mediation through 4-OH-PLA was observed, while PLA did not mediate the effect.
Example 3a: High fecal levels of 4-OH-PLA in infants at 2-6 months of age decreased the risk of later development of IgE-mediated diseases.
Surprisingly, children that did not develop several IgE-mediated atopic diseases during childhood had higher fecal levels of 4-OH-PLA, but did not have higher fecal levels of ILA and PLA. Higher fecal levels of 4-OH-PLA were observed at 2 months of age in children who did not develop food allergen sensitization < 5 years postpartum (Figure 3a) or atopic dermatitis at 2 years postpartum (Figure 3b). Higher maximum fecal levels of 4-OH-PLA at 2-6 months of age were also observed in children without repeated prescriptions of asthma medication < 14 years postpartum (Figure 3c) and repeated prescriptions of allergic rhinitis medication < 14 years postpartum (Figure 3d).
Children that did not develop food allergen sensitization < 5 years postpartum also had higher fecal levels of PLA (Figure 3e), while no differences were identified for fecal PLA levels in children with or without later dermatitis at 2 years (Figure 3f), or children with or without repeated prescriptions of asthma or allergic rhinitis medicine < 14 years (Figure 3g-h). Likewise, there were no differences in fecal levels of ILA at 2 months between children with and without later food allergen sensitization < 5 years postpartum or atopic dermatitis at 2 years (Figure 3i-j), and no differences in maximum fecal levels of ILA at 2-6 months between children with and without later repeated prescriptions of asthma or allergic rhinitis medicine (Figure 3k-l).
Importantly, among the two aromatic lactic acids displaying higher fecal levels at 2 months in children that did not develop food allergen sensitization < 5 years postpartum (4-OH-PLA (Figure 3A) and PLA (Figure 3E)), 4-OH-PLA was shown in a mediation analysis to completely mediate the association between aldh+ bifidobacteria and no food allergen sensitization < 5 years postpartum (Figure 4a, P = 0,002). Whereas, PLA, which also showed no in vitro effect, was demonstrated to not mediate the association (Figure 4b, P = 0.122). This demonstrates that 4-OH-PLA is the only one of the three gut bacteria-derived aromatic lactic acids with a causal mediating effect on the reduction of IgE-mediated diseases in vivo.
It was surprising and unexpected to observe that only 4-OH-PLA decreases the risk of later IgE-mediated disease development in vivo, considering that i) ILA has previously been reported to hold anti-inflammatory properties (Meng et al., 2020; Ehrlich et al., 2020; Laursen et al., 2021), ii) fecal ILA levels surpassed those of 4-OH-PLA and PLA (Figure 5), iii) observed plasma levels of ILA should be sufficient to activate the HCAR3 receptor given the ECso of 0.18 pM (Peters et al., 2019; Figure 6), iv) ILA inhibited in vitro IgE production in plasma B cells to a similar or higher degree than 4-OH-PLA did at concentrations above 5 pM (Figure 7, Figure 8 and Figure 9), and v) both ILA and 4- OH-PLA inhibited IL-12p70 production in human monocytes (Figure 10). 3b: Lower levels of 4-OH-PLA in fecal between 2 and 6 months in infants developing IgE-mediated diseases later in life
Levels of the three ALAs (PLA, ILA and 4-OH-PLA) and their sum in fecal samples of infants at 2 months of age (Figure 5a) and maximum levels between 2 and 6 months of age (Figure 5b) stratified via clinical outcome later in life are presented in Figure 5.
Total ALA levels: The summed 2 months fecal levels of ALAs ranged from about 1-600 nmol/g feces (Figure 5a), while the maximum summed levels of ALAs in infant fecal samples between 2 and 6 months of age (Figure 5b) ranged from about 40 to 680 nmol/g feces.
4-OH-PLA: Increased levels of 4-OH-PLA were linked to reduced risk of development of IgE-mediated atopic diseases in children (Figure 3 and Figure 4). Children that did not develop food allergen sensitization during the first 5 years of life displayed mean fecal levels of 4-OH-PLA above 61 nmol/g feces at 2 months of age, whereas children that did develop food allergen sensitization during the first 5 years of age displayed mean fecal levels of 4-OH-PLA below 37 nmol/g feces at 2 months of age (Figure 5a).
Children that were not diagnosed with atopic dermatitis at 2 years displayed mean fecal levels of 4-OH-PLA above 54 nmol/g feces at 2 months of age, whereas those diagnosed with atopic dermatitis at 2 years displayed mean levels of 4-OH-PLA below 34 nmol/g feces (Figure 5a).
Children that did not receive asthma medicine during the first 14 years postpartum displayed mean maximum 2-6 months fecal levels of 4-OH-PLA above 100 nmol/g feces, whereas children that did received more than one prescription for asthma medication during the first 14 years postpartum displayed mean maximum 2-6 months fecal levels of 4-OH-PLA below 61 nmol/g feces (Figure 5b).
Children that did not receive allergic rhinitis medication during the first 14 years postpartum displayed mean maximum 2-6 months fecal levels of 4-OH-PLA above 100 nmol/g feces, whereas children that did receive more than one prescription for allergic rhinitis medication during the first 14 years postpartum displayed mean maximum 2-6 months fecal levels of 4-OH-PLA below 72 nmol/g feces (Figure 5b). Example 3c: ILA levels detected in plasma at 6 months of age correlate with relative abundance of fecal aldh+ bacteria and with fecal ILA levels at 6 months of age.
Correlations between plasma concentrations (pM) of ILA and ILA concentrations in fecal water (nmol/g feces) in subjects at 6 months of age are presented in Figure 6. A concentration of 1 pM plasma ILA corresponded to approximately 30% of fecal aldh+ bacteria at 6 months of age (Figure 6a). Infants with above 95% of aldh+ gut bacteria at 6 months of age displayed plasma ILA concentrations up to 3 pM ILA (Figure 6a). 1 pM plasma ILA corresponds to ca. 40 nmol ILA/g feces (Figure 6b).
Example 4: Direct and consistent inhibitory effect of both ILA and 4-OH-PLA, but not PLA, on IgE production in human plasma B cells
Materials and Methods
Human blood samples
Human buffy coats were acguired from the Copenhagen University Hospital (Rigshospitalet, Copenhagen) from healthy anonymous donors. Use of the buffy coat material from healthy anonymous donors was approved by the Blood bank at Rigshospitalet, Copenhagen, under the jurisdiction of Region H, and handled in accordance with guidelines put forward in the “Transfusion Medicine Standards” by the Danish Society for Clinical Immunology. Prior written informed consent was obtained according to the Declaration of Helsinki.
Isolation of PBMCs and stimulation of IgE production in B cells
Peripheral blood mononuclear cells (PBMCs) were isolated from buffy coats using Ficoll-Pague (GE Healthcare) density centrifugation and used either fresh or cryopreserved at -140 °C in heat-inactivated fetal bovine serum (FBS; Lonza) with 10% DMSO until analysis. PBMCs were used directly or gently thawed on the day of experiment and plated at 250,000 PBMCs/well in preheated culture medium (RPMI 1640 (gibco) with 10% heat-inactivated FBS (Lonza), 1% penicillin/streptomycin (Lonza), 2 mM L-glutamine (Lonza), and 50 pM 2-ME (gibco)). Modifiers (ILA (Sigma- Aldrich I5508), 4-OH-PLA (Sigma-Aldrich H3253), D-PLA (Sigma-Aldrich 376906), or L- PLA (Sigma-Aldrich 113069)) dissolved in a maximum of 0.1% DMSO in culture medium were added to the PBMCs for final concentrations of 5 pM, 50 pM, 200 pM, or 1000 pM. A vehicle control (0.1% DMSO) was also included. B cells in the PBMC suspension were then activated for IgE production by adding an activation solution containing rhlL-4 (50 ng/mL, Miltenyi Biotec), CD40L (50 ng/mL, R&D systems), and cross-linking HA-Tag antibody (100 ng/mL or 500 ng/mL, R&D systems) in culture medium, and incubated in a humidified 37 °C, 5% CO2 incubator for a total of 10 days. At day 4 and 7, the medium was replenished by removing 50% of the supernatant and adding fresh culture medium with modifier followed by fresh culture medium with activation solution for the original assay concentration and volume. Cell-free supernatants were harvested and immediately stored at -80 °C until analysis, and cells were subjected to viability testing.
Viability testing
Cellular viability was examined using Live/Dead Fixable Violet stain (Thermofisher, cat. No. L34955) and count beads (123count eBeads), and analyzed using MACSQuant 16.
IgE determination
IgE production was quantified by IgE ELISA (Invitrogen) performed according to the manufacturer’s instructions.
Results
The direct effect of ILA and 4-OH-PLA on IgE production in human plasma B cells is demonstrated in Figure 7. Co-incubation with 200 pM ILA or 4-OH-PLA resulted in at least 50% inhibition of IgE production in human plasma B cells.
Additionally, IgE production in human plasma B cells is directly inhibited by ILA in a dose-dependent manner (Figure 8). ILA suppresses IgE production on average 58- 92% in the concentration range between 5 to 1000 pM. 4-OH-PLA has the biggest effect on IgE inhibition between 5 and 50 pM, with average suppressive effects of 60% in said range, while at concentrations of 200 and 1000 pM, a reduced effect was observed, with an average IgE inhibition of 26-37%.
The direct effect of ILA vs 4-OH-PLA vs PLA on IgE production in human plasma B cells is demonstrated in Figure 9. Cryopreserved human blood-derived B cells were thawed and stimulated for 10 days with 50 ng/mL CD40L, 100 ng/mL cross-linking HA antibody and 50 ng/mL rhlL-4 to induce IgE production. Following incubation with 200 pM ILA or 4-OH-PLA, IgE production from human plasma B cells was reduced consistently, whereas incubation with 200 pM L-PLA or D- PLA did not inhibit IgE production from human plasma B cells consistently. Cell viability was unaltered at these compound concentrations. Thus, these data demonstrate that specific ALAs; ILA and 4-OH-PLA have the capacity to inhibit IgE production from plasma B cells consistently and effectively across donors, whereas PLA does not consistently inhibit IgE production from B cells across donors.
Example 5: ILA and 4-OH-PLA, but not PLA, have an anti-inflammatory effect in LPS- primed human monocytes.
Materials and Methods
Monocyte experiment
PBMCs were isolated from buffy coats using Ficoll-Paque (GE Healthcare) density centrifugation, and monocytes were isolated from the PBMCs for a purity >92% using a Pan Monocyte Isolation kit (Miltenyi Biotec). The purity was determined by flow cytometry (MACSQuant 16) on cells stained with 1:25 dilutions of CD14-PE-Cy7 (eBiosciences) and CD16-FITC (Biolegend). Monocytes were incubated in culture medium (RPMI 1640 (Lonza) with 10% heat-inactivated fetal bovine serum (Lonza), 1% penicillin/streptomycin (Lonza), 2 mM L-glutamine (Lonza), and 50 pM 2-ME (Sigma- Aldrich)) in a humidified 37 °C, 5% CO2 incubator. Modifiers (ILA (Sigma-Aldrich I5508), 4-OH-PLA (Sigma-Aldrich H3253), D-PLA (Sigma-Aldrich 376906), or L-PLA (Sigma-Aldrich 113069) dissolved in a maximum of 0.1% DMSO in culture medium were added to the monocytes for a final concentration of 200 pM. A vehicle control (0.1% DMSO) was also included. Monocytes were then stimulated with lipopolysaccharide (LPS, TLR4 ligand, E. coli 026: B6, Sigma L2654) and IFN-y (RD285-IF-100) at final concentrations of 100 ng/mL of both, and incubated for 18 hours. Cellular viability was examined using Live/Dead Fixable Violet stain (Thermofisher, cat. No. L34955) and count beads (123count eBeads), analyzed using MACSQuant 16 and gated using FlowJo. Cell-free supernatants were harvested and immediately stored at -80 °C until analysis. I L-12p70 was quantified by ELISA according to manufacturer’s instructions (R&D systems). Results
The effect of ILA, 4-OH-PLA and PLA on the production of the pro-inflammatory cytokine; I L-12p70, in primary LPS-primed human monocytes is demonstrated in Figure 10. ILA and 4-OH-PLA inhibited I L-12p70 production in primary LPS-primed human monocytes, whereas D- and L-PLA had no significant effect.
Sequence overview
SEQ ID NO:1 - aldh (WT gene, Bifidobacterium longurri) tcacagcagc ccctcgcagt gttcacgcac gagttcggcg gaatggtgca gtttggcccg ctcgtcctcg ctcagatcga gttcgacgat ctcgttggca ccattggcgc gcagttcagt ggggacgccg aggaacacgt catgttcgcc gtattcgccg tccagcaggg tggagacggg cacgatgcgg cgttcgtccc acaggatggt ctgtacgatg ccggccacgg tggaggcgat gccgaagttg gtgccgccct tggccgcgac gatctcattg ccacgggtgc gggtcttctc ttcgatttcg gtggtggaca cggaggcgaa acggtcttgg ttgtcggcca ggaagcgggc gaatggcttg ccgccgagcg agacggtgga ccatgcggtg aactgggagt cgccgtgctc gcccatcacg aagccgccca cgttgcgcgg gtcgaggccg gtctcttcgc cgatgatggt cttgagacgc gaggtgtcga gcgcggtgcc ggtaccgagc acctgggtgc gaggcagacc ggaacgtttc caggcatacc aagccatcac gtcgaccggg ttggagacca tgacgatcac gccgtcaaaa ccggaggcca tgacgttgtc gaccacttcg cccacaagac ccacggtgaa gccgagttcg gccatgcggt tggagttggc cggcggcttg cggcccacgg tgatgacgac gatgtccgca tccttgcagt cggcgtagtc gccggcgcgc accttgacgt ggcggtcctg gaattcgctg ccatcatcga ggtcgcgggc ctcccccagg gccttggcgg cggaacagtc gatgagcaca agctcattgc acaggccgtg cgtgacgatg ccgaaggcgg cggtggcacc gacccggccg gtgccgacga taacgacttt gttgcggttc atagtgacca t
References
Stenius, F. et al. Lifestyle factors and sensitization in children - the ALADDIN birth cohort. Allergy 66, 1330-1338 (2011).
Nilsson, S. F., Lilja, G., Jarnbert-Pettersson, H. & Alm, J. Relevance of low specific IgE levels to egg, milk and peanut in infancy. Clin Exp Allergy 49, 308-316 (2019). Ballardini, N., Nilsson, C., Nilsson, M. & Lilja, G. ImmunoCAP Phadiatop lnfant--a new blood test for detecting IgE sensitisation in children at 2 years of age. Allergy 61, 337-343 (2006). Severity scoring of atopic dermatitis: the SCORAD index. Consensus Report of the European Task Force on Atopic Dermatitis. Dermatology. 1993; 186(1 ):23-31. doi: 10.1159/000247298. PMID: 8435513.
Fang, C. et al. Assessment of the cPAS-based BGISEQ-500 platform for metagenomic sequencing. Gigascience 7, 1-8 (2018)
Li, J. et al. An integrated catalog of reference genes in the human gut microbiome. Nature Biotechnology 32, 834-841 (2014)
Li, D., Liu, C. M., Luo, R., Sadakane, K. & Lam, T. W. MEGAHIT: an ultra-fast singlenode solution for large and complex metagenomics assembly via succinct de Bruijn graph. Bioinformatics 31 , 1674-1676 (2015)
Hyatt, D. et al. Prodigal: prokaryotic gene recognition and translation initiation site identification. BMC Bioinformatics 11 , 119 (2010).
Steinegger, M. & Sdding, J. MMseqs2 enables sensitive protein sequence searching for the analysis of massive data sets. Nat Biotechnol 35, 1026-1028 (2017).
Camacho, C. et al. BLAST+: architecture and applications. BMC Bioinformatics 10, 421 (2009)
Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754-1760 (2009).
Li, H. Toward better understanding of artifacts in variant calling from high-coverage samples. Bioinformatics 30, 2843-2851 (2014).
Nielsen, H. B. et al. Identification and assembly of genomes and genetic elements in complex metagenomic samples without using reference genomes. Nat Biotechnol 32, 822-828 (2014).
Laursen, M. F. et al. Bifidobacterium species associated with breastfeeding produce aromatic lactic acids in the infant gut. Nat Microbiol 6, 1367-1382 (2021).
Prescott, SL et al. Development of allergen-specific T-cell memory in atopic and normal children. Lancet 353, 196-200 (1999).
Meng D, Sommella E, Salviati E, et al (2020) lndole-3-lactic acid, a metabolite of tryptophan, secreted by Bifidobacterium longum subspecies infantis is anti- inflammatory in the immature intestine. Pediatr Res 88(2):209-217.
Ehrlich AM, Pacheco AR, Henrick BM, et al (2020) lndole-3-lactic acid associated with Bifidobacterium- dominated microbiota significantly decreases inflammation in intestinal epithelial cells. BMC Microbiol 20(1):357. Laursen MF, Sakanaka M, von Burg N, et al (2021) Bifidobacterium species associated with breastfeeding produce aromatic lactic acids in the infant gut. Nat Microbiol 6(11):1367-1382.
Peters A, Krumbholz P, Jager E, Heintz-Buschart A, Qakir MV, Rothemund S, Gaudl A, Ceglarek II, Schdneberg T, Staubert C. Metabolites of lactic acid bacteria present in fermented foods are highly potent agonists of human hydroxycarboxylic acid receptor 3. PLoS Genet. 2019 May 23;15(5):e1008145. doi: 10.1371/journal.pgen.1008145.
Erratum in: PLoS Genet. 2019 Jul 19; 15(7):e1008283. doi:
10.1371/journal.pgen.1008283. PMID: 31120900; PMCID: PMC6532841.

Claims

Claims
1. A compound of formula (1a) formula (1a), or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of an IgE-mediated disease in a subject.
2. A composition for use in the treatment and/or prevention of an IgE-mediated disease in a subject, the composition comprising a compound of formula (1a).
3. The compound or the composition for use according to any one of the preceding claims, wherein the compound is 4-hydroxyphenyllactic acid or a pharmaceutically acceptable salt thereof.
4. The compound or the composition for use according to any one of the preceding claims, wherein the subject is a human.
5. The compound or the composition for use according to any one of the preceding claims, wherein the subject is a non-infant.
6. The compound or the composition for use according to any one of the preceding claims, wherein the subject is an infant.
7. The compound or the composition for use according to any one of the preceding claims, wherein the subject is a pre-term infant or a term infant.
8. The compound or the composition for use according to any one of the preceding claims, wherein the infant is an infant born by C-section.
9. The compound or the composition for use according to any one of the preceding claims, wherein the infant is born from a first-time mother.
10. The compound or the composition for use according to any one of the preceding claims, wherein the infant has one or both parents affected by an IgE-mediated disease.
11. The compound or the composition for use according to any one of the preceding claims, wherein the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a low abundance of Aldh+ bacteria.
12. The compound or the composition for use according to any one of the preceding claims, wherein said Aldh+ bacteria comprises an Aldh gene encoding the enzyme aromatic lactate dehydrogenase, preferably an Aldh of SEQ ID NO: 1.
13. The compound or the composition for use according to any one of the preceding claims, wherein the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has an abundance of Aldh+ bacteria of at most 10% of total bacteria, such as at most 9% of total bacteria, such as at most 8% of total bacteria, such as at most 7% of total bacteria, such as at most 6% of total bacteria, such as at most 5% of total bacteria, such as at most 4% of total bacteria, such as at most 3% of total bacteria, such as at most 2% of total bacteria, such as at most 1 % of total bacteria.
14. The compound or the composition for use according to any one of the preceding claims, wherein the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has an abundance of Aldh+ bacteria of at most 5% of total bacteria.
15. The compound or the composition for use according to any one of the preceding claims, wherein the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a low load of Aldh+ bacteria.
16. The compound or the composition for use according to any one of the preceding claims, wherein the infant has a dysbiotic gut microbiota, wherein the dysbiotic gut has a load of Aldh+ bacteria of at most 3x109 Aldh+ bacteria/g feces.
17. The compound or the composition for use according to any one of the preceding claims, wherein the infant is between 3 weeks and 6 months of age, such as between 3 weeks and 5 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 3 months of age, such as between 3 weeks and 2 months of age, such as between 3 weeks and 1 month of age, such as between 1 month and 6 months of age, such as between 2 month and 6 months of age, such as between 3 month and 6 months of age, such as between 4 month and 6 months of age.
18. The compound or the composition for use according to any one of the preceding claims, wherein the infant has at most 3x109 Aldh+ bacteria/g feces.
19. The compound or the composition for use according to any one of the preceding claims, wherein the infant has at most 100 nmol/g feces of at least one aromatic lactic acid.
20. The compound or the composition for use according to any one of the preceding claims, wherein the infant has at most 100 nmol/g feces of at least one aromatic lactic acid, corresponding to a plasma concentration of at most 3 pM.
21 . The compound or the composition for use according to any one of the preceding claims, wherein the infant has at most 100 nmol/g feces of at least one aromatic lactic acid, wherein the concentration of the at least one aromatic lactic acid is determined by LC-MS.
22. The compound or the composition for use according to any one of the preceding claims, wherein the infant has at most 100 nmol/g feces of at least one aromatic lactic acid, such as is 4-hydroxyphenyllactic acid and/or indole-3- lactic acid and/or phenyllactic acid.
23. The compound or the composition for use according to any one of the preceding claims, wherein the infant has at most 100 nmol/g feces of the compound of formula (1a),
24. The compound or the composition for use according to any one of the preceding claims, wherein the infant has at most 100 nmol/g feces of 4- hydroxyphenyllactic acid in the gut.
25. The compound or the composition for use according to any one of the preceding claims, wherein the infant has at most 100 nmol/g feces of 4- hydroxyphenyllactic acid in the gut at between 3 weeks and 6 months of age, such as between 2-5 months of age.
26. The compound or the composition for use according to any one of the preceding claims, wherein the infant has less than 100 nmol/g feces of the compound of formula 1b, formula (1b), in the gut.
27. The compound or the composition for use according to any one of the preceding claims, wherein the infant has less than 100 nmol/g feces of indole-3- lactic acid in the gut.
28. The compound or the composition for use according to any one of the preceding claims, wherein the infant has less than 100nmol/g feces of the compound of formula (1c), formula (1c), in the gut.
29. The compound or the composition for use according to any one of the preceding claims, wherein the infant has less than 100nmol /g feces of phenyllactic acid in the gut.
30. The compound or the composition for use according to any one of the preceding claims, wherein the infant has less than 100nmol/g feces of the compound or indole-3-lactic acid in the gut between 3 weeks and 6 months of age, such as between 3 weeks and 5 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 4 months of age, such as between 3 weeks and 3 months of age, such as between 3 weeks and 2 months of age, such as between 3 weeks and 1 month of age, such as between 1 month and 6 months of age, such as between 2 month and 6 months of age, such as between 3 month and 6 months of age, such as between 4 month and 6 months of age.
31. The compound or the composition for use according to any of the preceding claims, wherein the compound is administered in an amount which is effective to decrease IgE production.
32. The compound or the composition for use according to any of the preceding claims, wherein the compound decreases IgE production, such as allergenspecific IgEproduction, in human B cells and/or plasma B cells.
33. The compound or the composition for use according to any of the preceding claims, wherein the compound the compound decreases systemic IgE production, such as allergen-specific IgE production, to 0.50 kll/L plasma IgE or below, such as to 0.35 kll/L or below, such to 0.1 kll/L or below.
34. The compound or the composition for use according to any of the preceding claims, wherein the compound decreases in vitro IgE production in human B cells and/or plasma cells to at most 25 ng/mL.
35. The compound or the composition for use according to any of the preceding claims, wherein the compound decreases in vitro IgE production in human B cells and/or plasma cells, wherein IgE production is determined by human B cell assays.
36. The compound or the composition for use according to any of the preceding claims, wherein the compound decreases IgE production in human B cells.
37. The compound or the composition for use according to any of the preceding claims, wherein the compound decreases in vitro IgE production in human B cells to at most 25 ng/mL.
38. The compound or the composition for use according to any of the preceding claims, wherein the compound decreases in vitro IgE production in plasma cells.
39. The compound or the composition for use according to any of the preceding claims, wherein the compound decreases in vitro IgE production in plasma cells to at most 25 ng/mL.
40. The compound or the composition for use according to any of the preceding claims, wherein the compound decreases systemic allergen-specific IgE production to 0.50 kll/L plasma IgE or less.
41. The compound or the composition for use according to any of the preceding claims, wherein the compound decreases systemic allergen-specific IgE production to 0.50 kll/L plasma IgE or less, such as to 0.35 kU/L or less, such as 0.1kU/L or less.
42. The compound or the composition for use according to any of the preceding claims, wherein the compound the compound decreases systemic IgE production of at most 0.50 kU/L plasma allergen-specific IgE.
43. The compound or the composition for use according to any of the preceding claims, wherein the compound decreases systemic IgE production to 0.50 kU/L or less plasma allergen-specific IgE, such as to 0.35 kll/L or less, such as to 0.1kll/L or less.
44. The compound or the composition for use according to any of the preceding claims, wherein allergen-specific IgE production in human B cells and/or plasma cells is decreased systemically to 0.35 kll/L or less, such as to 0.1 kll/L or less.
45. The compound or the composition for use according to any of the preceding claims, wherein the IgE-mediated disease is selected from the group consisting of: an allergic response to an allergen, allergic asthma, urticaria, angioedema, food allergy, an allergic response, atopic dermatitis, anaphylaxis, cutaneous mastocystosis, allergic rhinitis, allergic rhinoconjuncvititis, nasal polyposis, Kimura's disease, eosinophilic otitis media, eosinophilic gastroenteritis, latex allergy, bronchopulmonary allergic aspergillosis, or a combination thereof.
46. The compound or the composition for use according to any of the preceding claims, wherein the IgE-mediated disease is acute or chronic.
47. The compound or the composition for use according to any of the preceding claims, wherein the composition is administered to the subject prophylactically.
48. The compound or the composition for use according to any of the preceding claims, wherein the composition is administered to the subject orally and/or parenterally.
49. The compound or the composition for use according to any of the preceding claims, wherein the composition is administered to the subject orally.
50. The compound or the composition for use according to any of the preceding claims, wherein the composition is administered to the subject parenterally.
51. The compound or the composition for use according to any of the preceding claims, wherein the composition is administered to the subject via intramuscular, and/or subcutaneous and/or IV administration.
52. The compound or the composition for use according to any of the preceding claims, wherein the composition is administered with at least one or more therapeutic agent(s).
53. The compound or the composition for use according to any of the preceding claims, wherein the at least one or more therapeutic agent(s) is the standard of care treatment for IgE-mediated disease.
54. The compound or the composition for use according to any of the preceding claims, wherein the at least one or more therapeutic agent(s) is administered simultaneously, sequentially or separately.
55. The compound or the composition for use according to any of the preceding claims, wherein the composition is a pharmaceutical composition.
56. The compound or the composition for use according to any of the preceding claims, wherein a therapeutically effective amount of the composition is administered to the subject.
57. The compound or the composition for use according to any of the preceding claims, wherein a therapeutically effective amount of the composition is administered to the infant.
58. A compound of formula (1a) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in a subject, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kll/L, and, b. Administering a composition comprising a compound of formula (1a) or a pharmaceutically acceptable salt thereof, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM.
59. A compound of formula (1a) or a pharmaceutically acceptable salt thereof, for use in a method of reducing the risk of IgE-mediated disease in a subject, the method comprising: c. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kll/L, and, d. Administering a composition comprising a compound of formula (1a) or a pharmaceutically acceptable salt thereof, to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM and/or plasma IgE levels of 0.5 kll/L or below.
60. A method of reducing the risk of IgE-mediated disease in a subject, comprising: e. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kU/L, and, f. Administering a composition comprising a compound of formula (1a) or a pharmaceutically acceptable salt thereof, to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM.
61. A method of reducing the risk of IgE-mediated disease in a subject, comprising: g. Identifying a reduced total level of aromatic lactic acids in the gut and/or an allergen-specific plasma IgE level above 0.35 kU/L, and, h. Administering a composition comprising a compound of formula (1a) or a pharmaceutically acceptable salt thereof, to the subject, thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM and/or plasma IgE levels or 0.5 kU/L or below.
62. A compound of formula (1a) or a pharmaceutically acceptable salt thereof, formula (1a), wherein;
A represents a substituted or unsubstituted heteroaromatic ring or a substituted aromatic ring, for use in a method of reducing the risk of IgE-mediated disease in a subject, the method comprising: a. Identifying a reduced total level of aromatic lactic acids in the gut and/or a systemic allergen-specific I g E level above 0.35 kll/L, and, b. Administering a composition comprising one or more compounds of formula (II) or a pharmaceutically acceptable salt thereof to the subject thereby achieving a total level of aromatic lactic acids in plasma of at least 5 pM.
PCT/EP2025/051848 2024-01-26 2025-01-24 4-hydroxyphenyllactic acid for use in treating and reducing the risk of ige-mediated diseases Pending WO2025158027A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP24154241.4 2024-01-26
EP24154241 2024-01-26

Publications (1)

Publication Number Publication Date
WO2025158027A1 true WO2025158027A1 (en) 2025-07-31

Family

ID=89767776

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2025/051848 Pending WO2025158027A1 (en) 2024-01-26 2025-01-24 4-hydroxyphenyllactic acid for use in treating and reducing the risk of ige-mediated diseases

Country Status (1)

Country Link
WO (1) WO2025158027A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008120713A1 (en) * 2007-03-30 2008-10-09 Suntory Holdings Limited Medicinal composition, food or drink having effect of enhancing parasympathetic nervous actions
WO2022232661A1 (en) * 2021-04-29 2022-11-03 Evolve Biosystems, Inc. Personalized immunotherapy using intestinal metabolites
JP2023034914A (en) * 2021-08-31 2023-03-13 森永乳業株式会社 Composition for promoting production of aromatic lactic acid compound
CN117205206A (en) * 2023-08-16 2023-12-12 江南大学 A tryptophan metabolite that can improve the pathological characteristics of asthmatic mice and its application

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008120713A1 (en) * 2007-03-30 2008-10-09 Suntory Holdings Limited Medicinal composition, food or drink having effect of enhancing parasympathetic nervous actions
WO2022232661A1 (en) * 2021-04-29 2022-11-03 Evolve Biosystems, Inc. Personalized immunotherapy using intestinal metabolites
JP2023034914A (en) * 2021-08-31 2023-03-13 森永乳業株式会社 Composition for promoting production of aromatic lactic acid compound
CN117205206A (en) * 2023-08-16 2023-12-12 江南大学 A tryptophan metabolite that can improve the pathological characteristics of asthmatic mice and its application

Non-Patent Citations (20)

* Cited by examiner, † Cited by third party
Title
"Severity scoring of atopic dermatitis: the SCORAD index. Consensus Report of the European Task Force on Atopic Dermatitis.", DERMATOLOGY, vol. 186, no. 1, 1993, pages 23 - 31
BALLARDINI, N.NILSSON, C.NILSSON, MLILJA, G.: "ImmunoCAP Phadiatop Infant--a new blood test for detecting IgE sensitisation in children at 2 years of age", ALLERGY, vol. 61, 2006, pages 337 - 343, XP071459856, DOI: 10.1111/j.1398-9995.2005.00936.x
CAMACHO, C. ET AL.: "BLAST+: architecture and applications.", BMC BIOINFORMATICS, vol. 10, 2009, pages 421, XP021065535
EHRLICH AMPACHECO ARHENRICK BM ET AL.: "Indole-3-lactic acid associated with Bifidobacterium- dominated microbiota significantly decreases inflammation in intestinal epithelial cells.", BMC MICROBIOL, vol. 20, no. 1, 2020, pages 357, XP093092778, DOI: 10.1186/s12866-020-02023-y
FANG, C. ET AL.: "Assessment of the cPAS-based BGISEQ-500 platform for metagenomic sequencing.", GIGASCIENCE, vol. 7, 2018, pages 1 - 8
HYATT, D. ET AL.: "Prodigal: prokaryotic gene recognition and translation initiation site identification", BMC BIOINFORMATICS, vol. 11, 2010, pages 119, XP021071474, DOI: 10.1186/1471-2105-11-119
LAURSEN MARTIN F ET AL: "Bifidobacterium species associated with breastfeeding produce aromatic lactic acids in the infant gut", NATURE MICROBIOLOGY, NATURE PUBLISHING GROUP UK, LONDON, vol. 6, no. 11, 21 October 2021 (2021-10-21), pages 1367 - 1382, XP037603459, DOI: 10.1038/S41564-021-00970-4 *
LAURSEN MFSAKANAKA MVON BURG N ET AL.: "Bifidobacterium species associated with breastfeeding produce aromatic lactic acids in the infant gut.", NAT MICROBIOL, vol. 6, no. 11, 2021, pages 1367 - 1382, XP037603459, DOI: 10.1038/s41564-021-00970-4
LI, D.LIU, C. M.LUO, R.SADAKANE, K.LAM, T. W.: "MEGAHIT: an ultra-fast single-node solution for large and complex metagenomics assembly via succinct de Bruijn graph.", BIOINFORMATICS, vol. 31, 2015, pages 1674 - 1676, XP055469800, DOI: 10.1093/bioinformatics/btv033
LI, H.: "Toward better understanding of artifacts in variant calling from high-coverage samples", BIOINFORMATICS, vol. 30, 2014, pages 2843 - 2851
LI, H.DURBIN, R.: "Fast and accurate short read alignment with Burrows-Wheeler transform", BIOINFORMATICS, vol. 25, 2009, pages 1754 - 1760, XP055553969, DOI: 10.1093/bioinformatics/btp324
LI, J.: "An integrated catalog of reference genes in the human gut microbiome.", NATURE BIOTECHNOLOGY, vol. 32, 2014, pages 834 - 841, XP093174999, DOI: 10.1038/nbt.2942
MENG DSOMMELLA ESALVIATI E ET AL.: "Indole-3-lactic acid, a metabolite of tryptophan, secreted by Bifidobacterium longum subspecies infantis is anti-inflammatory in the immature intestine", PEDIATR RES, vol. 88, no. 2, 2020, pages 209 - 217
NIELSEN, H. B. ET AL.: "Identification and assembly of genomes and genetic elements in complex metagenomic samples without using reference genomes.", NAT BIOTECHNOL, vol. 32, 2014, pages 822 - 828
NILSSON, S. F.LILJA, G.JARNBERT-PETTERSSON, H.ALM, J.: "Relevance of low specific IgE levels to egg, milk and peanut in infancy.", CLIN EXP ALLERGY, vol. 49, 2019, pages 308 - 316, XP071888315, DOI: 10.1111/cea.13273
PETERS AKRUMBHOLZ PJAGER EHEINTZ-BUSCHART ACAKIR MVROTHEMUND SGAUDL ACEGLAREK USCH6NEBERG TSTÄUBERT C.: "Metabolites of lactic acid bacteria present in fermented foods are highly potent agonists of human hydroxycarboxylic acid receptor 3.", PLOS GENET., vol. 15, no. 5, 23 May 2019 (2019-05-23), pages e1008145
PLOS GENET., vol. 15, no. 7, 19 July 2019 (2019-07-19), pages e1008283
PRESCOTT, SL ET AL.: "Development of allergen-specific T-cell memory in atopic and normal children", LANCET, vol. 353, 1999, pages 196 - 200, XP005017870, DOI: 10.1016/S0140-6736(98)05104-6
STEINEGGER, M.SÖDING, J.: "MMseqs2 enables sensitive protein sequence searching for the analysis of massive data sets.", NAT BIOTECHNOL, vol. 35, 2017, pages 1026 - 1028
STENIUS, F. ET AL.: "Lifestyle factors and sensitization in children - the ALADDIN birth cohort.", ALLERGY, vol. 66, 2011, pages 1330 - 1338, XP071461324, DOI: 10.1111/j.1398-9995.2011.02662.x

Similar Documents

Publication Publication Date Title
Edwinson et al. Gut microbial β-glucuronidases regulate host luminal proteases and are depleted in irritable bowel syndrome
Danne et al. Neutrophils: from IBD to the gut microbiota
Diez-Martin et al. Inflammatory bowel disease: a comprehensive analysis of molecular bases, predictive biomarkers, diagnostic methods, and therapeutic options
Li et al. Fecal microbiota transplantation (FMT) could reverse the severity of experimental necrotizing enterocolitis (NEC) via oxidative stress modulation
WO2018136884A1 (en) Compositions and methods for treating obesity and inducing weight loss
Lee et al. Subjects with diarrhea-predominant IBS have increased rectal permeability responsive to tryptase
EP3900727A1 (en) Immunity mechanism and therapeutic drug for gastrointestinal diseases
Sun et al. The efficacy of anti‐proteolytic peptide R7I in intestinal inflammation, function, microbiota, and metabolites by multi‐omics analysis in murine bacterial enteritis
Zhang et al. Oral metformin inhibits choroidal neovascularization by modulating the gut-retina axis
US20210252107A1 (en) Novel therapy
Peng et al. Tryptophan metabolites relieve intestinal Candida albicans infection by altering the gut microbiota to reduce IL-22 release from group 3 innate lymphoid cells of the colon lamina propria
Dolislager et al. Campylobacter jejuni induces differentiation of human neutrophils to the CD16hi/CD62Llo subtype
Lu et al. Cytokines in cerebrospinal fluid of neurosyphilis patients: identification of Urokinase plasminogen activator using antibody microarrays
WO2025158027A1 (en) 4-hydroxyphenyllactic acid for use in treating and reducing the risk of ige-mediated diseases
Celik et al. Extracellular vesicle-associated TWEAK contributes to vascular inflammation and remodeling during acute cellular rejection
Yang et al. Protective effect of a combination of multiple strains of Lactobacillus acidophilus on collagen-induced arthritis
JP2019534247A (en) nNIF and nNIF-related peptides and related methods
Miller Targeting microbial pathogenic mechanisms as a novel therapeutic strategy in IBD
US20070128303A1 (en) Anti-inflammatory, cytoprotective factor derivable from a probiotic organism
Wishart et al. Neutralizing GM-CSF autoantibodies impair neutrophil antifungal effector function in a patient with aspergillosis
Watanabe et al. Enhanced resistance to Listeria infection in mice surviving sepsis: the role of lipid metabolism and myeloid cell reprogramming
Lioudakis THE INNATE IMMUNOMODULATORY EFFECTS OF AQUAMIN IN THE CONTEXT OF THE METABOLIC SYNDROME
WO2019165217A1 (en) Cd153 and/or cd30 in infection
Chen et al. MAPK signaling pathway induced LOX-1+ polymorphonuclear myeloid-derived suppressor cells in biliary atresia
WO2005077389A1 (en) An anti-inflammatory, cytoprotective factor derivable from a probiotic organism

Legal Events

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

Ref document number: 25702228

Country of ref document: EP

Kind code of ref document: A1