EP3471562A1 - Synthetic compositions comprising human milk oligosaccharides for use the prevention and treatment of disorders - Google Patents

Synthetic compositions comprising human milk oligosaccharides for use the prevention and treatment of disorders

Info

Publication number
EP3471562A1
EP3471562A1 EP17812794.0A EP17812794A EP3471562A1 EP 3471562 A1 EP3471562 A1 EP 3471562A1 EP 17812794 A EP17812794 A EP 17812794A EP 3471562 A1 EP3471562 A1 EP 3471562A1
Authority
EP
European Patent Office
Prior art keywords
hmos
human
cvd
fucosylated
use according
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.)
Withdrawn
Application number
EP17812794.0A
Other languages
German (de)
French (fr)
Other versions
EP3471562A4 (en
Inventor
Louise Kristine VIGSNÆS
Bruce Mcconnell
Emma ELISON
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.)
Glycom AS
Original Assignee
Glycom AS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US15/183,391 external-priority patent/US10835544B2/en
Priority claimed from US15/183,431 external-priority patent/US10987368B2/en
Priority claimed from US15/183,404 external-priority patent/US10881674B2/en
Application filed by Glycom AS filed Critical Glycom AS
Publication of EP3471562A1 publication Critical patent/EP3471562A1/en
Publication of EP3471562A4 publication Critical patent/EP3471562A4/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/125Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives containing carbohydrate syrups; containing sugars; containing sugar alcohols; containing starch hydrolysates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/702Oligosaccharides, i.e. having three to five saccharide radicals attached to each other by glycosidic linkages
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/20Reducing nutritive value; Dietetic products with reduced nutritive value
    • A23L33/21Addition of substantially indigestible substances, e.g. dietary fibres
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/40Complete food formulations for specific consumer groups or specific purposes, e.g. infant formula
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs

Definitions

  • This invention relates to a method and composition for regulating satiety and stabilising or reducing insulin resistance and risk of, preventing, or treating CVD and associated comorbidities in humans, in particular in overweight or obese human individuals.
  • Overweight and obesity is commonly associated with accumulated abdominal visceral fat and can be related to psycho-sociological behavioural disorders. It is often associated with the development of several chronic complications, such as high fasting glucose levels
  • hypoglycaemia hyperglycaemia
  • elevated triglyceride levels hypertriglyceridemia
  • low levels of high density lipoprotein dyslipidaemia
  • high blood pressure hypertension
  • Individuals who meet at least three of these criteria are clinically diagnosed as having metabolic syndrome, which increases the risk of developing metabolic diseases such as type 2 diabetes and cardiovascular diseases (CVD) (Boulange et al., Genome Medicine 8 , 42 (2016)) .
  • CVD cardiovascular diseases
  • Tissue resistance to insulin's actions has been postulated as the initial impairment underlying the onset of the metabolic comorbidities in these patients.
  • Type 2 diabetes is a metabolic disorder that is characterised by hyperglycaemia due to insulin resistance and relative lack of insulin and is a rapidly growing global epidemic.
  • IDF Diabetes Atlas International Diabetes Federation
  • WHO furthermore estimates that 90 percent of people around the world who suffer from diabetes suffer from type 2 diabetes. Long-term complications from high blood sugar can include heart disease, strokes, diabetic retinopathy, kidney failure, and poor blood flow in the limbs.
  • High low-density lipoprotein cholesterol (LDL-C) and triglyceride concentrations and low high- density lipoprotein cholesterol (HDL-C) in the blood is a precursor to hypertension, hyperlipidaemia, and causes the formation and build-up of atherosclerotic plaque in the arteries leading to higher risk of CVD.
  • Cardiovascular risk factors are not only observed in adults, but also obese children and young adults suffer from dyslipidaemia, hypertension, hyperinsulinemia or insulin resistance (Bridger, Paediatr. Child Health 1 4 , 177 (2009)) .
  • Cholesterol concentrations within the circulatory pool are products of input from gut absorption and endogenous synthesis relative to clearance through hepatic and extrahepatic tissue pathways. A disruption in any of these mechanisms can alter this balance, which is reflected in plasma cholesterol concentrations and subsequent CVD progression (Matthan et al. J. Am. Heart Assoc. 2 , e005066 (2013)) .
  • Gut microbiota is a specific entity within the body which has its own genome and whose gene pool is much more abundant than the one of its host. It has been estimated that the human intestine harbours 10 13 to 10 14 bacterial cells and the number of bacteria outnumbers the total number of cells in the body by a factor of 10 (Gill et al. , Science 31 2 , 1355 (2006)) . The physiologic functions attributed to gut microbiota have extended to extra-intestinal tissues, such as the liver, brain and adipose tissue, constructing connections with obesity and related disorders including type 2 diabetes and CVD.
  • gut microbiota has the potential to affect gut permeability and, consequently, give rise to metabolic endotoxemia and higher plasma lipopolysaccharide (LPS) .
  • gut peptides such as glucagon-like peptide 2 (GLP2) can play a key role in these processes (Tremaroli et al. Nature 489 , 242 (2012)) .
  • GLP2 which is secreted by intestine L cells, is a key regulator of intestinal permeability (Cani et al. Gut 58 , 1091 (2009)) .
  • Therapeutic regimes that target intestinal microbiota and intestinal barrier therefore show a broad prospect in treating metabolic diseases such as diabetes (Kootte et al. Diabetes, Obesity & Metabolism 1 4 , 112 (2012)) .
  • the gut microbiota does not only participate in whole-body metabolism by affecting energy balance (Turnbaugh et al. Nature 444 , 1027 (2006)) and glucose metabolism (Cani et al. Diabetes 57 , 1470 (2008)) but is also involved in development of the low-grade inflammation (Cani et al. Gut 58 , 1091 (2009)) associated with obesity and related metabolic disorders such as diabetes.
  • the association between inflammation and type 2 diabetes was described in the 1950s, when epidemiological studies showed a rise in acute-phase response proteins in serum of type 2 diabetic patients compared with controls (Fearnley et al. Lancet 274 , 1067 ( 1961)) .
  • GLP1 and PYY are of relevance to appetite and weight maintenance because they have action on the gastrointestinal tract as well as the direct regulation of appetite.
  • regulation of these hormones in humans can reduce food intake, appetite and hunger, and promote fullness and satiety with the ultimate result of promoting weight loss (Shah, Rev. Endocr. Metab. Disord. 1 5 , 181 (2014)) .
  • beneficial bacteria such as bifidobacteria have shown to ameliorate both metabolic and immunological dysfunctions related to obesity.
  • Bifidobacterium pseudocatenulatum has shown to reduce serum cholesterol, triglyceride and glucose levels and decrease insulin resistance and improve glucose tolerance in obese mice.
  • the species can reduce liver steatosis and the number of larger adipocytes in enterocytes of obese mice (Cano et al., Obesity, 21 , 2310 (2013)) .
  • bile salt hydrolase BSH
  • Some bifidobacteria have high BSH activity promoting deconjugation of bile acids in the gut to secondary amino acid conj ugates. When these secondary conj ugates are excreted, cholesterol is broken down to replace the processed bile salts. Overall, this process promotes the catabolism of cholesterol, leading to reduced serum levels (Ettinger et al., Gut Microbes 5 , 719 (2014)) .
  • SCFA short chain fatty acids
  • Acetate has shown to be negatively associated with visceral adipose tissue and insulin levels in obese individuals and propionate has shown to reduce lipogenesis and cholesterol synthesis inhibition (Verbeke et al. , Nutrition Research Reviews 28 , 42 (2015)) .
  • Selective stimulation of specific beneficial intestinal bacteria to promote their growth and metabolic activity could be a helpful approach in creating an intestinal environment that is able to regulate metabolic functions and energy balance.
  • a study has described that consuming inulin increased the gut microbiota fermentation, decreased appetite, improved postprandial glucose responses and higher concentrations of GLP1 and PYY after two weeks of prebiotic treatment (Cani et al. , Am. J. Clin. Nutr. 90 , 1236 (2009)) .
  • inulin may provoke side effects such as bloating, abdominal pain and increased flatulence.
  • Probiotic supplementation could be an approach, however, the addition of a small number of different probiotics to the intestine is unlikely to fully promote a beneficial intestinal microbiota composition with sufficient production of metabolites.
  • HMOs Human milk oligosaccharides
  • Human milk oligosaccharides are a heterogeneous mixture of soluble glycans found in human milk. They are the third most abundant solid component after lactose and lipids in human milk and are present in concentrations of 5-25 g/l (Gabrielli et al., Pediatrics 1 28 , 1520 (2011)) . HMOs are resistant to enzymatic hydrolysis in the small intestine and are thus largely undigested and unabsorbed (ten Bruggencate et al. , Nutrition Reviews 72 , 377 (2014)) . The majority of HMO that reaches the colon serves as a substrate to shape the gut ecosystem by selectively stimulating the growth of specific beneficial bacteria .
  • HMOs are believed to substantially modulate the infant gut microbiota and play a decisive role in the differences in the microbiota of formula-fed and breast-fed infants. These differences include the predominance of Bifidobacterium in the gut of breast-fed infants compared to a more diverse gut microbiota in formula-fed infants (Bezirtzoglou et al., Anaerobe 1 7 , 478 (2011) . This is viewed as beneficial for the infant because strains of Bifidobacterium species and their metabolites are believed to have a positive effect on human health (Chichlowski et al., J. Pediatr. Gastroenterol. Nutr. 55 , 321 (2012) ; Fukuda et al. , Nature 469 , 543 (2011)) .
  • sialylated and fucosylated HMOs has a positive effect on the growth of certain strains of bifidobacteria that are typically found in both infant and adult microbiota (WO 2013/154725) .
  • EP-A- 1332759 discloses that oral doses of 2'-FL, 3'-SL, 6'-SL, LNnT and sialic acid promote insulin secretion in type 2 diabetes-model mice.
  • EP-A-2143341 discloses that a mixture of GOS, sialylated oligosaccharides and N-acylated oligosaccharides reduces triglyceride concentration in liver in model mice.
  • EP-A-2332552 discloses that 3'-SL and 6'-SL reduce/prevent fat accumulation in the liver and other organs in high-fat diet mice and rats.
  • WO 2013/057061 discloses a composition for increasing insulin sensitivity and/or reducing insulin resistance that contains long chain polyunsaturated fatty acids, probiotics and a mixture of oligosaccharides containing at least one of lacto-N-neotetraose (LNnT) and lacto- N-tetraose (LNT), at least one N-acetylated oligosaccharide different from LNnT and LNT, at least one sialylated oligosaccharide and at least one neutral oligosaccharide.
  • This composition can also contain 2'-0-fucosyllactose (2'-FL) .
  • the composition is particularly adapted for use in infants who were born preterm and/or who experienced IUGR, and in pregnant women suffering from gestational diabetes. It is also stated that the composition can be given to children, adolescents and adults suffering from insulin resistance and/or type II diabetes. It is stated that the efficacy of the composition can be the result of the synergistic combination of immunity modulator effects triggered by the probiotics and the LC-PUFA through their stimulation with the specific oligosaccharide mixture.
  • WO 2013/036104 discloses a method for inter alia improving regulation of satiety in a human subject having an age of 0 to 36 months by feeding a nutritional composition comprising lipid globules.
  • WO 2014/ 187464 discloses a synthetic mixture of oligosaccharides comprising at least 6 oligosaccharides selected from fucosylated, sialylated, sulfated, GlcNAc- GalNAc- and mannose-containing oligosaccharides, for treating a microbiota of a human, to reduce or eliminate the activity and/or the proportion of a microbe in the microbiota that is associated with the development or maintenance of a cardiovascular disease.
  • a metabolic disorder for example obesity, obesity induced pre-diabetes and obesity induced type 2 diabetes
  • an obesity-related metabolic disorder for example obesity, obesity induced pre-diabetes and obesity induced type 2 diabetes; and/or
  • CVD or CVD-associated pathologic condition or disease in a human, preferably, in an overweight or obese human individual.
  • a first aspect of this invention relates to one or more human milk
  • an obesity-related metabolic disorder for example obesity, obesity induced pre-diabetes and obesity induced type 2 diabetes
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human preferably, in an overweight or obese human individual
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human preferably, in an overweight or obese human individual
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human preferably, in an overweight or obese human individual
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual.
  • the one or more HMOs is/are lacto-N-tetraose (LNT), lacto-N- neotetraose (LNnT), 2'-fucosyllactose (2'-FL), lacto-N-fucopentaose I (LNFP-I), 3- fucosyllactose (3-FL), difucosyllactose (DFL), 3'-sialyllactose (3'-SL) or 6'-sialyllactose (6'- SL), or a mixture thereof.
  • LNT lacto-N-tetraose
  • LNnT lacto-N- neotetraose
  • 2'-FL lacto-N-fucopentaose I
  • 3- fucosyllactose 3- fucosyllactose
  • DFL difucosyllactose
  • 3'-SL 3'-sialyllact
  • a second aspect of the invention relates to one or more human milk oligosaccharides (HMOs) selected from the group consisting of fucosylated HMOs and non-fucosylated HMOs, preferably a mixture of one or more fucosylated HMOs and one or more non-fucosylated neutral HMOs, for use in increasing the abundance of bifidobacteria in a human, preferably in an overweight or obese human having a propensity of, or diagnosed with a cardiovascular disease (CVD).
  • HMOs human milk oligosaccharides
  • a third aspect of the invention relates to a synthetic composition for use in
  • composition comprising an effective amount of one or more human milk oligosaccharides
  • an obesity-related metabolic disorder for example obesity, obesity induced pre-diabetes and obesity induced type 2 diabetes
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual, and/or
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human, preferably in an overweight or obese human individual
  • the synthetic composition comprising an effective amount of one or more human milk oligosaccharides (HMOs), for example those specified in the first aspect.
  • HMOs human milk oligosaccharides
  • the amount of a human milk oligosaccharide is effective to increase the abundance, particularly the relative abundance, of bifidobacteria in the gastrointestinal tract of the human.
  • the bifidobacteria increased is a member of the phylogenetic Bifidobacterium adolescentis group, for example, Bifidobacterium pseudocatenulatum and/or Bifidobacterium adolescentis, and, after about 14 days of treatment, are Bifidobacterium longum and/or Bifidobacterium bifidum .
  • a fourth aspect of the invention relates to a synthetic composition for use in increasing the abundance of bifidobacteria in a human, preferably, in an overweight or obese human having a propensity of, or diagnosed with a cardiovascular disease (CVD), the synthetic composition comprising one or more human milk oligosaccharides (HMOs) selected from the group consisting of fucosylated HMOs and non-fucosylated neutral HMOs, preferably a mixture of one or more fucosylated HMOs and one or more non-fucosylated neutral HMOs.
  • HMOs human milk oligosaccharides
  • a fifth aspect of this invention provides a method for
  • an obesity-related metabolic disorder for example obesity, obesity induced pre-diabetes and obesity induced type 2 diabetes
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual, and/or
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual
  • the method comprising administering, to the human, an effective amount of one or more human milk oligosaccharides, or a synthetic composition comprising an effective amount of one or more human milk oligosaccharides.
  • a sixth aspect of the invention relates to a method for increasing the a bundance of bifidobacteria in a human, preferably in an overweight or obese human having a propensity of, or diagnosed with a cardiovascular disease (CVD), the method comprising administering, to the human, one or more human milk oligosaccharides (HMOs) selected from the group consisting of fucosylated HMOs and core HMOs, preferably of a mixture of one or more fucosylated HMOs and one or more core HMOs, or a synthetic composition comprising one or more human milk oligosaccharides (HMOs) selected from the group consisting of fucosylated HMOs and core HMOs, preferably of a mixture of one or more fucosylated HMOs and one or more core HMOs.
  • HMOs human milk oligosaccharides
  • the one or more HMOs are administered to a human in need in two steps:
  • patient means a human individual who was diagnosed by a medical or health professional personal as having a disease, e.g . a metabolic disorder, and receiving or registered to receive medical treatment.
  • the patient can be a paediatric or adult patient.
  • An embodiment of the patent e.g. is an overweight or obese individual that is having a CVD or a CVD-associated pathological condition or disease.
  • Paediatric patient can be a human patient of 3-21 years old .
  • a “patient” can also be any other mammal.
  • non-infant human and “non-infant” all mean in the present context a human individual of at least 3 years old .
  • a human can be a child, a teenager, an adult or an elderly adult, preferably, the human is an individual of at least 3 years old that has an excess of body fat, more preferably, an individual whose excess body fat has accumulated to the extent that it may have a negative effect on health, i.e. an overweight or obese human individual.
  • Body fat percentage preferably means total mass of body fat divided by total mass of the body.
  • obese human or "obese human individual” means that a human individual that has a body mass index (BMI), a measurement obtained by dividing the individual's weight by the square of the individual's height, over 30 kg/m 2 , with the range 25-30 kg/m 2 defined as overweight.
  • BMI body mass index
  • Overweight and obesity for children and teens is defined as the following : overweight is defined as a BMI at or above the 85th percentile and below the 95th percentile for children and teens of the same age and sex. Obesity is defined as a BMI at or above the 95th percentile for children and teens of the same age and sex (see: Rolland-Cachera, Int. J. Pediatr. Obesity 6 , 325 (2011)) .
  • metabolic disorder also known as “metabolic syndrome” in the present context means an inherited or acquired genetic or physiological condition which is reflected by a change in metabolism of an individual, typically is a clustering of at least three of the five following medical conditions (giving a total of 16 possible combinations giving the syndrome) : abdominal (central) obesity (cf. TOFI)
  • HDL high-density lipoprotein
  • Metabolic syndrome is associated with the risk of
  • the invention relates to metabolic disorders, such as obesity, obesity induced pre-diabetes, obesity induced type 2 diabetes.
  • a metabolic disorder is insulin resistance or is associated with insulin resistance.
  • insulin resistance means a pathological condition in an individual who has a reduced insulin activity, e.g. due to the individual's cells fail to respond normally to the hormone insulin, or insulin is underproduced in the individual's body, or it has altered activity.
  • insulin resistance is in an overweight or obese individual.
  • Cardiovascular disease refers broadly to any disease of the heart and circulatory system (arteries and veins) . Cardiovascular disease generally refers to conditions that involve narrowed or blocked blood vessels that can lead to a heart attack, chest pain
  • angina or stroke.
  • Other heart conditions such as those that affect the heart muscle, valves or rhythm, also are also contemplated as forms of heart disease.
  • CVD include, but not limited to, coronary artery disease (blockage of blood vessels that serve the heart), acute coronary syndrome (symptoms such as pain, weakness, and tiredness caused by coronary artery disease), angina pectoris (pain resulting from coronary artery disease or other causes), myocardial infarction (heart attack, with damage to heart muscle caused by coronary artery disease), and left ventricular thrombus (blood clot in the left side of the heart that pumps blood into your body) .
  • coronary artery disease blockage of blood vessels that serve the heart
  • acute coronary syndrome symptoms such as pain, weakness, and tiredness caused by coronary artery disease
  • angina pectoris pain resulting from coronary artery disease or other causes
  • myocardial infarction heart attack, with damage to heart muscle caused by coronary artery disease
  • CVD may be accompanied with health complications (that are interchangeably referred herein as pathologic conditions) or associated diseases, which are also contemplated herein.
  • health complications that are interchangeably referred herein as pathologic conditions
  • CVD-associated diseases/pathologic conditions include heart failure (occurs when the heart cannot adequately pump blood throughout the body; this can cause shortness of breath, dizziness, confusion, and the build-up of fluid in the body, causing swelling), heart attack (occurs when the coronary arteries narrow so much that they cut off blood supply to the heart; the heart cells begin to die as they are deprived of oxygen and symptoms include shortness of breath and severe chest pain that may radiate to the back, jaw, or left arm), stroke (occurs due formation and lodging of blood clots in a blood vessel in the brain and cutting thus off blood flow; stroke symptoms include : numbness on one side of the body, confusion, trouble, speaking, loss of balance or coordination), pulmonary embolism (is similar to a stroke, but the blocked blood vessel is in the lungs instead of the
  • propensity in the present context means natural tendency of a human individual to develop later in life a medical condition, such as a disease, in particular a CVD or a CVD- associated pathological condition or disease.
  • preventing CVD and/or CVD associated pathological condition or disease in the present context means eliminating or minimising a chance of development of a CVD disease or a pathological condition or disease associated with an CVD. Both primary and secondary prevention are thus contemplated.
  • the primary prevention means preventing a CVD or associated disease or condition before it occurs, and the secondary prevention means preventing additional attacks of a CVD or development of associated condition or disease after the first attack has occurred .
  • enteral administration preferably means any conventional form for delivery of a composition to a human that causes the deposition of the composition in the gastrointestinal tract (including the stomach) .
  • Methods of enteral administration include feeding through a naso-gastric tube or jej unum tube, oral, sublingual and rectal.
  • oral administration preferably means delivery into the gastrointestinal tract through the oral cavity.
  • oral administration is a form of enteral administration.
  • the term "effective amount” preferably means an amount of a human milk oligosaccharide or an amount of a composition that provides a human milk oligosaccharide sufficient to render a desired treatment outcome in a patient.
  • An effective amount can be administered in one or more doses to the patient to achieve the desired treatment outcome.
  • the term "effective amount” may mean an amount of a single HMO, or a combination of different HMOs that is capable of increasing the abundance of bifidobacteria in the gastro- intestinal tract of a human individual of the invention, preferably, relative abundance of members of the Bifidobacterium adolescentis phylogenetic group in particular B. adolescentis and/or B. pseudocatenulatum .
  • the term "relative abundance of bifidobacteria” means the abundance of bifidobacteria relative to other genus in the microbiota of the gastro-intestinal tract.
  • human milk oligosaccharide or "HMO” preferably means a complex carbohydrate consisting of a small number, typically 3- 10, of monosaccharide units attached to each other by an interglycosidic linkage that can be found in human breast milk and that can be in acidic or neutral form. More than about 200 different HMO structures are known to exist in human breast milk (Urashima et al. : Milk Oligosaccharides, Nova Biomedical Books, New York, 2011) . HMOs can be core, fucosylated and sialylated oligosaccharides.
  • Core HMOs are non- fucosylated neutral (that is non-charged) HMOs and consist of Glu, Gal and GlcNAc (thus devoid of Fuc and sialic acid) .
  • core HMOs include lacto-N-tetraose (LNT), lacto- N-neotetraose (LNnT), lacto-N-neohexaose (LNnH) , lacto-N-hexaose (LNH) and p-lacto-N- neohexaose (pLNnH) .
  • Fucosyl HMOs are fucosylated lactoses or fucosylated core HMOs such as 2'-fucosyllactose (2'-FL), lacto-N-fucopentaose I (LNFP-I), lacto-N-difucohexaose I (LNDFH-I), 3-fucosyllactose (3-FL), difucosyllactose (DFL), lacto-N-fucopentaose III (LNFP- III), fucosyl-para-lacto-N-neohexaose (F-pLNnH), lacto-N-difucohexaose I (LNDFH-I), fucosyl-lacto-N-hexaose II (FLNH-II), lacto-N-fucopentaose V (LNFP-V), lacto-N- difucohexaose II (LNDFH-
  • Sialyl HMOs are sialylated lactoses or sialylated core HMOs such as 3',6-disialyllacto-N-tetraose (DSLNT), 6'-sialyllactose (6'-SL), 3'-sialyllactose (3'-SL), 6'-sialyllacto-N-neotetraose (LST c), 3'-sialyllacto-N-tetraose (LST a) and 6-sialyllacto-N-tetraose (LST b) .
  • DSLNT 3',6-disialyllacto-N-tetraose
  • 6'-SL 6'-sialyllactose
  • 3'-SL 6'-sialyllactose
  • LST c 6'-sialyllacto-N-neot
  • sialylated and fucosylated HMOs include disialyl-fucosyl-lacto-N-hexaose II (DSFLNH-II), fucosyl-sialyl-lacto-N-neohexaose I (FSLNnH-I), fucosyl-sialyl-lacto-N-hexaose I (FSLNH-I) and 3-fucosyl-3'-sialyllactose (FSL) .
  • DSFLNH-II disialyl-fucosyl-lacto-N-hexaose II
  • FSLNnH-I fucosyl-sialyl-lacto-N-neohexaose I
  • FSLNH-I fucosyl-sialyl-lacto-N-hexaose I
  • FSL 3-fucosyl-3'-sialyllactose
  • Microbiota preferably mean a community of living microorganisms that typically inhabits a bodily organ or part, particularly the gastro-intestinal organs of non-infant humans.
  • the most dominant members of the gastrointestinal microbiota include microorganisms of the phyla of Firmicutes, Bacteroidetes, Actinobacteria,
  • Proteobacteria Synergistetes, Verrucomicrobia, Fusobacteria, and Euryarchaeota; at genus level Bacteroides, Faecalibacterium, Bifidobacterium, Roseburia, Alistipes, Collinsella, Blautia, Coprococcus, Ruminococcus, Eubacterium and Dorea; at species level Bacteroides uniformis, Alistipes putredinis, Parabacteroides merdae, Ruminococcus bromii, Dorea longicatena, Bacteroides caccae, Bacteroides thetaiotaomicron, Eubacterium hallii, Ruminococcus torques, Faecalibacterium prausnitzii, Ruminococcus lactaris, Collinsella aerofaciens, Dorea
  • the gastrointestinal microbiota includes the mucosa-associated microbiota, which is located in or attached to the mucus layer covering the epithelium of the gastrointestinal tract, and luminal-associated microbiota, which is found in the lumen of the gastrointestinal tract.
  • bifidobacteria means a member of the Bifidobacterium genus commonly found in the human gastro-intestinal tract.
  • examples of bifidobacteria are Bifidobacterium longum, Bifidobacterium bifidum, and the members of the phylogenetic Bifidobacterium adolescentis group.
  • bifidobacteria preferably include members of the phylogenetic Bifidobacterium adolescentis group, for example Bifidobacterium pseudocatenulatum and/or Bifidobacterium adolescentis.
  • Bifidobacterium of the Bifidobacterium adolescentis phylogenetic group means a bacterium selected from a group consisting of Bifidobacterium adolescentis, Bifidobacterium angulatum , Bifidobacterium catenulatum, Bifidobacterium pseudocatenulatum ,
  • Bifidobacterium kashiwanohense, Bifidobacterium dentum and Bifidobacterium stercoris (Duranti et al. Appl. Environ. Microbiol. 79 , 336 (2013), Bottacini et al. Microbial Cell Fact. 1 3 : S4 (2014)) .
  • synthetic composition designates a composition which is artificially prepared and preferably means a composition containing at least one compound that is produced ex vivo chemically and/or biologically, e.g. by means of chemical reaction, enzymatic reaction or recombinantly.
  • a synthetic composition of the invention may be, but preferably is not, identical with a naturally occurring composition.
  • the synthetic composition of the invention typically comprises one or more compounds, advantageously HMOs, that are capable of preferentially increasing the abundance of bifidobacteria, in particular
  • Bifidobacterium of the following species Bifidobacterium longum, Bifidobacterium bifidum, and/or members of the phylogenetic Bifidobacterium adolescentis group.
  • the synthetic composition may comprise one or more compounds or components other than HMOs that may have an effect on bifidobacteria of a human subject microbiota in vivo, e.g . non-digestible oligosaccharides or prebiotics. Also in some
  • the synthetic compositions may comprise one or more nutritionally or pharmaceutically active components which do not affect adversely the efficacy of the above mentioned compounds.
  • a synthetic composition of the invention Some non-limiting embodiments of a synthetic composition of the invention are also described below.
  • the synthetic composition can take any suitable form.
  • the composition can be in the form of a nutritional composition which contains other macronutrients such as proteins, lipids or other carbohydrates.
  • the synthetic composition can also be a pharmaceutical composition.
  • intestinal permeability preferably means the permeability of the intestinal mucosa of a patient, permitting the absorption of vital nutrients from the gut lumen while presenting a barrier against the passage of pathogenic substances into the patient's body.
  • endotoxemia preferably means the presence of endotoxins, such as gut microbiota-derived lipopolysaccharides (LPS) in the blood of a patient.
  • endotoxins such as gut microbiota-derived lipopolysaccharides (LPS)
  • low-grade inflammation preferably means an immune system response of a patient characterized by altered levels of pro-inflammatory and anti-inflammatory cytokines as well as numerous other markers of immune system activity in response to an inj urious stimuli.
  • GI tissue index
  • AUC blood glucose response curve
  • the AUC of the test food is divided by the AUC of the standard (glucose, the standard, has a GI of 100) and multiplied by 100.
  • the average GI value is calculated from data collected in 10 human subjects.
  • initial period of treatment with an HMO or an HMO mixture is about 14 days from the start of the treatment; "additional period” of treatment means 1 or more days following the initial period of treatment.
  • the HMOs can be isolated or enriched by well-known processes from milk(s) secreted by mammals including, but not limited to human, bovine, ovine, porcine, or caprine species.
  • the HMOs can also be produced by well-known processes using microbial fermentation, enzymatic processes, chemical synthesis, or combinations of these technologies.
  • LNnT can be made as described in WO 2011/100980 and WO 2013/044928
  • LNT can be synthesized as described in WO 2012/155916 and WO 2013/044928
  • a mixture of LNT and LNnT ca n be made as described in WO 2013/091660
  • 2'-FL can be made as described in WO 2010/ 115934 and WO 2010/115935
  • 3-FL can be made as described in WO
  • sialylated oligosaccharides can be made as described in WO 2012/113404 and mixtures of human milk oligosaccharides can be made as described in WO 2012/113405.
  • sialylated oligosaccharides can be made as described in WO
  • fucosylated oligosaccharides can be made as described in WO 2012/127410, and advantageously diversified blends of human milk oligosaccharides can be made as described in WO 2012/156897 and WO 2012/156898.
  • WO 01/04341 and WO 2007/ 101862 describe how to make core human milk oligosaccharides optionally substituted by fucose or sialic acid using genetically modified £ coli.
  • Embodiments of the invention regulating satiety and/or reducing propensity to obesity
  • HMOs human milk oligosaccharides
  • HMOs can, by enteral ingestion, increase the production of GLP1, possibly through modulation of the intestinal microbiota in a human, in particular a non-infant human.
  • GLP1 possibly through modulation of the intestinal microbiota in a human, in particular a non-infant human.
  • a more beneficial intestinal microbial community can be shaped and maintained, and the regulation of satiety can be improved, leading to lower food intake. This can result in reduced propensity to obesity.
  • this invention in one aspect, relates to one or more human milk
  • oligosaccharides for use in a synthetic composition comprising an effective amount of one or more human milk oligosaccharides for use in, or a method for
  • a mixture of 2'-FL and LNnT may contain the amount of 2'- FU LNnT form about 1.5 : 1 to about 4: 1 by weight.
  • the HMO may be a single HMO or a mixture of any HMOs suitable for the purpose of the invention.
  • the HMO is a fucosylated or a non-fucosylated neutral HMO.
  • the invention relates to a mixture of HMOs, the mixture comprising at least a first HMO and at least a second HMO, wherein the first HMO is a fucosylated neutral HMO and the second HMO is a non-fucosylated neutral HMO.
  • the mixture of HMOs may contain a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP- III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH-III, FLNH-I, FLNH-II, FLNnH, FpLNH-I and F-pLNnH II, and a non-fucosylated HMO selected from the list consisting of LNT, LNnT, LNH, LNnH, pLNH and pLNnH .
  • a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP- III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH-III, FLNH-I, FLNH-II, FLNnH, Fp
  • the mixture of HMOs contains a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL and DFL, and a non-fucosylated HMO selected from the list consisting of LNT and LNnT; advantageously the mixture comprises 2'-FL and LNnT and/or LNT.
  • the mixture of HMOs essentially consists of two neutral HMOs, e.g.
  • a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP-III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH-III, FLNH-I, FLNH-II, FLNnH, FpLNH-I and F- pLNnH II, and a non-fucosylated HMO selected from the list consisting of LNT, LNnT, LNH, LNnH, pLNH and pLNnH .
  • the mixture essentially consists of a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL and DFL, and a non-fucosylated HMO selected from the list consisting of LNT and LNnT; in one preferred embodiment the mixture essentially consists of 2'-FL and LNnT, in another preferred embodiment the mixture essentially consists of 2'-FL and LNT.
  • a mixture of 2'-FL and LNnT may contain the amount of 2'- FL: LNnT form about 1.5: 1 to about 4: 1 by weight.
  • HMO may comprise a single HMO or a mixture of any HMOs suitable for the purpose of the invention as disclosed above.
  • the synthetic composition can take any suitable form.
  • the composition can be in the form of a nutritional composition which contains other macronutrients such as proteins, lipids or other carbohydrates (see below).
  • the synthetic composition can also be a
  • compositions comprise an effective amount one or more HMOs described above.
  • the synthetic composition may be a pharmaceutical composition.
  • the synthetic composition comprises an effective amount one or more HMOs described above.
  • the pharmaceutical composition can contain a pharmaceutically acceptable carrier, e.g.
  • the pharmaceutical composition can also contain other materials that do not produce an adverse, allergic or otherwise unwanted reaction when administered to humans.
  • the carriers and other materials can include solvents, dispersants, coatings, absorption promoting agents, controlled release agents, and one or more inert excipients, such as starches, polyols, granulating agents, microcrystalline cellulose, diluents, lubricants, binders, and disintegrating agents.
  • tablet dosages of the a nti- infective compositions can be coated by standard aqueous or non-aqueous techniques.
  • compositions can be administered orally, e.g. as a tablet, capsule, or pellet containing a predetermined amount, or as a powder or granules containing a predetermined concentration or a gel, paste, solution, suspension, emulsion, syrup, bolus, electuary, or slurry, in an aqueous or non-aqueous liquid, containing a predetermined concentration.
  • Orally administered compositions can include binders, lubricants, inert diluents, flavouring agents, and humectants.
  • Orally administered compositions such as tablets can optionally be coated and can be formulated so as to provide sustained, delayed or controlled release of the mixture therein.
  • compositions can also be administered by rectal suppository, aerosol tube, naso-gastric tube or direct infusion into the GI tract or stomach.
  • the pharmaceutical compositions can also include therapeutic agents such as antiviral agents, antibiotics, probiotics, analgesics, and anti-inflammatory agents.
  • the proper dosage of these compositions for a human can be determined in a conventional manner, based upon factors such as severity of the lactose intolerance, immune status, body weight and age.
  • a synthetic composition comprises an effective amount of a mixture of 2'-FL and LNnT, preferably the mass ratio between 2'-FL and LNnT in the composition is in the range from 5: 1 to 1 : 1.
  • the invention relates to the following methods:
  • a method for reducing propensity to obesity in a human having a metabolic disorder comprising administering to the human an effective amount of one or more human milk oligosaccharides.
  • the amount of human milk oligosaccharide(s) required to be administered to the person will vary depending upon factors such as the risk and condition severity, the age of the person, the form of the composition, and other medications being administered to the person.
  • the required amount can be readily set by a medical practitioner and would generally be in the range from about 10 mg to about 20 g per day, in certain embodiments from about 10 mg to about 15 g per day, from about 100 mg to about 10 g per day, in certain embodiments from about 500 mg to about 10 g per day, in certain embodiments from about 1 g to about 7.5 g per day.
  • an appropriate dose can be determined based on several factors, including, for example, the body weight and/or condition of the patient being treated, the severity of the condition, being treated, other ailments and/or diseases of the person, the incidence and/or severity of side effects and the manner of administration. Appropriate dose ranges can be determined by methods known to those skilled in the art.
  • the dosing can be higher (for example 200 mg to 20 g per day, preferably 500 mg to 15 g per day, more preferably 1 g to 10 g per day, in certain embodiments 2.5 g to 7.5 g per day).
  • the dosing can be reduced (for example, 10 mg to 10 g per day, preferably 100 mg to 7.5 g per day, more preferably 500 mg to 5 g per day, in certain embodiments 1 g to 2.5 g per day).
  • one or more HMOs or a composition comprising or essentially consisting thereof is administered to a human in need enteral, e.g. orally.
  • a synthetic composition of this invention can be co-administered to a patient who is also receiving a standard-of-care medication for obesity or diabetes.
  • Embodiments of the invention stabilising or reducing insulin resistance
  • human milk oligosaccharides advantageously 2'-FL, LNT and LNnT, not only modulate inflammation and microbiota in the GI tract, but also stabilise or reduce insulin resistance. Further, the abundance of members of the
  • Bifidobacterium adolescentis phylogenetic group is increased, in particular B. adolescentis and/or B. pseudocatenulatum .
  • B. adolescentis and/or B. pseudocatenulatum are increased, in particular B. adolescentis and/or B. pseudocatenulatum .
  • a combination of 2'-FL and LNnT preferentially increases the abundance of B. pseudocatenulatum . This can result in lower chronic inflammation, improved insulin sensitivity and reduced insulin resistance.
  • Obese and pre- diabetic patients can be stabilised and the progression to diabetes slowed, stopped or reversed .
  • Diabetic patients can be stabilised or at least the progression to diabetes with complications slowed .
  • this invention in another embodiments, relates to one or more human milk oligosaccharides (HMOs) for use in, or a synthetic composition comprising an effective amount of one or more human milk oligosaccharides for use in, or a method for stabilising or reducing insulin resistance in a human individual having an obesity-related metabolic disorder, for example obesity, obesity induced pre-diabetes and obesity induced type 2 diabetes.
  • HMOs human milk oligosaccharides
  • One embodiment of the invention relates to an HMO for use in stabilising or reducing insulin resistance in a human individual having an obesity-related metabolic disorder.
  • Another embodiment of the invention relates to a synthetic composition comprising an HMO for use in stabilising or reducing insulin resistance in a human individual having an obesity-related metabolic disorder.
  • Still another embodiment of the invention relates to a method for stabilising or reducing insulin resistance in a human individual having an obesity-related metabolic disorder, the method comprising enterally administering to the patient an effective amount of one or more human milk oligosaccharides.
  • the HMO in any of the above embodiments concerning stabilising or reducing insulin resistance in a human may be a single HMO or a mixture of any HMOs suitable for the purpose of the invention.
  • the HMO is a fucosylated or a non-fucosylated neutral HMO.
  • this particular aspect of the invention relates to a mixture of HMOs, the mixture comprising at least a first HMO and at least a second HMO, wherein the first HMO is a fucosylated neutral HMO and the second HMO is a non-fucosylated neutral HMO.
  • the mixture of HMOs may contain a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP-III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH- III, FLNH-I, FLNH-II, FLNnH, FpLNH-I and F-pLNnH II, and a non-fucosylated HMO selected from the list consisting of LNT, LNnT, LNH, LNnH, pLNH and pLNnH .
  • a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP-III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH- III, FLNH-I, FLNH-II, FLNnH, Fp
  • the mixture of HMOs contains a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL and DFL, and a non-fucosylated HMO selected from the list consisting of LNT and LNnT; advantageously the mixture comprises 2'-FL and LNnT and/or LNT.
  • the mixture of HMOs essentially consists of two neutral HMOs, e.g.
  • a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP-III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH-III, FLNH-I, FLNH-II, FLNnH, FpLNH-I and F-pLNnH II, and a non- fucosylated HMO selected from the list consisting of LNT, LNnT, LNH, LNnH, pLNH and pLNnH.
  • the mixture essentially consists of a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL and DFL, and a non-fucosylated HMO selected from the list consisting of LNT and LNnT; in one preferred embodiment the mixture essentially consists of 2'-FL and LNnT, in another preferred embodiment the mixture essentially consists of 2'-FL and LNT.
  • the invention relates in different embodiments to single HMOs as substantially pure single compounds, i.e. an HMO which grade of purity satisfies the demand of a medical or food authority for marketing, or mixtures of substantially pure HMOs, or artificial compositions comprising one or more HMOs.
  • the human milk oligosaccharides include one or more fucosylated HMOs, such as 2'-FL, and one or more core HMOs, such as LNT and LNnT. More preferably the composition comprises a mix of 2'-FL and LNnT; for example in a mass ratio of 5: 1 to 1 : 1; more preferably 4: 1 to 2: 1.
  • the human individual can be an obese paediatric or adult patient, preferably a prepubescent child.
  • the HMOs may be administered to the patient as a daily dose of about 1 g to about 15 g such as from about 3 g to about 10 g.
  • the patient can be administered a higher amount, preferably 5 g to 10 g per day, of the HMOs for an initial treatment period, followed by a lower amount, preferably 1 g to 5 g per day, for a
  • the initial treatment period can be 1 to 8 weeks.
  • the maintenance period is at least 1 month.
  • the synthetic composition can take any suitable form.
  • the composition can be in the form of a nutritional composition which contains other macronutrients such as proteins, lipids or other carbohydrates (see below).
  • the synthetic composition can also be a
  • compositions comprise an effective amount one or more HMOs described above.
  • the synthetic composition may be a pharmaceutical composition.
  • the synthetic composition comprises an effective amount one or more HMOs described above.
  • the pharmaceutical composition can contain a pharmaceutically acceptable carrier, e.g.
  • the pharmaceutical composition can also contain other materials that do not produce an adverse, allergic or otherwise unwanted reaction when administered to humans.
  • the carriers and other materials can include solvents, dispersants, coatings, absorption promoting agents, controlled release agents, and one or more inert excipients, such as starches, polyols, granulating agents, microcrystalline cellulose, diluents, lubricants, binders, and disintegrating agents.
  • tablet dosages of the a nti- infective compositions can be coated by standard aqueous or non-aqueous techniques.
  • compositions can be administered orally, e.g. as a tablet, capsule, or pellet containing a predetermined amount, or as a powder or granules containing a predetermined concentration or a gel, paste, solution, suspension, emulsion, syrup, bolus, electuary, or slurry, in an aqueous or non-aqueous liquid, containing a predetermined concentration.
  • Orally administered compositions can include binders, lubricants, inert diluents, flavouring agents, and humectants.
  • Orally administered compositions such as tablets can optionally be coated and can be formulated so as to provide sustained, delayed or controlled release of the mixture therein.
  • compositions can also be administered by rectal suppository, aerosol tube, naso-gastric tube or direct infusion into the GI tract or stomach.
  • the pharmaceutical compositions can also include therapeutic agents such as antiviral agents, antibiotics, probiotics, analgesics, and anti-inflammatory agents.
  • therapeutic agents such as antiviral agents, antibiotics, probiotics, analgesics, and anti-inflammatory agents.
  • the proper dosage of these compositions for a human can be determined in a conventional manner, based upon factors such as severity of the lactose intolerance, immune status, body weight and age.
  • the amount of human milk oligosaccharide(s) required to be administered to the person will vary depending upon factors such as the risk and condition severity, the age of the person, the form of the composition, and other medications being administered to the person.
  • the required amount can be readily set by a medical practitioner and would generally be in the range from about 200 mg to about 20 g per day, in certain embodiments from about 1 g to about 15 g per day, from about 3 g to about 10 g per day, in certain embodiments from about 3 g to about 7.5 g per day.
  • An appropriate dose can be determined based on several factors, including, for example, body weight and/or condition, the severity of the condition, being treated or prevented, other ailments and/or diseases, the incidence and/or severity of side effects and the manner of administration. Appropriate dose ranges may be determined by methods known to those skilled in the art.
  • the dosing can be higher or lower depending upon the need to boost bifidobacteria abundance or initial tolerance to HMOs.
  • the dosing can be set for chronic long term use.
  • the dosing can be 500 mg to 20 g per day, preferably 1 g to 15 g per day, more preferably 3 g to 10 g per day.
  • the dosing can be reduced to 200 mg to 10 g per day, preferably 500 mg to 7.5 g per day, more preferably 1 g to 5 g per day.
  • a synthetic composition of this invention can be co-administered to a patient who is also receiving a standard-of-care medication for obesity or diabetes.
  • HMO/HMOs or synthetic composition(s) comprising said HMO/HMOs disclosed herein is(are) preferably administered to a patient in need enterally, e.g . orally.
  • Embodiments of the invention reducing the risk of, preventing or treating CVD and associated co-morbidities in overweight or obese humans
  • HMOs oligosaccharides
  • HMOs can, by oral or enteral ingestion, increase the production of SCFA and activity of BSH, possibly through modulation of the intestinal microbiota in human.
  • a more beneficial intestinal microbial community can be shaped and maintained, which contributes to attenuation of hypercholesterolemia and hypertension . This can result in reduced risk of, prevention of and/or treatment of, CVD and associated co-morbidities.
  • this invention in other embodiments, relates to a human milk oligosaccharide or a mixture of two to five human milk oligosaccharides for use in, or a synthetic composition comprising an effective amount of a human milk oligosaccharide or an effective amount of a mixture of two to five human milk oligosaccharides for use in, or a method for
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual, and/or
  • CVD cardiovascular disease
  • CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual.
  • one or more HMOs selected from the group consisting of fucosylated HMOs and core HMOs, preferably of a mixture of one or more fucosylated HMOs and one or more core HMOs, for use in, or a synthetic composition comprising an effective amount of one or more HMOs selected from the group consisting of fucosylated HMOs and core HMOs, preferably of a mixture of one or more fucosylated HMOs and one or more core HMOs, for use in, or a method for increasing the abundance of bifidobacteria in a human, preferably, in an overweight or obese human having a propensity of, or diagnosed with a cardiovascular disease (CVD).
  • the HMOs suitable for the purpose according to this aspect of the invention are disclosed below.
  • the invention relates in different embodiments to single HMOs as substantially pure single compounds, i.e. an HMO which grade of purity satisfies the demand of a medical or food authority for marketing, or mixtures of 2 to 5 such substantially pure HMOs, or artificial compositions comprising one to five HMOs.
  • HMOs and compositions comprising thereof are described below.
  • CVD cardiovascular disease
  • CVD cardiovascular disease
  • CVD cardiovascular disease
  • the HMOs may be a single HMO or a mixture of two to five of any HMOs suitable for the purpose of the invention.
  • the HMO is a fucosylated or a non-fucosylated neutral HMO.
  • the invention relates to a mixture of HMOs, the mixture comprising at least a first HMO and at least a second HMO, wherein the first HMO is a fucosylated neutral HMO and the second HMO is a non-fucosylated neutral HMO.
  • the mixture may comprise further a third, a forth and a fifth HMO.
  • the mixture of HMOs may contain a fucosylated HMO selected from the list consisting of 2'- FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP-III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH-III, FLNH-I, FLNH-II, FLNnH, FpLNH-I and F-pLNnH II, and a non-fucosylated HMO selected from the list consisting of LNT, LNnT, LNH, LNnH, pLNH and pLNnH.
  • a fucosylated HMO selected from the list consisting of 2'- FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP-III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH-III, FLNH-I, FLNH-II, FLNnH, FpL
  • the mixture of HMOs contains a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL and DFL, and a non-fucosylated HMO selected from the list consisting of LNT and LNnT; advantageously the mixture comprises 2'-FL and LNnT and/or LNT.
  • the mixture of HMOs essentially consists of two neutral HMOs, e.g.
  • a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP-III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH- III, FLNH-I, FLNH-II, FLNnH, FpLNH-I and F-pLNnH II, and a non-fucosylated HMO selected from the list consisting of LNT, LNnT, LNH, LNnH, pLNH and pLNnH.
  • the mixture essentially consists of a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL and DFL, and a non-fucosylated HMO selected from the list consisting of LNT and LNnT; in one preferred embodiment the mixture essentially consists of 2'-FL and LNnT, in another preferred embodiment the mixture essentially consists of 2'-FL and LNT.
  • a mixture of 2'-FL and LNnT may contain the amount of 2'-FL: LNnT form about 1.5 : 1 to about 4: 1 by weight.
  • the invention relates to a synthetic composition for use in
  • CVD cardiovascular disease
  • CVD cardiovascular disease
  • CVD cardiovascular disease
  • HMO which may comprise a single HMO or a mixture of two to five of any HMOs suitable for the purpose of the invention as disclosed above.
  • the synthetic composition can take any suitable form.
  • the composition can be in the form of a nutritional composition which contains other macronutrients such as proteins, lipids or other carbohydrates (see below) .
  • the synthetic composition can also be a pharmaceutical composition .
  • a pharmaceutical composition contains an effective amount of HMO or an effective amount of mixture of two to five HMOs, wherein the HMOs are selected from any of described above.
  • the term "effective amount" in the present content means an amount of a single HMO, or a combination of different HMOs that is capable of increasing the abundance of bifidobacteria in the gastrointestinal tract of a human individual of the invention, preferably, relative abundance of members of the Bifidobacterium adolescentis phylogenetic group in particular B. adolescentis and/or B. pseudocatenulatum .
  • the pharmaceutical composition can further contain a pharmaceutically acceptable carrier, e.g. phosphate buffered saline solution, mixtures of ethanol in water, water and emulsions such as an oil/water or water/oil emulsion, as well as various wetting agents or excipients.
  • a pharmaceutically acceptable carrier e.g. phosphate buffered saline solution, mixtures of ethanol in water, water and emulsions such as an oil/water or water/oil emulsion, as well as various wetting agents or excipients.
  • the pharmaceutical composition can also contain other materials that do not produce an adverse, allergic or otherwise unwanted reaction when administered to humans.
  • the carriers and other materials can include solvents, dispersants, coatings, absorption promoting agents, controlled release agents, and one or more inert excipients, such as starches, polyols, granulating agents, microcrystalline cellulose, diluents, lubricants, binders, and disintegrating agents.
  • compositions can be administered orally, e.g. as a tablet, capsule, or pellet containing a predetermined amount, or as a powder or granules containing a predetermined concentration or a gel, paste, solution, suspension, emulsion, syrup, bolus, electuary, or slurry, in an aqueous or non-aqueous liquid, containing a predetermined concentration.
  • Orally administered compositions can include binders, lubricants, inert diluents, flavouring agents, and humectants.
  • Orally administered compositions such as tablets can optionally be coated and can be formulated so as to provide sustained, delayed or controlled release of the mixture therein.
  • compositions can also be administered by rectal suppository, aerosol tube, naso-gastric tube or direct infusion into the GI tract or stomach.
  • compositions can also include therapeutic agents most commonly prescribed for heart disease such as:
  • ACE inhibitors are a type of medication that dilates (widens) arteries to lower blood pressure and make it easier for the heart to pump blood. They also block some of the harmful actions of the endocrine system that may occur with heart failure;
  • Eplerenone (Inspra) and spironolactone (Aldoctone) and eplerenone are potassium-sparing diuretics. They can be prescribed to reduce the swelling and water build-up caused by heart failure. Diuretics cause the kidneys to send unneeded water and salt from the tissues and blood into the urine;
  • ARBs Angiotensin II Receptor Blocker
  • ARBs are used to decrease blood pressure in people with heart failure. ARBs decrease certain chemicals that narrow the blood vessels so blood can flow more easily through your body. They also decrease certain chemicals that cause salt and fluid build-up in the body;
  • Beta-Blockers block the effects of adrenaline (epinephrine) and thereby improve the heart's ability to perform. They also decrease the production of harmful substances produced by the body in response to heart failure. They cause the heart to beat more slowly and with less force, lowering blood pressure;
  • Calcium channel blockers are prescribed to treat angina (chest pain) and high blood pressure. Calcium channel blockers affect the movement of calcium in the cells of the heart and blood vessels. As a result, the drugs relax blood vessels and increase the supply of blood and oxygen to the heart, while reducing its workload;
  • Cholesterol -Lowering Drugs Cholesterol helps your body build new cells, insulate nerves, and produce hormones. But inflammation may lead to cholesterol build-up in the walls of arteries, increasing the risk of heart attack and stroke;
  • Digoxin helps an injured or weakened heart to work more efficiently and to send blood through the body. It strengthens the force of the heart muscle's contractions and may improve blood circulation; Diuretics: Diuretics, commonly known as "water pills," cause the kidneys to get rid of unneeded water and salt from the tissues and bloodstream into the urine. Getting rid of excess fluid makes it easier for your heart to pump. Diuretics are used to treat high blood pressure and reduce the swelling and water build-up caused by various medical problems, including heart failure;
  • Inotropic therapy is used to stimulate an injured or weakened heart to pump harder to send blood through the body. It helps the force of the heart muscle's contractions and relaxes constricted blood vessels so blood can flow more smoothly. Inotropic therapy may also speed up the heart's rhythm;
  • Potassium or Magnesium are minerals that can be lost because of increased urination when taking diuretics. Low levels in the body can be associated with abnormal heart rhythms. Some patients take them as supplements as directed by their doctor.
  • Vasodilators are used to treat heart failure and control high blood pressure by relaxing the blood vessels so blood can flow more easily through the body. Vasodilators are prescribed for patients who cannot take ACE inhibitors.
  • Warfarin is an anticoagulant medication. "Anti” means “against,” and “coagulant” means “causing blood clotting.” Therefore, warfarin helps prevent clots from forming in the blood.
  • the pharmaceutical composition may also contain other compounds such as antibiotics, probiotics, analgesics, and anti-inflammatory agents.
  • compositions for a human can be determined in a conventional manner, based upon factors such as severity of conditions of the human individual, e.g. the individual's blood pressure, immune status, body weight, age, etc.
  • the invention relates to a method for
  • CVD cardiovascular disease
  • pathologic condition or disease in a human, preferably, wherein said human is overweight or obese;
  • CVD cardiovascular disease
  • CVD-associated pathologic condition or disease in a human, preferably wherein said human is overweight or obese;
  • CVD cardiovascular disease
  • CVD-associated pathologic condition or disease in a human, preferably, wherein said human is overweight or obese;
  • HMO human milk oligosaccharide
  • the invention contemplates both prophylactic and therapeutic methods of treatment depending on different embodiments.
  • therapeutic method means a method co mprising treatment of disease or medical disorder by remedial agents and/or, e.g .
  • prophylactic method means a method comprising a measure taken to fend off a disease or another unwanted consequence of the disease, e.g . administering an HMO or a composition of the invention to a human of the invention to reduce the propensity of or prevent development of CVD or the associated pathological condition or disease in the human.
  • an HMO of the invention is administered to a human in need enteral, e.g. orally.
  • invention relates to a method increasing the abundance of a bacterium of the B. adolescentis phylogenetic group, especially Bifidobacterium adolescentis and/or B.
  • one or more HMOs may be any of the methods.
  • one or more HMOs preferably, one to five HMOs, may be any of the methods.
  • substantially pure compounds i.e. neat
  • diluted e.g . in form of a solution, power or syrup, or in the form of a synthetic composition, nutritional or
  • compositions as any of the described above, in one or more unit dosage forms, preferably in a single unit dosage form.
  • the HMOs are, or the synthetic, nutritional or pharmaceutical, composition contains, 2'-FL and LNnT, preferably the 2'-FL: LNnT ratio is about 1.5 : 1 to about 4: 1 by weight.
  • the dosage of one or more fucosylated HMOs and one or more core HMOs per administration may vary from about 2 g to about 10 g, preferably from about 3.5 g to about 7.5 g.
  • the HMOs are administered in a single dosage unit containing from about 2 g to about 10 g, preferably from about 3.5 g to about 7.5 g of one of more fucosylated HMOs and one or more core HMOs.
  • the patient may also additionally receive a dose of one or more species of probiotic bacteria, e.g . bifidobacteria.
  • the patient is administered a daily dose of at least 2 g of the mixture of one or more fucosylated HMOs and one or more core HMOs for at least 14 days, preferably, for more than 14 days.
  • a nutritional composition can contain sources of protein, lipids and/or digestible
  • composition can be designed to be the sole source of nutrition or a nutritional supplement.
  • Suitable protein sources include intact, hydrolysed, and partially hydrolysed protein, which can be derived from any suitable source such as milk (e.g., casein, whey), animal (e.g. meat, fish), cereal (e.g. rice, corn), and vegetable (e.g. soy, potato, pea), insect (e.g. locust) and combinations of these sources.
  • milk e.g., casein, whey
  • animal e.g. meat, fish
  • cereal e.g. rice, corn
  • vegetable e.g. soy, potato, pea
  • insect e.g. locust
  • Examples of the source of protein include whey protein concentrates, whey protein isolates, whey protein hydrolysates, acid caseins, sodium casemates, calcium casemates, potassium casemates, casein hydrolysates, milk protein concentrates, milk protein isolates, milk protein hydrolysates, non-fat dry milk, condensed skim milk, soy protein concentrates, soy protein isolates, soy protein hydrolysates, pea protein concentrates, pea protein isolates, pea protein hydrolysates, collagen proteins, and combinations of these sources.
  • the amount of protein is preferably sufficient to provide about 5 to about 30 % of the energy of the nutritional composition; for example about 10 % to about 25 % of the energy. Within these ranges, the amount of protein can vary depending upon the nutritional needs of the intended individual.
  • the nutritional compositions can also include free amino acids such as tryptophan, glutamine, tyrosine, methionine, cysteine, taurine, arginine, carnitine, threonine, serine and proline and combinations of these amino acids.
  • Threonine, serine and proline are important amino acids for the production of mucin which aids gut barrier function.
  • Any suitable source of other carbohydrates can be used. Examples include maltodextrin, hydrolysed or modified starch or corn starch, glucose polymers, corn syrup, corn syrup solids, rice-derived carbohydrates, sucrose, glucose, fructose, lactose, high fructose corn syrup, honey, sugar alcohols (e.g., maltitol, erythritol, sorbitol, etc.), isomaltulose, sucromalt, pullulan, potato starch, slowly-digested carbohydrates, dietary fibres such as oat fibre, soy fibre, gum arabic, sodium carboxymethylcellulose, methylcellulose, guar gum, gellan gum, locust bean gum, konjac flour, hydroxypropyl methylcellulose, tragacanth gum, karaya gum, gum acacia, chitosan, arabinogalactans, glucomannan, xanthan gum, alginate, pectin, low and high methoxy pectin, cereal
  • the carbohydrate source includes low glycemic index carbohydrates having a GI score of 55 or below.
  • low glycemic index carbohydrates include sucromalt,
  • Fibersol(TM) inulin
  • maltodextrins having a dextrose equivalence (DE) of less than
  • rice syrup having a dextrose equivalence of less than 15
  • fructooligosaccharides resistant starches, starches, fruit sourced fibres, vegetable sourced fibres, whole grains, beta-glucans, soy fibres, oat fibres, locust bean gum, konjac flour, hydroxy propyl methylcellulose, gum acacia, chitosan, arabinogalactans, xanthan gum, alginate, low and high methoxy pectin, carrageenan, psyllium, isomaltulose, glycerine and sugar alcohols.
  • DE dextrose equivalence
  • rice syrup having a dextrose equivalence of less than 15
  • fructooligosaccharides resistant starches, starches, fruit sourced fibres, vegetable sourced fibres, whole grains
  • the nutritional compositions can include carbohydrates in an amount sufficient to provide about 30 to about 70 % of the energy of the composition, for example about 35 to about 65 % of the energy. Within these parameters, the amount of carbohydrate can vary widely.
  • Suitable lipid sources include coconut oil, fractionated coconut oil, soy oil, corn oil, olive oil, safflower oil, high oleic safflower oil, medium chain triglycerides, sunflower oil, high oleic sunflower oil, palm and palm kernel oils, palm olein, canola oil, marine oils, cottonseed oils and combinations of these oils.
  • Fractionated coconut oils are a suitable source of medium chain triglycerides.
  • the lipids can contain polyunsaturated fatty acids such as n-3 LC-PUFA.
  • the n-3 LC-PUFA can be a C20 or a C22 n-3 fatty acid .
  • n-3 LC-PUFA is docosahexanoic acid (DHA, C22 : 6, n-3) .
  • the source of LC-PUFA can be, for example, egg lipids, fungal oil, low EPA fish oil or algal oil.
  • the nutritional compositions can include lipids in an amount sufficient to provide about 10 to about 50 % of energy of the nutritional composition, for example about 15 to about 40 % of the energy.
  • the nutritional composition preferably also includes vitamins and minerals. If the nutritional composition is intended to be a sole source of nutrition, it preferably includes a complete vitamin and mineral profile.
  • vitamins include vitamins A, B-complex (such as Bl, B2, B6 and B12), C, D, E and K, niacin and acid vitamins such as pantothenic acid, folic acid and biotin.
  • minerals include calcium, iron, zinc, magnesium, iodine, copper, phosphorus, manganese, potassium, chromium, molybdenum, selenium, nickel, tin, silicon, vanadium and boron .
  • the nutritional composition can also include a carotenoid such as lutein, lycopene, zeaxanthin, and beta -carotene.
  • a carotenoid such as lutein, lycopene, zeaxanthin, and beta -carotene.
  • the total amount of carotenoid included can vary from about 0.001 pg/ml to about 10 pg/ml.
  • Lutein can be included in an amount of from about 0.001 pg/ml to about 10 pg/ml, preferably from about 0.044 pg/ml to about 5 pg/ml of lutein.
  • Lycopene can be included in an amount from about 0.001 pg/ml to about 10 pg/ml, preferably about 0.0185 pg/ml to about 5 pg/ml of lycopene.
  • Beta-carotene can comprise from about 0.001 pg/ml to about 10 pg/ml, for example about 0.034 pg/ml to about 5 pg/ml of beta-carotene.
  • the nutritional composition can also include a source of anthocyanins. This can be in the form of a fruit or a fruit extract. Particularly useful fruits and fruit extracts include plum/prune, apple, pear, strawberry, blueberry, raspberry, cherry, and their combinations.
  • the nutritional composition can also contain various other conventional ingredients such as preservatives, emulsifying agents, thickening agents, buffers, fibres and prebiotics (e.g . fructooligosaccharides, galactooligosaccharides), probiotics (e.g . B. animalis subsp. lactis BB- 12, B. lactis HN019, B. lactis Bi07, B. infant is ATCC 15697, L. rhamnosus GG, L. rhamnosus HNOOI, L. acidophilus LA-5, L. acidophilus NCFM, L. fermentum CECT5716, B. longum BB536, B. longum AH 1205, B. longum AH 1206, B.
  • prebiotics e.g . fructooligosaccharides, galactooligosaccharides
  • probiotics e.g . B. animalis subsp. lactis BB- 12, B. lactis HN019, B
  • antioxidant/anti-inflammatory compounds including tocopherols, caroteinoids, ascorbate/vitamin C, ascorbyl palmitate, polyphenols, glutathione, and superoxide dismutase (melon), other bioactive factors (e.g . growth hormones, cytokines, TFG- ⁇ ), colorants, flavours, and stabilisers, lubricants, and so forth.
  • the nutritional composition can be in the form of a food, soluble powder, a liquid concentrate, or a ready-to-use formulation.
  • the composition can be eaten, drunk or can be fed via a nasogastric.
  • Various flavours, fibres and other additives can also be present.
  • the nutritional compositions can be prepared by any commonly used manufacturing techniques for preparing nutritional compositions in solid or liquid form.
  • the composition can be prepared by combining various feed solutions.
  • a protein-in-fat feed solution can be prepared by heating and mixing the lipid source and then adding an emulsifier (e.g . lecithin), fat soluble vitamins, and at least a portion of the protein source while heating and stirring.
  • a carbohydrate feed solution is then prepared by adding minerals, trace and ultra trace minerals, thickening or suspending agents to water while heating and stirring. The resulting solution is held for 10 minutes with continued heat and agitation before adding carbohydrates (e.g. the HMOs and digestible carbohydrate sources) .
  • the resulting feed solutions are then blended together while heating and agitating and the pH adj usted to 6.6-7.0, after which the composition is subjected to high-temperature short-time processing during which the composition is heat treated, emulsified and homogenized, and then allowed to cool.
  • Water soluble vitamins and ascorbic acid are added, the pH is adj usted to the desired range if necessary, flavours are added, and water is added to achieve the desired total solid level.
  • the resulting solution can then be aseptically packed to form an aseptically packaged nutritional composition.
  • the nutritional composition can be in ready-to-feed or concentrated liquid form.
  • the composition can be spray- dried and processed and packaged as a reconstitutable powder.
  • the nutritional composition can also be in the form of a food such as a nutritional bar, a yoghurt, etc. These forms can be produced using standard technologies and processes.
  • the total concentration of HMOs in the liquid, by weight of the liquid is from about 0.0001 % to about 2.0 %, including from about 0.001 % to about 1.5 %, including from about 0.01 % to about 1.0 %.
  • the total concentration of HMSs/HMOs in the liquid, by weight of the liquid is from about 0.0002 % to about 4.0 %, including from about 0.002 % to about 3.0 %, including from about 0.02 % to about 2.0 %.
  • the synthetic composition of this invention can also be in a unit dosage form such as a capsule, tablet or sachet.
  • the composition can be in a tablet form comprising the human milk oligosaccharides, and one or more additional components to aid formulation and administration, such as diluents, excipients, antioxidants, lubricants, colorants, binders, disintegrants, and the like.
  • Suitable diluents, excipients, lubricants, colorants, binders, and disintegrants include polyethylene, polyvinyl chloride, ethyl cellulose, acrylate polymers and their copolymers, hydroxyethyl-cellulose, hydroxypropylmethyl-cellulose (HPMC), sodium
  • PHEMA polyhydroxyethyl methacrylate
  • PVA polyvinyl alcohol
  • PVP polyvinyl pyrrolidone
  • PEO polyethylene oxide
  • PA polyacrylamide
  • carrageenan sodium alginate, polycarbophil, polyacrylic acid, tragacanth, methyl cellulose, pectin, natural gums, xanthan gum, guar gum, karaya gum, hypromellose, magnesium stearate,
  • Suitable antioxidants are vitamin A, carotenoids, vitamin C, vitamin E, selenium, flavonoids, polyphenols, lycopene, lutein, lignan, coenzyme Q10 ("CoQIO”) and glutathione.
  • the unit dosage forms can also include various nutrients including macronutrients.
  • the unit dosage forms can be administered orally, e.g. as a tablet, capsule, or pellet containing a predetermined amount, or as a powder or granules containing a predetermined concentration or a gel, paste, solution, suspension, emulsion, syrup, bolus, electuary, or slurry, in an aqueous or non-aqueous liquid, containing a predetermined concentration.
  • Orally administered compositions can include binders, lubricants, inert diluents, flavouring agents, and humectants.
  • Orally administered compositions such as tablets can optionally be coated and can be formulated so as to provide sustained, delayed or controlled release of the mixture therein.
  • Example 1 Treating high fat diet induced obesity and diabetes
  • mice 10-week-old C57BL/6J mice (100 mice) are housed in groups of five mice per cage, with free to water. The mice are divided into 10 groups of 10 mice, one control group and 9 treatment groups. All of the mice are fed a high-fat (HF) diet (60 % fat and 20 % carbohydrates
  • the 9 treatment groups are each administered one of the following : a) 2'-FL, b) 3-FL, c) 3'-SL, d) 6'-SL, e) LNT, f) LNnT, g) LNFP-I, h) DSLNT and i) a combination of these saccharides.
  • the control group is administered the HF diet only.
  • Intraperitoneal or oral glucose tolerance tests are performed as follows : 6-h-fasted mice are injected with glucose into the peritoneal cavity ( 1 g/kg glucose, 20 % glucose solution) or by gavage (3 g/kg glucose, 66 % glucose solution) . Blood glucose is determined with a glucose meter (Roche Diagnostics) on 3.5 ⁇ blood collected from the tip of the tail vein. A total of 20 ⁇ blood is sampled 30 min before and 15 or 30 min after the glucose load to assess plasma insulin concentration.
  • the intestinal permeability of 4000 Da fluorescent dextran-FITC (DX-4000-FITC) is measured .
  • M ice are fasted for 6 h before given DX-44-FITC by gavage (500 mg/kg body weight, 125 mg/ml) .
  • 120 ml of blood is collected from the tip of the tail vein.
  • the blood is centrifuged at 4 °C, 12 000 g for 3 min.
  • Plasma is diluted in an equal volume of PBS (pH 7.4) and analysed for DX- 4000-FITC concentration with a fluorescence spectrophotometer at an excitation wavelength of 485 nm and emission wavelength of 535 nm. Standard curves are obtained by diluting FITC-dextran in non-treated plasma diluted with PBS ( 1 : 3 v/v) .
  • Mice are anaesthetised (ketamine/xylazine, intraperineally, 100 and 10 mg/kg, respectively) after a 5 h period of fasting, and blood samples and tissues are harvested for further analysis. Mice are killed by cervical dislocation. Liver, caecum (full and empty), muscles (vastus lateralis), and adipose tissues (mesenteric and corresponding lymph nodes, epididymal, subcutaneous and visceral) are precisely dissected and weighed and stored at - 80 °C, for further analysis.
  • Total and active GLP1 are measured from blood with ELISA (M illipore, Molsheim, France) .
  • the caecal contents collected post mortem from mice are stored at -80 °C.
  • DNA is isolated from the caecal content samples using QIAamp DNA Stool Mini Kit. The DNA concentration of extracts is measured using NanoDrop.
  • Aliquots of 100 ng of extracted DNA are subjected to PCR using the 16S rDNA universal heteroduplex analysis (HDA) primers HDA1-GC 50-CGCCCGGGGCGCGCCCCGGGCGGG- GCGGGGGCACGGGGGGGGACTCCTACGGGAGGCAGCAGT-30 and HDA2 50- TTACCGCGGCTGCTGGCA-30 (both primers are disclosed in Walter et al. Appl. Environ.
  • HDA 16S rDNA universal heteroduplex analysis
  • Electrophoresis is carried out at 130 V for 4-5 hours at 60 °C. Polyacrylamide gels are stained with GelRede nucleic acid stain for 45 min, destained in ultrapure water and viewed under UV light. Bands of interest are excised from gels and lysed in ultrapure water. Extracted DNA is re-amplified using the same primers and PCR conditions. To purify the bacterial DNA, PCR products are reloaded on a denaturant gradient gel followed by excision and lysis of selected bands. DNA samples recovered from lysed bands of the second DGGE are re-amplified by PCR before purification using the QIAquick PCR Purification Kit and sequenced. Species identification is done using the Ribosomal Microbiome Database Project Classifier tool.
  • the V5-V6 region of 16S rRNA from caecal content DNA samples is amplified using the primers 784F 50- AGGATTAGATACCCT- GGTA-30 and 1061R 50-CRRCACGAGCTGACGAC-30 3640 (both primers are disclosed in Andersson et al. , PloS ONE 3 , e2836 (2008)) .
  • Amplicons are pyrosequenced using a Roche 454 GS-FLX system. Sequences of at least 240 nucleotides and containing no more than two undetermined bases are retained for taxonomic assignment.
  • the QIIME software is used for chimera check and the Greengenes database is used for classification. Bacterial diversity is determined at the phylum, family and genus levels.
  • HMOs are able to change the intestinal microbiota by increasing the abundance of bifidobacteria . Additionally, HMO supplementation increased the level of GLP1 and reduced body weight and adipose tissue. The level of GLP1 negatively correlated with food intake.
  • mice 10-week-old C57BL/6J mice ( 100 mice) are housed in groups of five mice per cage, with free access to food and water. The mice are divided into 10 groups of 10 mice, one control group and 9 treatment groups. All of the mice are fed a high-fat (HF) diet (60 % fat and 20 % carbohydrates [kcal/ 100 g], or an HF diet supplemented with HMO (20 g/kg of diet) for 8 weeks. Food and water intake are recorded twice a week.
  • HF high-fat
  • HMO 20 g/kg of diet
  • the 9 treatment groups are each administered one of the following : a) 2'-FL, b) 3-FL, c) 3'-SL, d) 6'-SL, e) LNT, f) LNnT, g) LNFP-I, h) DSLNT and i) a combination of these saccharides.
  • the control group is
  • Intraperitoneal or oral glucose tolerance tests are performed as follows : 6-h-fasted mice are injected with glucose into the peritoneal cavity ( 1 g/kg glucose, 20 % glucose solution) or by gavage (3 g/kg glucose, 66 % glucose solution) . Blood glucose is determined with a glucose meter (Roche Diagnostics) on 3.5 ⁇ blood collected from the tip of the tail vein. A total of 20 ⁇ blood is sampled 30 min before and 15 or 30 min after the glucose load to assess plasma insulin concentration.
  • the intestinal permeability of 4000 Da fluorescent dextran-FITC (DX-4000-FITC) is measured .
  • M ice are fasted for 6 h before given DX-44-FITC by gavage (500 mg/kg body weight, 125 mg/ml) .
  • 120 ml of blood is collected from the tip of the tail vein.
  • the blood is centrifuged at 4 °C, 12 000 g for 3 min.
  • Plasma is diluted in an equal volume of PBS (pH 7.4) and analysed for DX- 4000-FITC concentration with a fluorescence spectrophotometer at an excitation wavelength of 485 nm and emission wavelength of 535 nm. Standard curves are obtained by diluting FITC-dextran in non-treated plasma diluted with PBS ( 1 : 3 v/v) .
  • Plasma LPS, cytokines and gut hormones are determined as follows. Plasma LPS
  • Plasma cytokines interleukin (IL) la, ILlb, tumour necrosis factor (TNF) a, IL6, monocyte chemoattractant protein (MCP)- l, macrophage inflammatory protein ( ⁇ )- ⁇ , IL10, interferon (INF) c, IL15, IL18) and gut hormones (GLP1 (active), GIP (total), amylin (active), pancreatic polypeptide) are respectively determined in duplicate by using a Bio-Plex Multiplex kit, or a mouse gut hormones panel (LincoPlex), and measured by using Luminex technology, an EIA kit (GLP2 EIA kit) is used to quantify GLP2.
  • IL interleukin
  • TNF tumour necrosis factor
  • MCP monocyte chemoattractant protein
  • INF interferon
  • GLP1 active
  • GIP total
  • amylin active
  • Mice are anaesthetised (ketamine/xylazine, intraperineally, 100 and 10 mg/kg, respectively) after a 5 h period of fasting, and blood samples and tissues are harvested for further analysis. Mice are killed by cervical dislocation. Liver, caecum (full and empty), muscles (vastus lateralis), and adipose tissues (mesenteric and corresponding lymph nodes, epididymal, subcutaneous and visceral) are precisely dissected and weighed . The intestinal segments (jej unum, colon) are immersed in liquid nitrogen, and stored at -80 °C, for further analysis.
  • the caecal contents collected post mortem from mice are stored at -80 °C.
  • DNA is isolated from the caecal content samples using QIAamp DNA Stool Mini Kit. The DNA concentration of extracts is measured using NanoDrop. Aliquots of 100 ng of extracted DNA are subjected to PCR using the 16S rDNA universal heteroduplex analysis (HDA) primers HDA1-GC 50-CGCCCGGGGCGCGCCCCGGGCGGG- GCGGGGGCACGGGGGGGGACTCCTACGGGAGGCAGCAGT-30 and HDA2 50-
  • HDA 16S rDNA universal heteroduplex analysis
  • Electrophoresis is carried out at 130 V for 4-5 hours at 60 °C. Polyacrylamide gels are stained with GelRede nucleic acid stain for 45 min, destained in ultrapure water and viewed under UV light. Bands of interest are excised from gels and lysed in ultrapure water. Extracted DNA is re-amplified using the same primers and PCR conditions. To purify the bacterial DNA, PCR products are reloaded on a denaturant gradient gel followed by excision and lysis of selected bands. DNA samples recovered from lysed bands of the second DGGE are re-amplified by PCR before purification using the QIAquick PCR Purification Kit and sequenced. Species identification is done using the Ribosormal Microbiorme Database Project Classifier tool.
  • the V5-V6 region of 16S rRNA from caecal content DNA samples is amplified using the primers 784F 50- AGGATTAGATACCCT- GGTA-30 and 1061R 50-CRRCACGAGCTGACGAC-30 3640 (both primers are disclosed in Andersson et al. PloS ONE 3 , e2836 (2008)) .
  • Amplicons are pyrosequenced using a Roche 454 GS-FLX system. Sequences of at least 240 nucleotides and containing no more than two undetermined bases are retained for taxonomic assignment.
  • the QIIME software is used for chimera check and the Greengenes database is used for classification. Bacterial diversity is determined at the phylum, family and genus levels.
  • MAT mesenteric adipose tissue
  • MN lymph nodes
  • Quantification of bacterial DNA is performed by isolating genomic DNA from blood, MAT, MLN or intestine (contents and mucosa) . All bacterial DNA is quantified by quantitative real-time PCR targeting conserved regions of the 16S rRNA gene, with bacterial DNA as standard template for absolute quantification.
  • occludin and zonula occludens- 1 (ZO- 1) tight-junction proteins are assessed. Jej unum segments are immediately removed, washed with PBS, mounted in embedding medium, and stored at -80 °C until use.
  • Cryosections (5 mm) are fixed in acetone at -20 °C for 5 min for occludin and in ethanol for 30 min at room temperature and in acetone at -20 °C for 5 min for ZO- 1.
  • Non-specific background is blocked by incubation with 10% bovine serum albumin (BSA) in Tris-buffered saline (TBS) and 0.3% Triton X- 100 (30 min at room temperature) .
  • Sections are incubated with rabbit anti-occludin or rabbit anti-ZO- 1 ( 1 : 400 for ZO- 1 and 1 : 100 for occludin staining) for 2 h.
  • Sections are washed three times for 10 min in TBS and probed with goat anti-rabbit fluorescein isothiocyante (FITC)-conj ugated antibodies (1 : 50) . Slides are washed three times for 10 min in TBS and mounted in mounting medium. Sections are visualised on a
  • HMOs improve gut barrier function and reduce the metabolic inflammation and insulin resistance associated with obesity, and increase release of gut peptides, such as glucagon-like peptidel and 2 (GLP1 and GLP2) .
  • 10-week-old C57BL/6J mice 100 mice are housed in groups of five mice per cage, with free water. The mice are divided into 10 groups of 10 mice, one control group and 9 treatment groups. All of the mice are fed a high-fat (HF) diet (60 % fat and 20 % carbohydrates [kcal/100 g], or an HF diet supplemented with HMO (20 g/kg of diet) for 8 weeks. Food and water intake are recorded twice a week.
  • HF high-fat
  • the 9 treatment groups are each administered one of the following : a) 2'-FL, b) 3-FL, c) 3'-SL, d) 6'-SL, e) LNT, f) LNnT, g) LNFP-I, h) DSLNT and i) a combination of these saccharides.
  • the control group is administered the HF diet only.
  • Intraperitoneal or oral glucose tolerance tests are performed as follows : 6-h-fasted mice are injected with glucose into the peritoneal cavity ( 1 g/kg glucose, 20 % glucose solution) or by gavage (3 g/kg glucose, 66 % glucose solution) . Blood glucose is determined with a glucose meter (Roche Diagnostics) on 3.5 ⁇ blood collected from the tip of the tail vein. A total of 20 ⁇ blood is sampled 30 min before and 15 or 30 min after the glucose load to assess plasma insulin concentration.
  • Plasma triglyceride and cholesterol is measured from blood taken during the treatment period.
  • the intestinal permeability of 4000 Da fluorescent dextran-FITC (DX-4000-FITC) is measured .
  • M ice are fasted for 6 h before given DX-44-FITC by gavage (500 mg/kg body weight, 125 mg/ml) .
  • 120 ml of blood is collected from the tip of the tail vein.
  • the blood is centrifuged at 4 °C, 12 000 g for 3 min.
  • Plasma is diluted in an equal volume of PBS (pH 7.4) and analysed for DX- 4000-FITC concentration with a fluorescence spectrophotometer at an excitation wavelength of 485 nm and emission wavelength of 535 nm. Standard curves are obtained by diluting FITC-dextran in non-treated plasma diluted with PBS ( 1 : 3 v/v) .
  • Mice are anaesthetised (ketamine/xylazine, intraperineally, 100 and 10 mg/kg, respectively) after a 5 h period of fasting, blood samples and tissues are harvested for further analysis. Mice are killed by cervical dislocation. Liver, caecum (full and empty), and adipose tissues (mesenteric and corresponding lymph nodes, epididymal, subcutaneous and visceral) are precisely dissected, weighed and stored at -80 °C, for further analysis.
  • Total and active GLP1 are measured from blood with ELISA (M illipore, Molsheim, France) .
  • the caecal contents collected post mortem from mice are stored at -80 °C.
  • DNA is isolated from the caecal content samples using QIAamp DNA Stool Mini Kit. The DNA concentration of extracts is measured using NanoDrop.
  • Aliquots of 100 ng of extracted DNA are subjected to PCR using the 16S rDNA universal heteroduplex analysis (HDA) primers HDA1-GC 50-CGCCCGGGGCGCGCCCCGGGCGGG- GCGGGGGCACGGGGGGGGACTCCTACGGGAGGCAGCAGT-30 and HDA2 50- TTACCGCGGCTGCTGGCA-30 (both primers are disclosed in Walter et al. Appl. Environ.
  • HDA 16S rDNA universal heteroduplex analysis
  • Electrophoresis is carried out at 130 V for 4-5 hours at 60 °C. Polyacrylamide gels are stained with GelRede nucleic acid stain for 45 min, destained in ultrapure water and viewed under UV light. Bands of interest are excised from gels and lysed in ultrapure water. Extracted DNA is re-amplified using the same primers and PCR conditions. To purify the bacterial DNA, PCR products are reloaded on a denaturant gradient gel followed by excision and lysis of selected bands. DNA samples recovered from lysed bands of the second DGGE are re-amplified by PCR before purification using the QIAquick PCR Purification Kit and sequenced. Species
  • the V5-V6 region of 16S rRNA from caecal content DNA samples is amplified using the primers 784F 50- AGGATTAGATACCCT- GGTA-30 and 1061R 50-CRRCACGAGCTGACGAC-30 3640 (both primers are disclosed in Andersson et al. PloS ONE 3 , e2836 (2008)) .
  • Amplicons are pyrosequenced using a Roche 454 GS-FLX system. Sequences of at least 240 nucleotides and containing no more than two undetermined bases are retained for taxonomic assignment.
  • the QIIME software is used for chimera check and the Greengenes database is used for classification. Bacterial diversity is determined at the phylum, family and genus levels.
  • HMOs are able to change the intestinal microbiota by increasing the abundance of bifidobacteria . Additionally, HMO supplementation reduces cholesterol, body weight, fat accumulation and glucose tolerance.
  • mice Six-week-old ob/ob mice ( 120 mice) on C57BL/6 background are housed in a controlled environment ( 12 h daylight cycle) in groups of 2 mice/cage, and kept with free access to food and drinking water. The mice are separated into 10 groups of 10 mice, one control group and 9 treatment groups. One group is fed a control diet, and the 9 treatment groups each receive a control diets containing one of the following HMOs (20 g/kg of diet) for five weeks: a) 2'-FL, b) 3-FL, c) 3'-SL, d) 6'-SL, e) LNT, f) LNnT, g) LNFP-I, h) DSLNT, and i) a combination of these saccharides. Fresh food is given daily.
  • the investigational products contain 4.5 grams of either 2'-FL alone or a combination of 2'-FL and LNnT in a 4: 1 ratio while the placebo product contains 4.5 grams of glucose. All products are in powder form in a unit dosage container.
  • the patients are eligible to participate if: they are between 5 and 12 years of age, have a BMI SDS (Standard Deviation Score) of ⁇ 2.0 and are enrolled in the childhood obesity treatment program at the Children's Obesity Clinic. All recruited patients and their representatives are able and willing to understand and comply with the study procedures.
  • BMI SDS Standard Deviation Score
  • Patients are excluded if: they have participated in a clinical study one month prior to the screening visit and throughout the study; have any gastrointestinal disease(s) that may cause symptoms or may interfere with the trial outcome; have other severe disease(s) such as malignancy, kidney disease or neurological disease; have psychiatric disease; have used highly dosed probiotic supplements (yoghurt allowed) 3 months prior to screening and throughout the study; have consumed antibiotic drugs 3 months prior to screening and throughout the study; and consume on a regular basis medication that might interfere with symptom evaluation 2 weeks prior to screening and throughout the study.
  • Eligibility criteria are checked and for children who are enrolled to the study, medical history and concomitant medication are registered. A physical examination is done and pubertal staging is determined. Blood pressure, pulse rate, height and bodyweight are measured, and body composition is determined by a DXA (dual energy x-ray absorptiometry)-scan and bioimpedance. BMI SDS is calculated, waist and hip circumferences measured and food intake registered. Fasting blood samples are collected for safety and biomarker studies and for biobanking.
  • the serum from the blood samples is transferred to cryotubes and stored at -80° C.
  • the following biomarkers are measured; Lipopolysaccharides (LPS), hsCRP, free fatty acids, total cholesterol, HDL, LDL, HbAlc, glucose, insulin, triglycerides, TNF-a, IL- ⁇ , IL-6, IL-8, IL-10, GLP1, GLP2, Adiponectin, and Zonulin.
  • faecal samples Equipment for collecting faecal samples is distributed.
  • the faecal samples are stored at -80° C until analysis.
  • Microbiological analysis is performed on the faecal samples using 16S rRNA gene sequencing.
  • GSRS Gastrointestinal Symptom Rating Scale
  • BSFS Bristol Stool Form Scales
  • faecal samples are collected and equipment for collection of new samples is distributed.
  • BSFS is collected and new BSFS is distributed.
  • Study products are distributed together with a compliance form (diary). Patients and their representatives are reminded to follow the healthy dietary habits.
  • the study runs for 8 weeks with the patients consuming either a placebo or one of two investigational products daily. Patients are instructed to consume the products in the morning with breakfast. Compliance is monitored via a compliance form (diary) to be filled in daily.
  • each patient has a visit with the medical team. Patients and their representatives are asked about adverse events and any changes in the patient's usual medication. Study products and compliance forms are collected to check compliance.
  • BSFS and faecal samples are collected and equipment for collection of new samples is distributed. A physical examination is done and pubertal staging is determined. Blood pressure, pulse rate, height and bodyweight are measured, and body composition is determined by a DXA (dual energy x-ray absorptiometry)-scan and bioimpedance. BMI SDS is calculated, waist and hip circumferences measured and food intake registered. Fasting blood samples are collected for safety and biomarker studies and for biobanking, and equipment for collecting faecal samples is distributed. The GSRS questionnaire is completed on site by the participating child's representative(s).
  • BMI SDS Absorptiometry-scan and bioimpedance.
  • BMI SDS is calculated, waist and hip circumferences measured and food intake registered.
  • Fasting blood samples are collected for safety and biomarker studies and for biobanking. Faecal samples are collected.
  • results show that oral ingestion of HMOs modulate the intestinal microbiota, and specifically stimulate the growth of bifidobacteria.
  • the blood biomarker analysis indicates that the patients given the investigational products have increased levels of GLP1.
  • the level of GLP1 correlated positively with the abundance of bifidobacteria.
  • HMOs are able to increase bifidobacteria and change the intestinal environment, and by this, increasing the level of GLP1 in obese children.
  • results also show that oral ingestion of HMOs modulate the intestinal microbiota, and specifically stimulate the growth of bifidobacteria, particularly species belonging to the B. adolescentis phylogenetic group, and change the SCFA profile.
  • the blood biomarker analysis indicated that the patients given the investigational products have a lipid profile with lower triglyceride levels and higher high-density lipoprotein cholesterol. Additionally, the blood pressure and body composition is decreased . The abundance of bifidobacteria correlates negatively with the level of low-density lipoprotein cholesterol and positively with the level of high-density lipoprotein cholesterol. Collectively, HMOs are able to increase bifidobacteria and change the intestinal environment, and by this, improve the lipid profile, hypertension and body composition, all incidence reducing the risk of CVD.
  • the patients receiving the intervention product have lower HOMA-IR scores. Further, the patients given the investigational products show a greater reduction of body fat, body weight and BMI SDS as compared to the placebo group.
  • the blood biomarker analysis indicates that the patients given the investigational products have increased levels of GLP1 and GLP2, reduced levels of metabolic endotoxemia and inflammatory markers and reduced gut permeability indicating an improved mucosal barrier compared to the placebo.
  • the faecal analysis indicates that the patients given the investigational products have reduced bacterial dysbiosis and a higher level of bifidobacteria compared to the placebo, particularly
  • a ready to feed nutritional composition is prepared from water, maltodextrin, milk protein concentrate, Sucromalt, glycerine, cocoa powder, soy protein isolate, fructose, high oleic safflower oil, soy oil, canola oil, plant sterol esters, HMOs, soy lecithin, magnesium chloride, calcium phosphate, carrageenan, sodium ascorbate, potassium citrate, sodium phosphate, calcium citrate, choline chloride, potassium chloride, sodium citrate, magnesium oxide, taurine, L-carnitine, alpha-tocopheryl acetate, zinc sulphate, ferrous sulphate, niacinamide, calcium pantothenate, vitamin A palmitate, citric acid, manganese sulphate, pyridoxine hydrochloride, vitamin D3, copper sulphate, thiamine mononitrate, riboflavin, beta carotene, folic acid, biotin, potassium iodide
  • the composition has an energy density of 0.8 kcal/ml with an energy distribution (% of kcal) as follows : protein : 20 %, carbohydrate : 48 %, fat: 32 %.
  • a tablet is prepared from HMO, hydroxypropyl methylcellulose, sodium alginate, gum, microcrystalline cellulose, colloidal silicon dioxide, and magnesium stearate. All raw materials except the magnesium stearate are placed into a high shear granulator and premixed . Water is sprayed onto the premix while continuing to mix at 300 rpm. The granulate is transferred to a fluidised bed drier and dried at 75 °C. The dried powder is sieved and sized using a mill. The resulting powder is then lubricated with magnesium stearate and pressed into tablets. The tablets each contain 325 mg of HMO. The tablets each have a weight of 750 mg.
  • a capsule is prepared by filling about 1 g of HMO into a 000 gelatine capsule using a filing machine. The capsules are then closed. The HMO are in free flowing, powder form.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Polymers & Plastics (AREA)
  • Nutrition Science (AREA)
  • Engineering & Computer Science (AREA)
  • Food Science & Technology (AREA)
  • Mycology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

This invention relates to a method and composition for regulating satiety and stabilising or reducing insulin resistance and risk of, preventing, or treating CVD and associated co‐morbidities in humans, in particular in overweight or obese human individuals.

Description

SYNTHETIC COMPOSITIONS COMPRISING HUMAN MILK OLIGOSACCHARIDES FOR USE IN THE PREVENTION AND TREATMENT OF DISORDERS
Fl ELD OF I N VENTI ON
This invention relates to a method and composition for regulating satiety and stabilising or reducing insulin resistance and risk of, preventing, or treating CVD and associated comorbidities in humans, in particular in overweight or obese human individuals.
BACKGROU N D OF TH E I NVENTI ON
The increasing trend of obese individuals has become a major health issue over the past several decades and World Health Organization (WHO) has declared obesity as a global epidemic. According to WHO, it was estimated that more than 1.9 billion adults were overweight in 2014, and among them, at least 600 million were obese. This means that obesity has more than doubled since 1980 worldwide (WHO, fact sheet from January 2015) . The ra pid increase in obesity over such a short time frame makes a novel genetic cause per se unlikely and strongly favours modified environmental factors over the past 30 years. Such environmental factors include dietary habits, exercise or energy expenditure, and lifestyle. Indeed, there appears to be a strong correlation between Westernization in terms of diet and lifestyle and obesity. A shift from more traditional diets, rich in whole-plant foods like whole- grain cereals, fruits, and vegetables, to modern Western-style diets rich in refined carbohydrates, fat, and red/processed meats and low in fibre and whole-plant foods, is strongly correlated with increased body weight, obesity, and the diseases of obesity
(Conterno et al. Genes Nutr. 6 , 241) .
Overweight and obesity is commonly associated with accumulated abdominal visceral fat and can be related to psycho-sociological behavioural disorders. It is often associated with the development of several chronic complications, such as high fasting glucose levels
(hyperglycaemia), elevated triglyceride levels (hypertriglyceridemia), low levels of high density lipoprotein (dyslipidaemia) and high blood pressure (hypertension) . Individuals who meet at least three of these criteria are clinically diagnosed as having metabolic syndrome, which increases the risk of developing metabolic diseases such as type 2 diabetes and cardiovascular diseases (CVD) (Boulange et al., Genome Medicine 8 , 42 (2016)) .
Many obese patients have metabolic dysfunctions, even at early stages and tend to develop comorbidities such as type 2 diabetes. Tissue resistance to insulin's actions (insulin resistance) has been postulated as the initial impairment underlying the onset of the metabolic comorbidities in these patients.
Type 2 diabetes is a metabolic disorder that is characterised by hyperglycaemia due to insulin resistance and relative lack of insulin and is a rapidly growing global epidemic. The
International Diabetes Federation (IDF) reports that as of 2013 there were more than 382 million people living with diabetes, and a further 316 million with impaired glucose tolerance who are at high risk from the disease (IDF Diabetes Atlas, 6 edn.) . WHO furthermore estimates that 90 percent of people around the world who suffer from diabetes suffer from type 2 diabetes. Long-term complications from high blood sugar can include heart disease, strokes, diabetic retinopathy, kidney failure, and poor blood flow in the limbs.
High low-density lipoprotein cholesterol (LDL-C) and triglyceride concentrations and low high- density lipoprotein cholesterol (HDL-C) in the blood is a precursor to hypertension, hyperlipidaemia, and causes the formation and build-up of atherosclerotic plaque in the arteries leading to higher risk of CVD. Cardiovascular risk factors are not only observed in adults, but also obese children and young adults suffer from dyslipidaemia, hypertension, hyperinsulinemia or insulin resistance (Bridger, Paediatr. Child Health 1 4 , 177 (2009)) .
Cholesterol concentrations within the circulatory pool are products of input from gut absorption and endogenous synthesis relative to clearance through hepatic and extrahepatic tissue pathways. A disruption in any of these mechanisms can alter this balance, which is reflected in plasma cholesterol concentrations and subsequent CVD progression (Matthan et al. J. Am. Heart Assoc. 2 , e005066 (2013)) .
Diet, and the gut microbiota is proposed to play a key role in the rapid rise in obesity and metabolic diseases (Burcelin et al. Frontiers in Bioscience 1 4 , 5107 (2009) . The extent to how the gut microbiota play a causal role in the development of obesity and metabolic diseases is unclear. However, in diet-induced obesity, over-nutrition can alter composition of the gut microbiota, with dietary nutrients influencing the growth of certain species. Diets rich in cholesterol, saturated fats, and simple carbohydrates are associated with a gut microbiota rich in particular organisms belonging to the Firmicutes phylum. In line with this, marked differences in the gut microbiota have been observed between healthy, obese, and type 2 diabetic patients (Backhed et al. , PNAS 1 01 , 15718 (2004), Qin et al. , Nature 490 , 55 (2012)) with fewer Bacteroidetes and more Firmicutes in obese than lean people. However, this proportion has shown to change with weight loss leading to increase in the abundance of Bacteroidetes and decrease in the abundance of Firmicutes (Ley et al. , Nature 444 , 1022 (2006)) . Additionally, specific changes at genus level has been observed with lower number of bifidobacteria in obese versus lean and diabetic versus non-diabetic individuals (Schwiertz et al. , Obesity 1 8 , 190 (2009)) . Hence, compelling evidence suggests that the gut microbiota serves as a pivotal contributing factor in the development of diet-related obesity in humans, and can affect metabolic regulation and alter energy homeostasis (Zhang et al. , EBioMedicine 2 , 966 (2015)) .
Gut microbiota is a specific entity within the body which has its own genome and whose gene pool is much more abundant than the one of its host. It has been estimated that the human intestine harbours 1013 to 1014 bacterial cells and the number of bacteria outnumbers the total number of cells in the body by a factor of 10 (Gill et al. , Science 31 2 , 1355 (2006)) . The physiologic functions attributed to gut microbiota have extended to extra-intestinal tissues, such as the liver, brain and adipose tissue, constructing connections with obesity and related disorders including type 2 diabetes and CVD. The dysbiosis of gut microbiota has the potential to affect gut permeability and, consequently, give rise to metabolic endotoxemia and higher plasma lipopolysaccharide (LPS) . In addition, gut peptides such as glucagon-like peptide 2 (GLP2) can play a key role in these processes (Tremaroli et al. Nature 489 , 242 (2012)) . For example, GLP2, which is secreted by intestine L cells, is a key regulator of intestinal permeability (Cani et al. Gut 58 , 1091 (2009)) . Therapeutic regimes that target intestinal microbiota and intestinal barrier therefore show a broad prospect in treating metabolic diseases such as diabetes (Kootte et al. Diabetes, Obesity & Metabolism 1 4 , 112 (2012)) .
However, the gut microbiota does not only participate in whole-body metabolism by affecting energy balance (Turnbaugh et al. Nature 444 , 1027 (2006)) and glucose metabolism (Cani et al. Diabetes 57 , 1470 (2008)) but is also involved in development of the low-grade inflammation (Cani et al. Gut 58 , 1091 (2009)) associated with obesity and related metabolic disorders such as diabetes. The association between inflammation and type 2 diabetes was described in the 1950s, when epidemiological studies showed a rise in acute-phase response proteins in serum of type 2 diabetic patients compared with controls (Fearnley et al. Lancet 274 , 1067 ( 1959)) . Later, a specific link between inflammatory and metabolic responses was made with the discovery that compared with lean tissue, obese adipose tissue secretes inflammatory cytokines and that these inflammatory cytokines themselves can inhibit insulin signalling (Hotamisligil et al. Science 271 , 665 ( 1996)) . The definitive proof of a connection between inflammatory mediators and insulin resistance in obesity and type 2 diabetes came from genetic studies that interfered with inflammatory mediators and demonstrated beneficial effects of this interference on insulin action (Uysal et al. Nature 389 , 610 (1997)) .
In recent years, gut microbiota derived LPS has been shown to be involved in the onset and progression of inflammation, and in pathological situations, such as obesity and type 2 diabetes, LPS play a major role in the onset of disease (Cani et al. Diabetes 57 , 1470
(2008)) . After only one week of a high-fat diet in mice, commensal intestinal bacteria are translocated from the intestine into adipose tissue and the blood where they can induce inflammation (Amar et al. EMBO Mol. Med. 3 , 559 (2011)) . This metabolic bacteraemia is characterized by an increased co-localization with dendritic cells from the intestinal lamina propria and by an augmented intestinal mucosal adherence of non-pathogenic Escherichia coli. The bacterial translocation process from intestine towards tissue with resulting inflammation was reversed by six weeks of treatment with the probiotic strain
Bifidobacterium animalis subsp. lactis 420, suggesting an involvement of the microbiota . Normally insulin secretion is proportional to blood glucose levels. However, in some individuals, body tissue does not respond properly to insulin. The insulin receptors in body tissue do not function properly and cells inadequately recognise the presence of insulin. As a result, the pancreas needs to secrete more insulin. This phenomenon is called insulin resistance (or impaired insulin sensitivity) . Reducing insulin resistance, or at least preventing its increase, is therefore desired, especially in obese people to prevent or slow down disease progression.
An dysbiotic gut microbiota can also play a key role in food intake and appetite sensation, since gut bacteria and especially their SCFA metabolites can influence gut satiety hormone levels such as glucagon-like peptide 1 (GLP1) and peptide YY (PYY) (Sanchez et al. , Int. J. Environ. Res. Public Health 1 2 , 162 (2015)) . GLP1 and PYY is of relevance to appetite and weight maintenance because they have action on the gastrointestinal tract as well as the direct regulation of appetite. Hence, regulation of these hormones in humans can reduce food intake, appetite and hunger, and promote fullness and satiety with the ultimate result of promoting weight loss (Shah, Rev. Endocr. Metab. Disord. 1 5 , 181 (2014)) .
Hence, it would be advantageous to be able to prevent or reduce the damaging consequences of a dysbiotic microbiota in overweight and obesity. Modulation of the microbiota increasing the abundance of beneficial bacteria could be a way to help regulate food intake and interrupt the processers involved in type 2 diabetes and CVD. Beneficial bacteria such as bifidobacteria have shown to ameliorate both metabolic and immunological dysfunctions related to obesity. As an example, Bifidobacterium pseudocatenulatum has shown to reduce serum cholesterol, triglyceride and glucose levels and decrease insulin resistance and improve glucose tolerance in obese mice. Additionally, the species can reduce liver steatosis and the number of larger adipocytes in enterocytes of obese mice (Cano et al., Obesity, 21 , 2310 (2013)) .
One mode of action for lowering cholesterol by bifidobacteria is the processing of bile salts. Metabolism of cholesterol, a precursor of bile acids, is mediated through the bacteria expressing the enzyme bile salt hydrolase (BSH) . Some bifidobacteria have high BSH activity promoting deconjugation of bile acids in the gut to secondary amino acid conj ugates. When these secondary conj ugates are excreted, cholesterol is broken down to replace the processed bile salts. Overall, this process promotes the catabolism of cholesterol, leading to reduced serum levels (Ettinger et al., Gut Microbes 5 , 719 (2014)) . Another mechanism is through bacterial metabolites like short chain fatty acids (SCFA), including acetate, propionate and butyrate. Acetate has shown to be negatively associated with visceral adipose tissue and insulin levels in obese individuals and propionate has shown to reduce lipogenesis and cholesterol synthesis inhibition (Verbeke et al. , Nutrition Research Reviews 28 , 42 (2015)) .
Selective stimulation of specific beneficial intestinal bacteria to promote their growth and metabolic activity (e.g . production of SCFA) could be a helpful approach in creating an intestinal environment that is able to regulate metabolic functions and energy balance. For example, a study has described that consuming inulin increased the gut microbiota fermentation, decreased appetite, improved postprandial glucose responses and higher concentrations of GLP1 and PYY after two weeks of prebiotic treatment (Cani et al. , Am. J. Clin. Nutr. 90 , 1236 (2009)) . However, in some individuals, inulin may provoke side effects such as bloating, abdominal pain and increased flatulence. Probiotic supplementation could be an approach, however, the addition of a small number of different probiotics to the intestine is unlikely to fully promote a beneficial intestinal microbiota composition with sufficient production of metabolites.
Human milk oligosaccharides (HMOs) are a heterogeneous mixture of soluble glycans found in human milk. They are the third most abundant solid component after lactose and lipids in human milk and are present in concentrations of 5-25 g/l (Gabrielli et al., Pediatrics 1 28 , 1520 (2011)) . HMOs are resistant to enzymatic hydrolysis in the small intestine and are thus largely undigested and unabsorbed (ten Bruggencate et al. , Nutrition Reviews 72 , 377 (2014)) . The majority of HMO that reaches the colon serves as a substrate to shape the gut ecosystem by selectively stimulating the growth of specific beneficial bacteria . HMOs are believed to substantially modulate the infant gut microbiota and play a decisive role in the differences in the microbiota of formula-fed and breast-fed infants. These differences include the predominance of Bifidobacterium in the gut of breast-fed infants compared to a more diverse gut microbiota in formula-fed infants (Bezirtzoglou et al., Anaerobe 1 7 , 478 (2011) . This is viewed as beneficial for the infant because strains of Bifidobacterium species and their metabolites are believed to have a positive effect on human health (Chichlowski et al., J. Pediatr. Gastroenterol. Nutr. 55 , 321 (2012) ; Fukuda et al. , Nature 469 , 543 (2011)) .
Recently, it has also been demonstrated that some sialylated and fucosylated HMOs has a positive effect on the growth of certain strains of bifidobacteria that are typically found in both infant and adult microbiota (WO 2013/154725) .
EP-A- 1332759 discloses that oral doses of 2'-FL, 3'-SL, 6'-SL, LNnT and sialic acid promote insulin secretion in type 2 diabetes-model mice.
EP-A-2143341 discloses that a mixture of GOS, sialylated oligosaccharides and N-acylated oligosaccharides reduces triglyceride concentration in liver in model mice.
EP-A-2332552 discloses that 3'-SL and 6'-SL reduce/prevent fat accumulation in the liver and other organs in high-fat diet mice and rats.
WO 2013/057061 discloses a composition for increasing insulin sensitivity and/or reducing insulin resistance that contains long chain polyunsaturated fatty acids, probiotics and a mixture of oligosaccharides containing at least one of lacto-N-neotetraose (LNnT) and lacto- N-tetraose (LNT), at least one N-acetylated oligosaccharide different from LNnT and LNT, at least one sialylated oligosaccharide and at least one neutral oligosaccharide. This composition can also contain 2'-0-fucosyllactose (2'-FL) . The composition is particularly adapted for use in infants who were born preterm and/or who experienced IUGR, and in pregnant women suffering from gestational diabetes. It is also stated that the composition can be given to children, adolescents and adults suffering from insulin resistance and/or type II diabetes. It is stated that the efficacy of the composition can be the result of the synergistic combination of immunity modulator effects triggered by the probiotics and the LC-PUFA through their stimulation with the specific oligosaccharide mixture. WO 2013/036104 discloses a method for inter alia improving regulation of satiety in a human subject having an age of 0 to 36 months by feeding a nutritional composition comprising lipid globules.
WO 2014/ 187464 discloses a synthetic mixture of oligosaccharides comprising at least 6 oligosaccharides selected from fucosylated, sialylated, sulfated, GlcNAc- GalNAc- and mannose-containing oligosaccharides, for treating a microbiota of a human, to reduce or eliminate the activity and/or the proportion of a microbe in the microbiota that is associated with the development or maintenance of a cardiovascular disease.
However, there remains a need for effective interventions which are able to manage food intake by improving regulation of satiety, stabilise or improve insulin resistance in patients having an obesity-related, metabolic disorder and/or prevent or reduce CVD and long-term effects of CVD in CVD patients having an obesity-related, metabolic disorder (especially where the patients are overweight or obese), which are safe, well tolerated and well accepted .
SU M MARY OF TH E I N VEN TI ON
The present invention provides one or more HMOs and synthetic compositions comprising one or more HMOs that can be advantageously used for
- for regulating satiety in a human individual having a metabolic disorder, for example obesity, obesity induced pre-diabetes and obesity induced type 2 diabetes;
- for stabilising or reducing insulin resistance in a human individual having an obesity- related metabolic disorder, for example obesity, obesity induced pre-diabetes and obesity induced type 2 diabetes; and/or
to reduce the risk of, prevent or treat CVD or CVD-associated pathologic condition or disease in a human, preferably, in an overweight or obese human individual.
Accordingly, a first aspect of this invention relates to one or more human milk
oligosaccharides for use in
- regulating satiety in a human having a metabolic disorder,
- reducing propensity to obesity in a human having a metabolic disorder,
- stabilising or reducing insulin resistance in a human individual having an obesity- related metabolic disorder, for example obesity, obesity induced pre-diabetes and obesity induced type 2 diabetes,
- reducing the propensity of a cardiovascular disease (CVD) and/or a CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual, - for preventing development of a cardiovascular disease (CVD) and/or a CVD- associated pathological condition or disease in a human, preferably, in an overweight or obese human individual, and/or
- treating a cardiovascular disease (CVD) and/or a CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual.
In one embodiment, the one or more HMOs is/are lacto-N-tetraose (LNT), lacto-N- neotetraose (LNnT), 2'-fucosyllactose (2'-FL), lacto-N-fucopentaose I (LNFP-I), 3- fucosyllactose (3-FL), difucosyllactose (DFL), 3'-sialyllactose (3'-SL) or 6'-sialyllactose (6'- SL), or a mixture thereof.
A second aspect of the invention relates to one or more human milk oligosaccharides (HMOs) selected from the group consisting of fucosylated HMOs and non-fucosylated HMOs, preferably a mixture of one or more fucosylated HMOs and one or more non-fucosylated neutral HMOs, for use in increasing the abundance of bifidobacteria in a human, preferably in an overweight or obese human having a propensity of, or diagnosed with a cardiovascular disease (CVD).
A third aspect of the invention relates to a synthetic composition for use in
- regulating satiety in a human having a metabolic disorder, the composition comprising an effective amount of one or more human milk oligosaccharides,
- reducing propensity to obesity in a human having a metabolic disorder, the
composition comprising an effective amount of one or more human milk
oligosaccharides,
- stabilising or reducing insulin resistance in a human individual having an obesity- related metabolic disorder, for example obesity, obesity induced pre-diabetes and obesity induced type 2 diabetes,
- reducing the propensity of a cardiovascular disease (CVD) and/or a CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual,
- for preventing development of a cardiovascular disease (CVD) and/or a CVD- associated pathological condition or disease in a human, preferably, in an overweight or obese human individual, and/or
- treating a cardiovascular disease (CVD) and/or a CVD-associated pathological condition or disease in a human, preferably in an overweight or obese human individual,
the synthetic composition comprising an effective amount of one or more human milk oligosaccharides (HMOs), for example those specified in the first aspect. Preferably the amount of a human milk oligosaccharide is effective to increase the abundance, particularly the relative abundance, of bifidobacteria in the gastrointestinal tract of the human. In one embodiment, in a period of about 14 days of treatment the bifidobacteria increased is a member of the phylogenetic Bifidobacterium adolescentis group, for example, Bifidobacterium pseudocatenulatum and/or Bifidobacterium adolescentis, and, after about 14 days of treatment, are Bifidobacterium longum and/or Bifidobacterium bifidum .
A fourth aspect of the invention relates to a synthetic composition for use in increasing the abundance of bifidobacteria in a human, preferably, in an overweight or obese human having a propensity of, or diagnosed with a cardiovascular disease (CVD), the synthetic composition comprising one or more human milk oligosaccharides (HMOs) selected from the group consisting of fucosylated HMOs and non-fucosylated neutral HMOs, preferably a mixture of one or more fucosylated HMOs and one or more non-fucosylated neutral HMOs.
A fifth aspect of this invention provides a method for
- regulating satiety in a human having a metabolic disorder,
- reducing propensity to obesity in a human having a metabolic disorder,
- stabilising or reducing insulin resistance in a human individual having an obesity- related metabolic disorder, for example obesity, obesity induced pre-diabetes and obesity induced type 2 diabetes,
- reducing the propensity of a cardiovascular disease (CVD) and/or a CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual,
- for preventing development of a cardiovascular disease (CVD) and/or a CVD- associated pathological condition or disease in a human, preferably, in an overweight or obese human individual, and/or
- treating a cardiovascular disease (CVD) and/or a CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual,
the method comprising administering, to the human, an effective amount of one or more human milk oligosaccharides, or a synthetic composition comprising an effective amount of one or more human milk oligosaccharides.
A sixth aspect of the invention relates to a method for increasing the a bundance of bifidobacteria in a human, preferably in an overweight or obese human having a propensity of, or diagnosed with a cardiovascular disease (CVD), the method comprising administering, to the human, one or more human milk oligosaccharides (HMOs) selected from the group consisting of fucosylated HMOs and core HMOs, preferably of a mixture of one or more fucosylated HMOs and one or more core HMOs, or a synthetic composition comprising one or more human milk oligosaccharides (HMOs) selected from the group consisting of fucosylated HMOs and core HMOs, preferably of a mixture of one or more fucosylated HMOs and one or more core HMOs.
In one embodiment, with regard to any of the aspects mentioned above, the one or more HMOs are administered to a human in need in two steps:
(a) in a first step, during an initial treatment period of about 14 days, to increase the relative abundance of bifidobacteria of the phylogenetic Bifidobacterium adolescentis group; and
(b) in a second step, during an additional period of treatment of 1 or more days following the initial treatment period , to increase the relative abundance of
Bifidobacterium longum and/or Bifidobacterium bifidum,
in the microbiota in the gastro-intestinal tract of said human.
D ETAI LED DESCRI PTI ON OF TH E I NVENTI ON
Terms and definitions
The term "patient" means a human individual who was diagnosed by a medical or health professional personal as having a disease, e.g . a metabolic disorder, and receiving or registered to receive medical treatment. The patient can be a paediatric or adult patient. An embodiment of the patent e.g. is an overweight or obese individual that is having a CVD or a CVD-associated pathological condition or disease.
"Paediatric patient" can be a human patient of 3-21 years old . In some embodiments, a "patient" can also be any other mammal.
The terms "non-infant human" and "non-infant" all mean in the present context a human individual of at least 3 years old . A human can be a child, a teenager, an adult or an elderly adult, preferably, the human is an individual of at least 3 years old that has an excess of body fat, more preferably, an individual whose excess body fat has accumulated to the extent that it may have a negative effect on health, i.e. an overweight or obese human individual.
Body fat percentage preferably means total mass of body fat divided by total mass of the body.
The term "obese human" or "obese human individual" means that a human individual that has a body mass index (BMI), a measurement obtained by dividing the individual's weight by the square of the individual's height, over 30 kg/m2, with the range 25-30 kg/m2 defined as overweight.
Overweight and obesity for children and teens (human individuals aged 3- 19 years old) is defined as the following : overweight is defined as a BMI at or above the 85th percentile and below the 95th percentile for children and teens of the same age and sex. Obesity is defined as a BMI at or above the 95th percentile for children and teens of the same age and sex (see: Rolland-Cachera, Int. J. Pediatr. Obesity 6 , 325 (2011)) .
The term "metabolic disorder" (also known as "metabolic syndrome") in the present context means an inherited or acquired genetic or physiological condition which is reflected by a change in metabolism of an individual, typically is a clustering of at least three of the five following medical conditions (giving a total of 16 possible combinations giving the syndrome) : abdominal (central) obesity (cf. TOFI)
elevated blood pressure
elevated fasting plasma glucose
- high serum triglycerides
low high-density lipoprotein (HDL) levels.
A metabolic disorder often happens when abnormal chemical reactions in the body alter the normal metabolic process, e.g . disrupt the normal production and utilizing of energy in the body from the food uptake. Metabolic syndrome is associated with the risk of
developing cardiovascular disease and type 2 diabetes. In some preferred embodiments, the invention relates to metabolic disorders, such as obesity, obesity induced pre-diabetes, obesity induced type 2 diabetes. In one preferred embodiment, a metabolic disorder is insulin resistance or is associated with insulin resistance. The term "insulin resistance" means a pathological condition in an individual who has a reduced insulin activity, e.g. due to the individual's cells fail to respond normally to the hormone insulin, or insulin is underproduced in the individual's body, or it has altered activity. In one preferred embodiment of the invention, insulin resistance is in an overweight or obese individual.
The term "cardiovascular disease (CVD)" refers broadly to any disease of the heart and circulatory system (arteries and veins) . Cardiovascular disease generally refers to conditions that involve narrowed or blocked blood vessels that can lead to a heart attack, chest pain
(angina) or stroke. Other heart conditions, such as those that affect the heart muscle, valves or rhythm, also are also contemplated as forms of heart disease. Examples of CVD include, but not limited to, coronary artery disease (blockage of blood vessels that serve the heart), acute coronary syndrome (symptoms such as pain, weakness, and tiredness caused by coronary artery disease), angina pectoris (pain resulting from coronary artery disease or other causes), myocardial infarction (heart attack, with damage to heart muscle caused by coronary artery disease), and left ventricular thrombus (blood clot in the left side of the heart that pumps blood into your body) . CVD may be accompanied with health complications (that are interchangeably referred herein as pathologic conditions) or associated diseases, which are also contemplated herein. Some non-limiting examples of relevant contemplated health complications and CVD-associated diseases/pathologic conditions include heart failure (occurs when the heart cannot adequately pump blood throughout the body; this can cause shortness of breath, dizziness, confusion, and the build-up of fluid in the body, causing swelling), heart attack (occurs when the coronary arteries narrow so much that they cut off blood supply to the heart; the heart cells begin to die as they are deprived of oxygen and symptoms include shortness of breath and severe chest pain that may radiate to the back, jaw, or left arm), stroke (occurs due formation and lodging of blood clots in a blood vessel in the brain and cutting thus off blood flow; stroke symptoms include : numbness on one side of the body, confusion, trouble, speaking, loss of balance or coordination), pulmonary embolism (is similar to a stroke, but the blocked blood vessel is in the lungs instead of the brain; symptoms include shortness of breath, chest pain on breathing, and bluish skin), cardiac arrest (occurs when the heart suddenly stops beating ; this will lead to death if not treated immediately), Peripheral Artery Disease (PAD) (occurs due to narrowing in the arteries that supply blood to the arms and legs; the main symptom of PAD is severe leg pain when walking) .
The term "propensity "in the present context means natural tendency of a human individual to develop later in life a medical condition, such as a disease, in particular a CVD or a CVD- associated pathological condition or disease.
The term "preventing CVD and/or CVD associated pathological condition or disease" in the present context means eliminating or minimising a chance of development of a CVD disease or a pathological condition or disease associated with an CVD. Both primary and secondary prevention are thus contemplated. The primary prevention means preventing a CVD or associated disease or condition before it occurs, and the secondary prevention means preventing additional attacks of a CVD or development of associated condition or disease after the first attack has occurred .
The term "enteral administration" preferably means any conventional form for delivery of a composition to a human that causes the deposition of the composition in the gastrointestinal tract (including the stomach) . Methods of enteral administration include feeding through a naso-gastric tube or jej unum tube, oral, sublingual and rectal.
The term "oral administration" preferably means delivery into the gastrointestinal tract through the oral cavity. As such, oral administration is a form of enteral administration.
The term "effective amount" preferably means an amount of a human milk oligosaccharide or an amount of a composition that provides a human milk oligosaccharide sufficient to render a desired treatment outcome in a patient. An effective amount can be administered in one or more doses to the patient to achieve the desired treatment outcome. In some embodiments, the term "effective amount" may mean an amount of a single HMO, or a combination of different HMOs that is capable of increasing the abundance of bifidobacteria in the gastro- intestinal tract of a human individual of the invention, preferably, relative abundance of members of the Bifidobacterium adolescentis phylogenetic group in particular B. adolescentis and/or B. pseudocatenulatum . The term "relative abundance of bifidobacteria" means the abundance of bifidobacteria relative to other genus in the microbiota of the gastro-intestinal tract.
The term "human milk oligosaccharide" or "HMO" preferably means a complex carbohydrate consisting of a small number, typically 3- 10, of monosaccharide units attached to each other by an interglycosidic linkage that can be found in human breast milk and that can be in acidic or neutral form. More than about 200 different HMO structures are known to exist in human breast milk (Urashima et al. : Milk Oligosaccharides, Nova Biomedical Books, New York, 2011) . HMOs can be core, fucosylated and sialylated oligosaccharides. Core HMOs are non- fucosylated neutral (that is non-charged) HMOs and consist of Glu, Gal and GlcNAc (thus devoid of Fuc and sialic acid) . Examples of core HMOs include lacto-N-tetraose (LNT), lacto- N-neotetraose (LNnT), lacto-N-neohexaose (LNnH) , lacto-N-hexaose (LNH) and p-lacto-N- neohexaose (pLNnH) . Fucosyl HMOs are fucosylated lactoses or fucosylated core HMOs such as 2'-fucosyllactose (2'-FL), lacto-N-fucopentaose I (LNFP-I), lacto-N-difucohexaose I (LNDFH-I), 3-fucosyllactose (3-FL), difucosyllactose (DFL), lacto-N-fucopentaose III (LNFP- III), fucosyl-para-lacto-N-neohexaose (F-pLNnH), lacto-N-difucohexaose I (LNDFH-I), fucosyl-lacto-N-hexaose II (FLNH-II), lacto-N-fucopentaose V (LNFP-V), lacto-N- difucohexaose II (LNDFH-II), fucosyl-lacto-N-hexaose I (FLNH-I), fucosyl-lacto-N-hexaose III (FLNH-III) and fucosyl-para-lacto-N-neohexaose (F-pLNnH) . Sialyl HMOs are sialylated lactoses or sialylated core HMOs such as 3',6-disialyllacto-N-tetraose (DSLNT), 6'-sialyllactose (6'-SL), 3'-sialyllactose (3'-SL), 6'-sialyllacto-N-neotetraose (LST c), 3'-sialyllacto-N-tetraose (LST a) and 6-sialyllacto-N-tetraose (LST b) . Examples for sialylated and fucosylated HMOs include disialyl-fucosyl-lacto-N-hexaose II (DSFLNH-II), fucosyl-sialyl-lacto-N-neohexaose I (FSLNnH-I), fucosyl-sialyl-lacto-N-hexaose I (FSLNH-I) and 3-fucosyl-3'-sialyllactose (FSL) .
"Microbiota", "microflora" and "microbiome" preferably mean a community of living microorganisms that typically inhabits a bodily organ or part, particularly the gastro-intestinal organs of non-infant humans. The most dominant members of the gastrointestinal microbiota include microorganisms of the phyla of Firmicutes, Bacteroidetes, Actinobacteria,
Proteobacteria, Synergistetes, Verrucomicrobia, Fusobacteria, and Euryarchaeota; at genus level Bacteroides, Faecalibacterium, Bifidobacterium, Roseburia, Alistipes, Collinsella, Blautia, Coprococcus, Ruminococcus, Eubacterium and Dorea; at species level Bacteroides uniformis, Alistipes putredinis, Parabacteroides merdae, Ruminococcus bromii, Dorea longicatena, Bacteroides caccae, Bacteroides thetaiotaomicron, Eubacterium hallii, Ruminococcus torques, Faecalibacterium prausnitzii, Ruminococcus lactaris, Collinsella aerofaciens, Dorea
formicigenerans, Bacteroides vulgatus and Roseburia intestinalis. The gastrointestinal microbiota includes the mucosa-associated microbiota, which is located in or attached to the mucus layer covering the epithelium of the gastrointestinal tract, and luminal-associated microbiota, which is found in the lumen of the gastrointestinal tract.
The term "bifidobacteria" means a member of the Bifidobacterium genus commonly found in the human gastro-intestinal tract. Examples of bifidobacteria are Bifidobacterium longum, Bifidobacterium bifidum, and the members of the phylogenetic Bifidobacterium adolescentis group. In non-infant humans, bifidobacteria preferably include members of the phylogenetic Bifidobacterium adolescentis group, for example Bifidobacterium pseudocatenulatum and/or Bifidobacterium adolescentis.
The term "Bifidobacterium of the Bifidobacterium adolescentis phylogenetic group" means a bacterium selected from a group consisting of Bifidobacterium adolescentis, Bifidobacterium angulatum , Bifidobacterium catenulatum, Bifidobacterium pseudocatenulatum ,
Bifidobacterium kashiwanohense, Bifidobacterium dentum and Bifidobacterium stercoris (Duranti et al. Appl. Environ. Microbiol. 79 , 336 (2013), Bottacini et al. Microbial Cell Fact. 1 3 : S4 (2014)) .
The term "synthetic composition" designates a composition which is artificially prepared and preferably means a composition containing at least one compound that is produced ex vivo chemically and/or biologically, e.g. by means of chemical reaction, enzymatic reaction or recombinantly. In some embodiments a synthetic composition of the invention may be, but preferably is not, identical with a naturally occurring composition. The synthetic composition of the invention typically comprises one or more compounds, advantageously HMOs, that are capable of preferentially increasing the abundance of bifidobacteria, in particular
Bifidobacterium of the following species : Bifidobacterium longum, Bifidobacterium bifidum, and/or members of the phylogenetic Bifidobacterium adolescentis group. In some
embodiments, the synthetic composition may comprise one or more compounds or components other than HMOs that may have an effect on bifidobacteria of a human subject microbiota in vivo, e.g . non-digestible oligosaccharides or prebiotics. Also in some
embodiments, the synthetic compositions may comprise one or more nutritionally or pharmaceutically active components which do not affect adversely the efficacy of the above mentioned compounds. Some non-limiting embodiments of a synthetic composition of the invention are also described below. The synthetic composition can take any suitable form. For example, the composition can be in the form of a nutritional composition which contains other macronutrients such as proteins, lipids or other carbohydrates. The synthetic composition can also be a pharmaceutical composition.
The term "intestinal permeability" preferably means the permeability of the intestinal mucosa of a patient, permitting the absorption of vital nutrients from the gut lumen while presenting a barrier against the passage of pathogenic substances into the patient's body.
The term "endotoxemia" preferably means the presence of endotoxins, such as gut microbiota-derived lipopolysaccharides (LPS) in the blood of a patient.
The term "low-grade inflammation" preferably means an immune system response of a patient characterized by altered levels of pro-inflammatory and anti-inflammatory cytokines as well as numerous other markers of immune system activity in response to an inj urious stimuli. The term "glycemic index" or "GI" is defined as the incremental area under the two-hour blood glucose response curve (AUC) following a 12-hour fast and ingestion of a food with a certain quantity of available carbohydrate (usually 50 g) . The AUC of the test food is divided by the AUC of the standard (glucose, the standard, has a GI of 100) and multiplied by 100. The average GI value is calculated from data collected in 10 human subjects. Both the standard and test food must contain an equal amount of available carbohydrate. The result gives a relative ranking for each tested food. Tables reporting commonly accepted GI values for a variety of foods are available including the international GI database maintained by the University of Sydney, and available on the internet at: www.glycemicindex.com.
The term "initial period" of treatment with an HMO or an HMO mixture is about 14 days from the start of the treatment; "additional period" of treatment means 1 or more days following the initial period of treatment.
The term "about" in the present context means up to 2.5 % deviation from the corresponded value.
The term "preferably" is used herein to indicate the best mode of invention, but to limit the scope of invention.
The HMOs can be isolated or enriched by well-known processes from milk(s) secreted by mammals including, but not limited to human, bovine, ovine, porcine, or caprine species. The HMOs can also be produced by well-known processes using microbial fermentation, enzymatic processes, chemical synthesis, or combinations of these technologies. As examples, using chemistry LNnT can be made as described in WO 2011/100980 and WO 2013/044928, LNT can be synthesized as described in WO 2012/155916 and WO 2013/044928, a mixture of LNT and LNnT ca n be made as described in WO 2013/091660, 2'-FL can be made as described in WO 2010/ 115934 and WO 2010/115935, 3-FL can be made as described in WO
2013/139344, 6'-SL and salts thereof can be made as described in WO 2010/100979, sialylated oligosaccharides can be made as described in WO 2012/113404 and mixtures of human milk oligosaccharides can be made as described in WO 2012/113405. As examples of enzymatic production, sialylated oligosaccharides can be made as described in WO
2012/007588, fucosylated oligosaccharides can be made as described in WO 2012/127410, and advantageously diversified blends of human milk oligosaccharides can be made as described in WO 2012/156897 and WO 2012/156898. With regard to biotechnological methods, WO 01/04341 and WO 2007/ 101862 describe how to make core human milk oligosaccharides optionally substituted by fucose or sialic acid using genetically modified £ coli.
Embodiments of the invention : regulating satiety and/or reducing propensity to obesity
It has now been surprisingly found that administration of human milk oligosaccharides (HMOs) to patient having a metabolic disorder, for example obesity, obesity induced prediabetes and obesity induced type 2 diabetes, preferentially increases the abundance of bifidobacteria in the gastro-intestinal tract and changes appetite sensation. The increased abundance of bifidobacteria leads to production of SCFAs through the fermentation of HMOs. These metabolites contribute to increasing production of GLP1, which is associated with the feeling of satiety (Chakraborti, World J. Gastrointest. Pathophysiol. 6 , 110 (2015)) . Thus, it has been discovered that HMOs can, by enteral ingestion, increase the production of GLP1, possibly through modulation of the intestinal microbiota in a human, in particular a non-infant human. As an outcome, a more beneficial intestinal microbial community can be shaped and maintained, and the regulation of satiety can be improved, leading to lower food intake. This can result in reduced propensity to obesity.
Accordingly, this invention, in one aspect, relates to one or more human milk
oligosaccharides for use in, a synthetic composition comprising an effective amount of one or more human milk oligosaccharides for use in, or a method for
- regulating satiety, and/or
- reducing propensity to obesity
in a human having a metabolic disorder.
In a preferred embodiment, a mixture of 2'-FL and LNnT may contain the amount of 2'- FU LNnT form about 1.5 : 1 to about 4: 1 by weight.
An HMO for use in
- regulating satiety in a human having a metabolic disorder, and/or
- reducing propensity to obesity in a human having a metabolic disorder,
may be a single HMO or a mixture of any HMOs suitable for the purpose of the invention. Preferably, the HMO is a fucosylated or a non-fucosylated neutral HMO. More preferably, the invention relates to a mixture of HMOs, the mixture comprising at least a first HMO and at least a second HMO, wherein the first HMO is a fucosylated neutral HMO and the second HMO is a non-fucosylated neutral HMO. Particularly, the mixture of HMOs may contain a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP- III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH-III, FLNH-I, FLNH-II, FLNnH, FpLNH-I and F-pLNnH II, and a non-fucosylated HMO selected from the list consisting of LNT, LNnT, LNH, LNnH, pLNH and pLNnH . Preferably, the mixture of HMOs contains a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL and DFL, and a non-fucosylated HMO selected from the list consisting of LNT and LNnT; advantageously the mixture comprises 2'-FL and LNnT and/or LNT. In some embodiments, the mixture of HMOs essentially consists of two neutral HMOs, e.g. a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP-III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH-III, FLNH-I, FLNH-II, FLNnH, FpLNH-I and F- pLNnH II, and a non-fucosylated HMO selected from the list consisting of LNT, LNnT, LNH, LNnH, pLNH and pLNnH . Preferably, the mixture essentially consists of a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL and DFL, and a non-fucosylated HMO selected from the list consisting of LNT and LNnT; in one preferred embodiment the mixture essentially consists of 2'-FL and LNnT, in another preferred embodiment the mixture essentially consists of 2'-FL and LNT.
In a preferred embodiment, a mixture of 2'-FL and LNnT may contain the amount of 2'- FL: LNnT form about 1.5: 1 to about 4: 1 by weight.
The synthetic composition for use in
- regulating satiety in a human having a metabolic disorder, and/or
- reducing propensity to obesity in a human having a metabolic disorder,
may comprise a single HMO or a mixture of any HMOs suitable for the purpose of the invention as disclosed above.
The synthetic composition can take any suitable form. For example, the composition can be in the form of a nutritional composition which contains other macronutrients such as proteins, lipids or other carbohydrates (see below). The synthetic composition can also be a
pharmaceutical composition. The synthetic compositions comprise an effective amount one or more HMOs described above.
In one embodiment, the synthetic composition may be a pharmaceutical composition. The synthetic composition comprises an effective amount one or more HMOs described above. The pharmaceutical composition can contain a pharmaceutically acceptable carrier, e.g.
phosphate buffered saline solution, mixtures of ethanol in water, water and emulsions such as an oil/water or water/oil emulsion, as well as various wetting agents or excipients. The pharmaceutical composition can also contain other materials that do not produce an adverse, allergic or otherwise unwanted reaction when administered to humans. The carriers and other materials can include solvents, dispersants, coatings, absorption promoting agents, controlled release agents, and one or more inert excipients, such as starches, polyols, granulating agents, microcrystalline cellulose, diluents, lubricants, binders, and disintegrating agents. If desired, tablet dosages of the a nti- infective compositions can be coated by standard aqueous or non-aqueous techniques.
The pharmaceutical compositions can be administered orally, e.g. as a tablet, capsule, or pellet containing a predetermined amount, or as a powder or granules containing a predetermined concentration or a gel, paste, solution, suspension, emulsion, syrup, bolus, electuary, or slurry, in an aqueous or non-aqueous liquid, containing a predetermined concentration. Orally administered compositions can include binders, lubricants, inert diluents, flavouring agents, and humectants. Orally administered compositions such as tablets can optionally be coated and can be formulated so as to provide sustained, delayed or controlled release of the mixture therein.
The pharmaceutical compositions can also be administered by rectal suppository, aerosol tube, naso-gastric tube or direct infusion into the GI tract or stomach. The pharmaceutical compositions can also include therapeutic agents such as antiviral agents, antibiotics, probiotics, analgesics, and anti-inflammatory agents. The proper dosage of these compositions for a human can be determined in a conventional manner, based upon factors such as severity of the lactose intolerance, immune status, body weight and age. In one embodiment, a synthetic composition comprises an effective amount of a mixture of 2'-FL and LNnT, preferably the mass ratio between 2'-FL and LNnT in the composition is in the range from 5: 1 to 1 : 1.
Furthermore, the invention relates to the following methods:
- a method for regulating satiety in in a human having a metabolic disorder, the
method comprising administering to the human an effective amount of one or more human milk oligosaccharides; and/or
- a method for reducing propensity to obesity in a human having a metabolic disorder, the method comprising administering to the human an effective amount of one or more human milk oligosaccharides.
The HMOs suitable for the purpose of the method are disclosed above.
For increasing the levels of the gut hormones GLP1 and GLP2 in a person, the amount of human milk oligosaccharide(s) required to be administered to the person will vary depending upon factors such as the risk and condition severity, the age of the person, the form of the composition, and other medications being administered to the person. However, the required amount can be readily set by a medical practitioner and would generally be in the range from about 10 mg to about 20 g per day, in certain embodiments from about 10 mg to about 15 g per day, from about 100 mg to about 10 g per day, in certain embodiments from about 500 mg to about 10 g per day, in certain embodiments from about 1 g to about 7.5 g per day. An appropriate dose can be determined based on several factors, including, for example, the body weight and/or condition of the patient being treated, the severity of the condition, being treated, other ailments and/or diseases of the person, the incidence and/or severity of side effects and the manner of administration. Appropriate dose ranges can be determined by methods known to those skilled in the art. During an initial treatment phase, the dosing can be higher (for example 200 mg to 20 g per day, preferably 500 mg to 15 g per day, more preferably 1 g to 10 g per day, in certain embodiments 2.5 g to 7.5 g per day). During a maintenance phase, the dosing can be reduced (for example, 10 mg to 10 g per day, preferably 100 mg to 7.5 g per day, more preferably 500 mg to 5 g per day, in certain embodiments 1 g to 2.5 g per day).
Preferably, one or more HMOs or a composition comprising or essentially consisting thereof is administered to a human in need enteral, e.g. orally.
A synthetic composition of this invention can be co-administered to a patient who is also receiving a standard-of-care medication for obesity or diabetes. Embodiments of the invention : stabilising or reducing insulin resistance
It has also been surprisingly found that human milk oligosaccharides, advantageously 2'-FL, LNT and LNnT, not only modulate inflammation and microbiota in the GI tract, but also stabilise or reduce insulin resistance. Further, the abundance of members of the
Bifidobacterium adolescentis phylogenetic group is increased, in particular B. adolescentis and/or B. pseudocatenulatum . In particular, a combination of 2'-FL and LNnT preferentially increases the abundance of B. pseudocatenulatum . This can result in lower chronic inflammation, improved insulin sensitivity and reduced insulin resistance. Obese and pre- diabetic patients can be stabilised and the progression to diabetes slowed, stopped or reversed . Diabetic patients can be stabilised or at least the progression to diabetes with complications slowed .
Accordingly, this invention, in another embodiments, relates to one or more human milk oligosaccharides (HMOs) for use in, or a synthetic composition comprising an effective amount of one or more human milk oligosaccharides for use in, or a method for stabilising or reducing insulin resistance in a human individual having an obesity-related metabolic disorder, for example obesity, obesity induced pre-diabetes and obesity induced type 2 diabetes.
One embodiment of the invention relates to an HMO for use in stabilising or reducing insulin resistance in a human individual having an obesity-related metabolic disorder.
Another embodiment of the invention relates to a synthetic composition comprising an HMO for use in stabilising or reducing insulin resistance in a human individual having an obesity- related metabolic disorder.
Still another embodiment of the invention relates to a method for stabilising or reducing insulin resistance in a human individual having an obesity-related metabolic disorder, the method comprising enterally administering to the patient an effective amount of one or more human milk oligosaccharides.
The HMO in any of the above embodiments concerning stabilising or reducing insulin resistance in a human may be a single HMO or a mixture of any HMOs suitable for the purpose of the invention. Preferably, the HMO is a fucosylated or a non-fucosylated neutral HMO. More preferably, this particular aspect of the invention relates to a mixture of HMOs, the mixture comprising at least a first HMO and at least a second HMO, wherein the first HMO is a fucosylated neutral HMO and the second HMO is a non-fucosylated neutral HMO.
Particularly, the mixture of HMOs may contain a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP-III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH- III, FLNH-I, FLNH-II, FLNnH, FpLNH-I and F-pLNnH II, and a non-fucosylated HMO selected from the list consisting of LNT, LNnT, LNH, LNnH, pLNH and pLNnH . Preferably, the mixture of HMOs contains a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL and DFL, and a non-fucosylated HMO selected from the list consisting of LNT and LNnT; advantageously the mixture comprises 2'-FL and LNnT and/or LNT. In some embodiments, the mixture of HMOs essentially consists of two neutral HMOs, e.g. a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP-III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH-III, FLNH-I, FLNH-II, FLNnH, FpLNH-I and F-pLNnH II, and a non- fucosylated HMO selected from the list consisting of LNT, LNnT, LNH, LNnH, pLNH and pLNnH. Preferably, the mixture essentially consists of a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL and DFL, and a non-fucosylated HMO selected from the list consisting of LNT and LNnT; in one preferred embodiment the mixture essentially consists of 2'-FL and LNnT, in another preferred embodiment the mixture essentially consists of 2'-FL and LNT.
The invention relates in different embodiments to single HMOs as substantially pure single compounds, i.e. an HMO which grade of purity satisfies the demand of a medical or food authority for marketing, or mixtures of substantially pure HMOs, or artificial compositions comprising one or more HMOs.
Preferably, in this aspect of the invention, the human milk oligosaccharides include one or more fucosylated HMOs, such as 2'-FL, and one or more core HMOs, such as LNT and LNnT. More preferably the composition comprises a mix of 2'-FL and LNnT; for example in a mass ratio of 5: 1 to 1 : 1; more preferably 4: 1 to 2: 1.
In this aspect of the invention, the human individual can be an obese paediatric or adult patient, preferably a prepubescent child.
In this aspect of the invention, the HMOs may be administered to the patient as a daily dose of about 1 g to about 15 g such as from about 3 g to about 10 g. The patient can be administered a higher amount, preferably 5 g to 10 g per day, of the HMOs for an initial treatment period, followed by a lower amount, preferably 1 g to 5 g per day, for a
maintenance period. The initial treatment period can be 1 to 8 weeks. The maintenance period is at least 1 month.
The synthetic composition can take any suitable form. For example, the composition can be in the form of a nutritional composition which contains other macronutrients such as proteins, lipids or other carbohydrates (see below). The synthetic composition can also be a
pharmaceutical composition. The synthetic compositions comprise an effective amount one or more HMOs described above.
In one embodiment, the synthetic composition may be a pharmaceutical composition. The synthetic composition comprises an effective amount one or more HMOs described above. The pharmaceutical composition can contain a pharmaceutically acceptable carrier, e.g.
phosphate buffered saline solution, mixtures of ethanol in water, water and emulsions such as an oil/water or water/oil emulsion, as well as various wetting agents or excipients. The pharmaceutical composition can also contain other materials that do not produce an adverse, allergic or otherwise unwanted reaction when administered to humans. The carriers and other materials can include solvents, dispersants, coatings, absorption promoting agents, controlled release agents, and one or more inert excipients, such as starches, polyols, granulating agents, microcrystalline cellulose, diluents, lubricants, binders, and disintegrating agents. If desired, tablet dosages of the a nti- infective compositions can be coated by standard aqueous or non-aqueous techniques.
The pharmaceutical compositions can be administered orally, e.g. as a tablet, capsule, or pellet containing a predetermined amount, or as a powder or granules containing a predetermined concentration or a gel, paste, solution, suspension, emulsion, syrup, bolus, electuary, or slurry, in an aqueous or non-aqueous liquid, containing a predetermined concentration. Orally administered compositions can include binders, lubricants, inert diluents, flavouring agents, and humectants. Orally administered compositions such as tablets can optionally be coated and can be formulated so as to provide sustained, delayed or controlled release of the mixture therein.
The pharmaceutical compositions can also be administered by rectal suppository, aerosol tube, naso-gastric tube or direct infusion into the GI tract or stomach.
The pharmaceutical compositions can also include therapeutic agents such as antiviral agents, antibiotics, probiotics, analgesics, and anti-inflammatory agents. The proper dosage of these compositions for a human can be determined in a conventional manner, based upon factors such as severity of the lactose intolerance, immune status, body weight and age. For stabilising or reducing insulin resistance in a patient having an obesity-related, metabolic disorder, the amount of human milk oligosaccharide(s) required to be administered to the person will vary depending upon factors such as the risk and condition severity, the age of the person, the form of the composition, and other medications being administered to the person. However, the required amount can be readily set by a medical practitioner and would generally be in the range from about 200 mg to about 20 g per day, in certain embodiments from about 1 g to about 15 g per day, from about 3 g to about 10 g per day, in certain embodiments from about 3 g to about 7.5 g per day. An appropriate dose can be determined based on several factors, including, for example, body weight and/or condition, the severity of the condition, being treated or prevented, other ailments and/or diseases, the incidence and/or severity of side effects and the manner of administration. Appropriate dose ranges may be determined by methods known to those skilled in the art.
During an initial treatment phase, the dosing can be higher or lower depending upon the need to boost bifidobacteria abundance or initial tolerance to HMOs. During a maintenance phase, the dosing can be set for chronic long term use. For example, during the initial treatment phase, the dosing can be 500 mg to 20 g per day, preferably 1 g to 15 g per day, more preferably 3 g to 10 g per day. During the maintenance phase, the dosing can be reduced to 200 mg to 10 g per day, preferably 500 mg to 7.5 g per day, more preferably 1 g to 5 g per day. A synthetic composition of this invention can be co-administered to a patient who is also receiving a standard-of-care medication for obesity or diabetes.
HMO/HMOs or synthetic composition(s) comprising said HMO/HMOs disclosed herein is(are) preferably administered to a patient in need enterally, e.g . orally.
Embodiments of the invention : reducing the risk of, preventing or treating CVD and associated co-morbidities in overweight or obese humans
In addition, it has also been surprisingly found that administration of human milk
oligosaccharides (HMOs) to an obese patient, preferentially increases the abundance of bifidobacteria in the gastro-intestinal tract, reducing cholesterol and/or hypertension, and through this reduces the risk of CVD and associated co-morbidities. Further, the abundance of members of the Bifidobacterium adolescentis phylogenetic group is increased, in particular B. adolescentis and/or B. pseudocatenulatum .
The increased abundance of bifidobacteria leads to production of SCFAs though the fermentation of HMOs and increased activity of BSH. Thus, it has been discovered that HMOs can, by oral or enteral ingestion, increase the production of SCFA and activity of BSH, possibly through modulation of the intestinal microbiota in human. As an outcome, a more beneficial intestinal microbial community can be shaped and maintained, which contributes to attenuation of hypercholesterolemia and hypertension . This can result in reduced risk of, prevention of and/or treatment of, CVD and associated co-morbidities.
Accordingly, this invention, in other embodiments, relates to a human milk oligosaccharide or a mixture of two to five human milk oligosaccharides for use in, or a synthetic composition comprising an effective amount of a human milk oligosaccharide or an effective amount of a mixture of two to five human milk oligosaccharides for use in, or a method for
- reducing the propensity of a cardiovascular disease (CVD) and/or a CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual,
- preventing development of a cardiovascular disease (CVD) and/or a CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual, and/or
- treating a cardiovascular disease (CVD) and/or a CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual.
Within this aspect of the invention, it is provided one or more HMOs selected from the group consisting of fucosylated HMOs and core HMOs, preferably of a mixture of one or more fucosylated HMOs and one or more core HMOs, for use in, or a synthetic composition comprising an effective amount of one or more HMOs selected from the group consisting of fucosylated HMOs and core HMOs, preferably of a mixture of one or more fucosylated HMOs and one or more core HMOs, for use in, or a method for increasing the abundance of bifidobacteria in a human, preferably, in an overweight or obese human having a propensity of, or diagnosed with a cardiovascular disease (CVD). The HMOs suitable for the purpose according to this aspect of the invention are disclosed below.
In this aspect of the invention, the invention relates in different embodiments to single HMOs as substantially pure single compounds, i.e. an HMO which grade of purity satisfies the demand of a medical or food authority for marketing, or mixtures of 2 to 5 such substantially pure HMOs, or artificial compositions comprising one to five HMOs. Embodiments of HMOs and compositions comprising thereof are described below.
In particular, in this aspect of the invention, different embodiments of the invention relate to HMOs for use in
- reducing the propensity of a cardiovascular disease (CVD) in a human individual, preferably in an overweight or obese human individual,
- preventing development of a cardiovascular disease (CVD) in in a human individual, preferably in an overweight or obese human individual, and/or
- treating a cardiovascular disease (CVD) in in a human individual, preferably in an overweight or obese human individual,
where the HMOs may be a single HMO or a mixture of two to five of any HMOs suitable for the purpose of the invention. Preferably, the HMO is a fucosylated or a non-fucosylated neutral HMO. More preferably, the invention relates to a mixture of HMOs, the mixture comprising at least a first HMO and at least a second HMO, wherein the first HMO is a fucosylated neutral HMO and the second HMO is a non-fucosylated neutral HMO. In other embodiments the mixture may comprise further a third, a forth and a fifth HMO. Particularly, the mixture of HMOs may contain a fucosylated HMO selected from the list consisting of 2'- FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP-III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH-III, FLNH-I, FLNH-II, FLNnH, FpLNH-I and F-pLNnH II, and a non-fucosylated HMO selected from the list consisting of LNT, LNnT, LNH, LNnH, pLNH and pLNnH. Preferably, the mixture of HMOs contains a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL and DFL, and a non-fucosylated HMO selected from the list consisting of LNT and LNnT; advantageously the mixture comprises 2'-FL and LNnT and/or LNT. In some embodiments, the mixture of HMOs essentially consists of two neutral HMOs, e.g. a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL, DFL, LNFP-I, LNFP-II, LNFP-III, LNFP-V, LNDFH-I, LNDFH-II, LNDFH- III, FLNH-I, FLNH-II, FLNnH, FpLNH-I and F-pLNnH II, and a non-fucosylated HMO selected from the list consisting of LNT, LNnT, LNH, LNnH, pLNH and pLNnH. Preferably, the mixture essentially consists of a fucosylated HMO selected from the list consisting of 2'-FL, 3-FL and DFL, and a non-fucosylated HMO selected from the list consisting of LNT and LNnT; in one preferred embodiment the mixture essentially consists of 2'-FL and LNnT, in another preferred embodiment the mixture essentially consists of 2'-FL and LNT. In a preferred embodiment of this aspect of the invention, a mixture of 2'-FL and LNnT may contain the amount of 2'-FL: LNnT form about 1.5 : 1 to about 4: 1 by weight.
In other embodiments of this aspect of the invention, the invention relates to a synthetic composition for use in
- reducing the propensity of a cardiovascular disease (CVD) in a human individual, preferably in an overweight or obese human individual,
- preventing development of a cardiovascular disease (CVD) in a human individual, preferably in an overweight or obese human individual, and/or
- treating a cardiovascular disease (CVD) in a human individual, preferably in an overweight or obese human individual,
which may comprise a single HMO or a mixture of two to five of any HMOs suitable for the purpose of the invention as disclosed above.
The synthetic composition can take any suitable form. For example, the composition can be in the form of a nutritional composition which contains other macronutrients such as proteins, lipids or other carbohydrates (see below) .
The synthetic composition can also be a pharmaceutical composition . A pharmaceutical composition contains an effective amount of HMO or an effective amount of mixture of two to five HMOs, wherein the HMOs are selected from any of described above. The term "effective amount" in the present content means an amount of a single HMO, or a combination of different HMOs that is capable of increasing the abundance of bifidobacteria in the gastrointestinal tract of a human individual of the invention, preferably, relative abundance of members of the Bifidobacterium adolescentis phylogenetic group in particular B. adolescentis and/or B. pseudocatenulatum .
The pharmaceutical composition can further contain a pharmaceutically acceptable carrier, e.g. phosphate buffered saline solution, mixtures of ethanol in water, water and emulsions such as an oil/water or water/oil emulsion, as well as various wetting agents or excipients. The pharmaceutical composition can also contain other materials that do not produce an adverse, allergic or otherwise unwanted reaction when administered to humans. The carriers and other materials can include solvents, dispersants, coatings, absorption promoting agents, controlled release agents, and one or more inert excipients, such as starches, polyols, granulating agents, microcrystalline cellulose, diluents, lubricants, binders, and disintegrating agents. If desired, tablet dosages of the a nti- infective compositions can be coated by standard aqueous or non-aqueous techniques.
The pharmaceutical compositions can be administered orally, e.g. as a tablet, capsule, or pellet containing a predetermined amount, or as a powder or granules containing a predetermined concentration or a gel, paste, solution, suspension, emulsion, syrup, bolus, electuary, or slurry, in an aqueous or non-aqueous liquid, containing a predetermined concentration. Orally administered compositions can include binders, lubricants, inert diluents, flavouring agents, and humectants. Orally administered compositions such as tablets can optionally be coated and can be formulated so as to provide sustained, delayed or controlled release of the mixture therein.
The pharmaceutical compositions can also be administered by rectal suppository, aerosol tube, naso-gastric tube or direct infusion into the GI tract or stomach.
The pharmaceutical compositions can also include therapeutic agents most commonly prescribed for heart disease such as:
ACE Inhibitors: ACE inhibitors are a type of medication that dilates (widens) arteries to lower blood pressure and make it easier for the heart to pump blood. They also block some of the harmful actions of the endocrine system that may occur with heart failure;
Aldosterone Inhibitor: Eplerenone (Inspra) and spironolactone (Aldoctone) and eplerenone are potassium-sparing diuretics. They can be prescribed to reduce the swelling and water build-up caused by heart failure. Diuretics cause the kidneys to send unneeded water and salt from the tissues and blood into the urine;
They may improve heart failure symptoms that are still present despite use of other treatments. These drugs protect the heart by blocking a chemical (aldosterone) in the body that causes salt and fluid build-up. This medication is used to treat patients with certain types of severe heart failure;
Angiotensin II Receptor Blocker (ARBs) : ARBs are used to decrease blood pressure in people with heart failure. ARBs decrease certain chemicals that narrow the blood vessels so blood can flow more easily through your body. They also decrease certain chemicals that cause salt and fluid build-up in the body;
Beta-Blockers: Beta-blockers block the effects of adrenaline (epinephrine) and thereby improve the heart's ability to perform. They also decrease the production of harmful substances produced by the body in response to heart failure. They cause the heart to beat more slowly and with less force, lowering blood pressure;
Calcium Channel Blockers: Calcium channel blockers are prescribed to treat angina (chest pain) and high blood pressure. Calcium channel blockers affect the movement of calcium in the cells of the heart and blood vessels. As a result, the drugs relax blood vessels and increase the supply of blood and oxygen to the heart, while reducing its workload;
Cholesterol -Lowering Drugs: Cholesterol helps your body build new cells, insulate nerves, and produce hormones. But inflammation may lead to cholesterol build-up in the walls of arteries, increasing the risk of heart attack and stroke;
Digoxin : Digoxin helps an injured or weakened heart to work more efficiently and to send blood through the body. It strengthens the force of the heart muscle's contractions and may improve blood circulation; Diuretics: Diuretics, commonly known as "water pills," cause the kidneys to get rid of unneeded water and salt from the tissues and bloodstream into the urine. Getting rid of excess fluid makes it easier for your heart to pump. Diuretics are used to treat high blood pressure and reduce the swelling and water build-up caused by various medical problems, including heart failure;
Inotropic Therapy: Inotropic therapy is used to stimulate an injured or weakened heart to pump harder to send blood through the body. It helps the force of the heart muscle's contractions and relaxes constricted blood vessels so blood can flow more smoothly. Inotropic therapy may also speed up the heart's rhythm;
Potassium or Magnesium : Potassium and magnesium are minerals that can be lost because of increased urination when taking diuretics. Low levels in the body can be associated with abnormal heart rhythms. Some patients take them as supplements as directed by their doctor.
Vasodilators: Vasodilators are used to treat heart failure and control high blood pressure by relaxing the blood vessels so blood can flow more easily through the body. Vasodilators are prescribed for patients who cannot take ACE inhibitors.
Warfarin : Warfarin is an anticoagulant medication. "Anti" means "against," and "coagulant" means "causing blood clotting." Therefore, warfarin helps prevent clots from forming in the blood.
The pharmaceutical composition may also contain other compounds such as antibiotics, probiotics, analgesics, and anti-inflammatory agents.
The proper dosage of these compositions for a human can be determined in a conventional manner, based upon factors such as severity of conditions of the human individual, e.g. the individual's blood pressure, immune status, body weight, age, etc.
In other embodiments of this aspect of the invention, the invention relates to a method for
- reducing the propensity of a cardiovascular disease (CVD), or an CVD-associated
pathologic condition or disease, in a human, preferably, wherein said human is overweight or obese;
- preventing development of a cardiovascular disease (CVD), or an CVD-associated pathologic condition or disease, in a human, preferably wherein said human is overweight or obese;
- treating a cardiovascular disease (CVD), or an CVD-associated pathologic condition or disease, in a human, preferably, wherein said human is overweight or obese; and/or
- increasing the abundance of bifidobacteria in a human having an CVD disease, or an CVD-associated pathologic condition or disease, preferably, wherein said human is overweight or obese, said the method comprising administering to the patient, preferably daily, at least 2 g of a human milk oligosaccharide (HMO) selected from the group consisting of fucosylated HMOs and core HMOs, more preferably at least 2 g of a mixture of two to five HMOs consisting of one or more fucosylated HMOs and one or more core HMOs. The HMOs suitable for the purpose of the method according to this aspect of the invention are disclosed above.
The invention contemplates both prophylactic and therapeutic methods of treatment depending on different embodiments. The term "therapeutic method" means a method co mprising treatment of disease or medical disorder by remedial agents and/or, e.g .
administering an HMO(s) or a composition of the invention to a CVD patient of the invention to cure the CVD or the associated pathological condition or disease. The term "prophylactic method" means a method comprising a measure taken to fend off a disease or another unwanted consequence of the disease, e.g . administering an HMO or a composition of the invention to a human of the invention to reduce the propensity of or prevent development of CVD or the associated pathological condition or disease in the human.
Preferably, an HMO of the invention is administered to a human in need enteral, e.g. orally. Preferably, invention relates to a method increasing the abundance of a bacterium of the B. adolescentis phylogenetic group, especially Bifidobacterium adolescentis and/or B.
pseudocatenulatum .
In any of the methods, one or more HMOs, preferably, one to five HMOs, may be
administered as substantially pure compounds (i.e. neat) or diluted, e.g . in form of a solution, power or syrup, or in the form of a synthetic composition, nutritional or
pharmaceutical composition, as any of the described above, in one or more unit dosage forms, preferably in a single unit dosage form.
Preferably, the HMOs are, or the synthetic, nutritional or pharmaceutical, composition contains, 2'-FL and LNnT, preferably the 2'-FL: LNnT ratio is about 1.5 : 1 to about 4: 1 by weight.
The dosage of one or more fucosylated HMOs and one or more core HMOs per administration may vary from about 2 g to about 10 g, preferably from about 3.5 g to about 7.5 g. Typically, the HMOs are administered in a single dosage unit containing from about 2 g to about 10 g, preferably from about 3.5 g to about 7.5 g of one of more fucosylated HMOs and one or more core HMOs. The patient may also additionally receive a dose of one or more species of probiotic bacteria, e.g . bifidobacteria.
Typically, the patient is administered a daily dose of at least 2 g of the mixture of one or more fucosylated HMOs and one or more core HMOs for at least 14 days, preferably, for more than 14 days. Nutritional compositions
A nutritional composition can contain sources of protein, lipids and/or digestible
carbohydrates and can be in solid, powdered or liquid forms. The composition can be designed to be the sole source of nutrition or a nutritional supplement.
Suitable protein sources include intact, hydrolysed, and partially hydrolysed protein, which can be derived from any suitable source such as milk (e.g., casein, whey), animal (e.g. meat, fish), cereal (e.g. rice, corn), and vegetable (e.g. soy, potato, pea), insect (e.g. locust) and combinations of these sources. Examples of the source of protein include whey protein concentrates, whey protein isolates, whey protein hydrolysates, acid caseins, sodium casemates, calcium casemates, potassium casemates, casein hydrolysates, milk protein concentrates, milk protein isolates, milk protein hydrolysates, non-fat dry milk, condensed skim milk, soy protein concentrates, soy protein isolates, soy protein hydrolysates, pea protein concentrates, pea protein isolates, pea protein hydrolysates, collagen proteins, and combinations of these sources.
The amount of protein is preferably sufficient to provide about 5 to about 30 % of the energy of the nutritional composition; for example about 10 % to about 25 % of the energy. Within these ranges, the amount of protein can vary depending upon the nutritional needs of the intended individual.
The nutritional compositions can also include free amino acids such as tryptophan, glutamine, tyrosine, methionine, cysteine, taurine, arginine, carnitine, threonine, serine and proline and combinations of these amino acids. Threonine, serine and proline are important amino acids for the production of mucin which aids gut barrier function.
Any suitable source of other carbohydrates can be used. Examples include maltodextrin, hydrolysed or modified starch or corn starch, glucose polymers, corn syrup, corn syrup solids, rice-derived carbohydrates, sucrose, glucose, fructose, lactose, high fructose corn syrup, honey, sugar alcohols (e.g., maltitol, erythritol, sorbitol, etc.), isomaltulose, sucromalt, pullulan, potato starch, slowly-digested carbohydrates, dietary fibres such as oat fibre, soy fibre, gum arabic, sodium carboxymethylcellulose, methylcellulose, guar gum, gellan gum, locust bean gum, konjac flour, hydroxypropyl methylcellulose, tragacanth gum, karaya gum, gum acacia, chitosan, arabinogalactans, glucomannan, xanthan gum, alginate, pectin, low and high methoxy pectin, cereal beta-glucans (i.e., oat beta-glucan, barley beta- glucan), carrageenan and psyllium, Fibersol(TM), other resistant starches, and combinations of these carbohydrate.
Preferably the carbohydrate source includes low glycemic index carbohydrates having a GI score of 55 or below. Examples of low glycemic index carbohydrates include sucromalt,
Fibersol(TM) (inulin), maltodextrins having a dextrose equivalence (DE) of less than 15, rice syrup having a dextrose equivalence of less than 15, fructooligosaccharides, resistant starches, starches, fruit sourced fibres, vegetable sourced fibres, whole grains, beta-glucans, soy fibres, oat fibres, locust bean gum, konjac flour, hydroxy propyl methylcellulose, gum acacia, chitosan, arabinogalactans, xanthan gum, alginate, low and high methoxy pectin, carrageenan, psyllium, isomaltulose, glycerine and sugar alcohols.
The nutritional compositions can include carbohydrates in an amount sufficient to provide about 30 to about 70 % of the energy of the composition, for example about 35 to about 65 % of the energy. Within these parameters, the amount of carbohydrate can vary widely.
Suitable lipid sources include coconut oil, fractionated coconut oil, soy oil, corn oil, olive oil, safflower oil, high oleic safflower oil, medium chain triglycerides, sunflower oil, high oleic sunflower oil, palm and palm kernel oils, palm olein, canola oil, marine oils, cottonseed oils and combinations of these oils. Fractionated coconut oils are a suitable source of medium chain triglycerides. The lipids can contain polyunsaturated fatty acids such as n-3 LC-PUFA. The n-3 LC-PUFA can be a C20 or a C22 n-3 fatty acid . Preferably the n-3 LC-PUFA is docosahexanoic acid (DHA, C22 : 6, n-3) . The source of LC-PUFA can be, for example, egg lipids, fungal oil, low EPA fish oil or algal oil.
The nutritional compositions can include lipids in an amount sufficient to provide about 10 to about 50 % of energy of the nutritional composition, for example about 15 to about 40 % of the energy.
The nutritional composition preferably also includes vitamins and minerals. If the nutritional composition is intended to be a sole source of nutrition, it preferably includes a complete vitamin and mineral profile. Examples of vitamins include vitamins A, B-complex (such as Bl, B2, B6 and B12), C, D, E and K, niacin and acid vitamins such as pantothenic acid, folic acid and biotin. Examples of minerals include calcium, iron, zinc, magnesium, iodine, copper, phosphorus, manganese, potassium, chromium, molybdenum, selenium, nickel, tin, silicon, vanadium and boron .
The nutritional composition can also include a carotenoid such as lutein, lycopene, zeaxanthin, and beta -carotene. The total amount of carotenoid included can vary from about 0.001 pg/ml to about 10 pg/ml. Lutein can be included in an amount of from about 0.001 pg/ml to about 10 pg/ml, preferably from about 0.044 pg/ml to about 5 pg/ml of lutein. Lycopene can be included in an amount from about 0.001 pg/ml to about 10 pg/ml, preferably about 0.0185 pg/ml to about 5 pg/ml of lycopene. Beta-carotene can comprise from about 0.001 pg/ml to about 10 pg/ml, for example about 0.034 pg/ml to about 5 pg/ml of beta-carotene. The nutritional composition can also include a source of anthocyanins. This can be in the form of a fruit or a fruit extract. Particularly useful fruits and fruit extracts include plum/prune, apple, pear, strawberry, blueberry, raspberry, cherry, and their combinations.
The nutritional composition can also contain various other conventional ingredients such as preservatives, emulsifying agents, thickening agents, buffers, fibres and prebiotics (e.g . fructooligosaccharides, galactooligosaccharides), probiotics (e.g . B. animalis subsp. lactis BB- 12, B. lactis HN019, B. lactis Bi07, B. infant is ATCC 15697, L. rhamnosus GG, L. rhamnosus HNOOI, L. acidophilus LA-5, L. acidophilus NCFM, L. fermentum CECT5716, B. longum BB536, B. longum AH 1205, B. longum AH 1206, B. breve M- 16V, L. reuteri ATCC 55730, L. reuteri ATCC PTA-6485, L. reuteri DSM 17938), antioxidant/anti-inflammatory compounds including tocopherols, caroteinoids, ascorbate/vitamin C, ascorbyl palmitate, polyphenols, glutathione, and superoxide dismutase (melon), other bioactive factors (e.g . growth hormones, cytokines, TFG-β), colorants, flavours, and stabilisers, lubricants, and so forth.
The nutritional composition can be in the form of a food, soluble powder, a liquid concentrate, or a ready-to-use formulation. The composition can be eaten, drunk or can be fed via a nasogastric. Various flavours, fibres and other additives can also be present.
The nutritional compositions can be prepared by any commonly used manufacturing techniques for preparing nutritional compositions in solid or liquid form. For example, the composition can be prepared by combining various feed solutions. A protein-in-fat feed solution can be prepared by heating and mixing the lipid source and then adding an emulsifier (e.g . lecithin), fat soluble vitamins, and at least a portion of the protein source while heating and stirring. A carbohydrate feed solution is then prepared by adding minerals, trace and ultra trace minerals, thickening or suspending agents to water while heating and stirring. The resulting solution is held for 10 minutes with continued heat and agitation before adding carbohydrates (e.g. the HMOs and digestible carbohydrate sources) . The resulting feed solutions are then blended together while heating and agitating and the pH adj usted to 6.6-7.0, after which the composition is subjected to high-temperature short-time processing during which the composition is heat treated, emulsified and homogenized, and then allowed to cool. Water soluble vitamins and ascorbic acid are added, the pH is adj usted to the desired range if necessary, flavours are added, and water is added to achieve the desired total solid level.
For a liquid product, the resulting solution can then be aseptically packed to form an aseptically packaged nutritional composition. In this form, the nutritional composition can be in ready-to-feed or concentrated liquid form. Alternatively, the composition can be spray- dried and processed and packaged as a reconstitutable powder.
The nutritional composition can also be in the form of a food such as a nutritional bar, a yoghurt, etc. These forms can be produced using standard technologies and processes.
When the nutritional product is a ready-to-feed nutritional liquid, the total concentration of HMOs in the liquid, by weight of the liquid, is from about 0.0001 % to about 2.0 %, including from about 0.001 % to about 1.5 %, including from about 0.01 % to about 1.0 %. When the nutritional product is a concentrated nutritional liquid, the total concentration of HMSs/HMOs in the liquid, by weight of the liquid, is from about 0.0002 % to about 4.0 %, including from about 0.002 % to about 3.0 %, including from about 0.02 % to about 2.0 %. Unit dosage forms
The synthetic composition of this invention can also be in a unit dosage form such as a capsule, tablet or sachet. For example, the composition can be in a tablet form comprising the human milk oligosaccharides, and one or more additional components to aid formulation and administration, such as diluents, excipients, antioxidants, lubricants, colorants, binders, disintegrants, and the like.
Suitable diluents, excipients, lubricants, colorants, binders, and disintegrants include polyethylene, polyvinyl chloride, ethyl cellulose, acrylate polymers and their copolymers, hydroxyethyl-cellulose, hydroxypropylmethyl-cellulose (HPMC), sodium
carboxymethylcellulose, polyhydroxyethyl methacrylate (PHEMA), polyvinyl alcohol (PVA), polyvinyl pyrrolidone (PVP), polyethylene oxide (PEO), or polyacrylamide (PA), carrageenan, sodium alginate, polycarbophil, polyacrylic acid, tragacanth, methyl cellulose, pectin, natural gums, xanthan gum, guar gum, karaya gum, hypromellose, magnesium stearate,
microcrystalline cellulose, and colloidal silicon dioxide. Suitable antioxidants are vitamin A, carotenoids, vitamin C, vitamin E, selenium, flavonoids, polyphenols, lycopene, lutein, lignan, coenzyme Q10 ("CoQIO") and glutathione.
The unit dosage forms, especially those in sachet form, can also include various nutrients including macronutrients.
The unit dosage forms can be administered orally, e.g. as a tablet, capsule, or pellet containing a predetermined amount, or as a powder or granules containing a predetermined concentration or a gel, paste, solution, suspension, emulsion, syrup, bolus, electuary, or slurry, in an aqueous or non-aqueous liquid, containing a predetermined concentration. Orally administered compositions can include binders, lubricants, inert diluents, flavouring agents, and humectants. Orally administered compositions such as tablets can optionally be coated and can be formulated so as to provide sustained, delayed or controlled release of the mixture therein.
EXAM PLES
Examples are now described to further illustrate the invention :
Example 1 - Treating high fat diet induced obesity and diabetes
10-week-old C57BL/6J mice (100 mice) are housed in groups of five mice per cage, with free to water. The mice are divided into 10 groups of 10 mice, one control group and 9 treatment groups. All of the mice are fed a high-fat (HF) diet (60 % fat and 20 % carbohydrates
[kcal/100 g], or an HF diet supplemented with HMO (20 g/kg of diet) for 8 weeks. Food and water intake are recorded twice a week. The 9 treatment groups are each administered one of the following : a) 2'-FL, b) 3-FL, c) 3'-SL, d) 6'-SL, e) LNT, f) LNnT, g) LNFP-I, h) DSLNT and i) a combination of these saccharides. The control group is administered the HF diet only. Intraperitoneal or oral glucose tolerance tests are performed as follows : 6-h-fasted mice are injected with glucose into the peritoneal cavity ( 1 g/kg glucose, 20 % glucose solution) or by gavage (3 g/kg glucose, 66 % glucose solution) . Blood glucose is determined with a glucose meter (Roche Diagnostics) on 3.5 μΙ blood collected from the tip of the tail vein. A total of 20 μΙ blood is sampled 30 min before and 15 or 30 min after the glucose load to assess plasma insulin concentration.
To assess intestinal permeability in vivo, the intestinal permeability of 4000 Da fluorescent dextran-FITC (DX-4000-FITC) is measured . M ice are fasted for 6 h before given DX-44-FITC by gavage (500 mg/kg body weight, 125 mg/ml) . After 1 h and 4 h, 120 ml of blood is collected from the tip of the tail vein. The blood is centrifuged at 4 °C, 12 000 g for 3 min. Plasma is diluted in an equal volume of PBS (pH 7.4) and analysed for DX- 4000-FITC concentration with a fluorescence spectrophotometer at an excitation wavelength of 485 nm and emission wavelength of 535 nm. Standard curves are obtained by diluting FITC-dextran in non-treated plasma diluted with PBS ( 1 : 3 v/v) .
Mice are anaesthetised (ketamine/xylazine, intraperineally, 100 and 10 mg/kg, respectively) after a 5 h period of fasting, and blood samples and tissues are harvested for further analysis. Mice are killed by cervical dislocation. Liver, caecum (full and empty), muscles (vastus lateralis), and adipose tissues (mesenteric and corresponding lymph nodes, epididymal, subcutaneous and visceral) are precisely dissected and weighed and stored at - 80 °C, for further analysis.
Total and active GLP1 are measured from blood with ELISA (M illipore, Molsheim, France) .
To assess the microbiota profile, the caecal contents collected post mortem from mice are stored at -80 °C. DNA is isolated from the caecal content samples using QIAamp DNA Stool Mini Kit. The DNA concentration of extracts is measured using NanoDrop. Aliquots of 100 ng of extracted DNA are subjected to PCR using the 16S rDNA universal heteroduplex analysis (HDA) primers HDA1-GC 50-CGCCCGGGGCGCGCCCCGGGCGGG- GCGGGGGCACGGGGGGACTCCTACGGGAGGCAGCAGT-30 and HDA2 50- TTACCGCGGCTGCTGGCA-30 (both primers are disclosed in Walter et al. Appl. Environ.
Microbiol. 66 , 297 (2000)) at 56 °C for strand annealing. Initial denaturation at 94 ° C for 4 min is followed by thirty cycles of 30 s at 94 °C, 30 s at 56 °C and 1 min at 72 °C. The quality of PCR products is verified by agarose gel electrophoresis. Amplified 16S rDNA fragments are separated by denaturing gradient gel electrophoresis (DGGE) using an INGENYphorU system equipped with 6% polyacrylamide gels with a denaturant in the range of 30-55%, where 100% denaturant is equivalent to 7M-urea and 40 % formamide.
Electrophoresis is carried out at 130 V for 4-5 hours at 60 °C. Polyacrylamide gels are stained with GelRede nucleic acid stain for 45 min, destained in ultrapure water and viewed under UV light. Bands of interest are excised from gels and lysed in ultrapure water. Extracted DNA is re-amplified using the same primers and PCR conditions. To purify the bacterial DNA, PCR products are reloaded on a denaturant gradient gel followed by excision and lysis of selected bands. DNA samples recovered from lysed bands of the second DGGE are re-amplified by PCR before purification using the QIAquick PCR Purification Kit and sequenced. Species identification is done using the Ribosomal Microbiome Database Project Classifier tool.
Because of the limited sensitivity of DGGE to quantify microbial diversity, the microbial composition of DNA samples is also analysed using high-throughput sequencing . The V5-V6 region of 16S rRNA from caecal content DNA samples is amplified using the primers 784F 50- AGGATTAGATACCCT- GGTA-30 and 1061R 50-CRRCACGAGCTGACGAC-30 3640 (both primers are disclosed in Andersson et al. , PloS ONE 3 , e2836 (2008)) . Amplicons are pyrosequenced using a Roche 454 GS-FLX system. Sequences of at least 240 nucleotides and containing no more than two undetermined bases are retained for taxonomic assignment. The QIIME software is used for chimera check and the Greengenes database is used for classification. Bacterial diversity is determined at the phylum, family and genus levels.
The results show that HMOs are able to change the intestinal microbiota by increasing the abundance of bifidobacteria . Additionally, HMO supplementation increased the level of GLP1 and reduced body weight and adipose tissue. The level of GLP1 negatively correlated with food intake.
Example 2 - Treating high fat diet induced obesity and diabetes
10-week-old C57BL/6J mice ( 100 mice) are housed in groups of five mice per cage, with free access to food and water. The mice are divided into 10 groups of 10 mice, one control group and 9 treatment groups. All of the mice are fed a high-fat (HF) diet (60 % fat and 20 % carbohydrates [kcal/ 100 g], or an HF diet supplemented with HMO (20 g/kg of diet) for 8 weeks. Food and water intake are recorded twice a week. The 9 treatment groups are each administered one of the following : a) 2'-FL, b) 3-FL, c) 3'-SL, d) 6'-SL, e) LNT, f) LNnT, g) LNFP-I, h) DSLNT and i) a combination of these saccharides. The control group is
administered the HF diet only. Fresh food is given daily.
Intraperitoneal or oral glucose tolerance tests are performed as follows : 6-h-fasted mice are injected with glucose into the peritoneal cavity ( 1 g/kg glucose, 20 % glucose solution) or by gavage (3 g/kg glucose, 66 % glucose solution) . Blood glucose is determined with a glucose meter (Roche Diagnostics) on 3.5 μΙ blood collected from the tip of the tail vein. A total of 20 μΙ blood is sampled 30 min before and 15 or 30 min after the glucose load to assess plasma insulin concentration.
To assess intestinal permeability in vivo, the intestinal permeability of 4000 Da fluorescent dextran-FITC (DX-4000-FITC) is measured . M ice are fasted for 6 h before given DX-44-FITC by gavage (500 mg/kg body weight, 125 mg/ml) . After 1 h and 4 h, 120 ml of blood is collected from the tip of the tail vein. The blood is centrifuged at 4 °C, 12 000 g for 3 min. Plasma is diluted in an equal volume of PBS (pH 7.4) and analysed for DX- 4000-FITC concentration with a fluorescence spectrophotometer at an excitation wavelength of 485 nm and emission wavelength of 535 nm. Standard curves are obtained by diluting FITC-dextran in non-treated plasma diluted with PBS ( 1 : 3 v/v) .
Plasma LPS, cytokines and gut hormones are determined as follows. Plasma LPS
concentration is measured using a kit based upon a Limulus amoebocyte extract (LAL kit endpoint- QCL1000) . Samples are diluted 1/40 to 1/100 and heated for 20 cycles of 10 min at 68 °C and 10 min at 4 °C. An internal control for LPS recovery is included in the calculation. Plasma cytokines (interleukin (IL) la, ILlb, tumour necrosis factor (TNF) a, IL6, monocyte chemoattractant protein (MCP)- l, macrophage inflammatory protein (ΜΙΡ)- Ια, IL10, interferon (INF) c, IL15, IL18) and gut hormones (GLP1 (active), GIP (total), amylin (active), pancreatic polypeptide) are respectively determined in duplicate by using a Bio-Plex Multiplex kit, or a mouse gut hormones panel (LincoPlex), and measured by using Luminex technology, an EIA kit (GLP2 EIA kit) is used to quantify GLP2.
Mice are anaesthetised (ketamine/xylazine, intraperineally, 100 and 10 mg/kg, respectively) after a 5 h period of fasting, and blood samples and tissues are harvested for further analysis. Mice are killed by cervical dislocation. Liver, caecum (full and empty), muscles (vastus lateralis), and adipose tissues (mesenteric and corresponding lymph nodes, epididymal, subcutaneous and visceral) are precisely dissected and weighed . The intestinal segments (jej unum, colon) are immersed in liquid nitrogen, and stored at -80 °C, for further analysis.
To assess the microbiota profile, the caecal contents collected post mortem from mice are stored at -80 °C. DNA is isolated from the caecal content samples using QIAamp DNA Stool Mini Kit. The DNA concentration of extracts is measured using NanoDrop. Aliquots of 100 ng of extracted DNA are subjected to PCR using the 16S rDNA universal heteroduplex analysis (HDA) primers HDA1-GC 50-CGCCCGGGGCGCGCCCCGGGCGGG- GCGGGGGCACGGGGGGACTCCTACGGGAGGCAGCAGT-30 and HDA2 50-
TTACCGCGGCTGCTGGCA-30 (both primers are disclosed in Walter et al. Appl. Environ.
Microbiol. 66 , 297 (2000)) at 56 °C for strand annealing. Initial denaturation at 94 ° C for 4 min is followed by thirty cycles of 30 s at 94 °C, 30 s at 56 °C and 1 min at 72 °C. The quality of PCR products is verified by agarose gel electrophoresis. Amplified 16S rDNA fragments are separated by denaturing gradient gel electrophoresis (DGGE) using an
INGENYphorU system equipped with 6% polyacrylamide gels with a denaturant in the range of 30-55%, where 100% denaturant is equivalent to 7M-urea and 40 % formamide.
Electrophoresis is carried out at 130 V for 4-5 hours at 60 °C. Polyacrylamide gels are stained with GelRede nucleic acid stain for 45 min, destained in ultrapure water and viewed under UV light. Bands of interest are excised from gels and lysed in ultrapure water. Extracted DNA is re-amplified using the same primers and PCR conditions. To purify the bacterial DNA, PCR products are reloaded on a denaturant gradient gel followed by excision and lysis of selected bands. DNA samples recovered from lysed bands of the second DGGE are re-amplified by PCR before purification using the QIAquick PCR Purification Kit and sequenced. Species identification is done using the Ribosormal Microbiorme Database Project Classifier tool.
Because of the limited sensitivity of DGGE to quantify microbial diversity, the microbial composition of DNA samples is also analysed using high-throughput sequencing . The V5-V6 region of 16S rRNA from caecal content DNA samples is amplified using the primers 784F 50- AGGATTAGATACCCT- GGTA-30 and 1061R 50-CRRCACGAGCTGACGAC-30 3640 (both primers are disclosed in Andersson et al. PloS ONE 3 , e2836 (2008)) . Amplicons are pyrosequenced using a Roche 454 GS-FLX system. Sequences of at least 240 nucleotides and containing no more than two undetermined bases are retained for taxonomic assignment. The QIIME software is used for chimera check and the Greengenes database is used for classification. Bacterial diversity is determined at the phylum, family and genus levels.
To assess bacterial translocation from intestine into tissues, mesenteric adipose tissue (MAT) and corresponding lymph nodes (MLN) are harvested, and luminal and mucosal contents of each intestinal segment separated . Quantification of bacterial DNA is performed by isolating genomic DNA from blood, MAT, MLN or intestine (contents and mucosa) . All bacterial DNA is quantified by quantitative real-time PCR targeting conserved regions of the 16S rRNA gene, with bacterial DNA as standard template for absolute quantification.
In order to assess barrier permeability, the expression of occludin and zonula occludens- 1 (ZO- 1) tight-junction proteins are assessed. Jej unum segments are immediately removed, washed with PBS, mounted in embedding medium, and stored at -80 °C until use.
Cryosections (5 mm) are fixed in acetone at -20 °C for 5 min for occludin and in ethanol for 30 min at room temperature and in acetone at -20 °C for 5 min for ZO- 1. Non-specific background is blocked by incubation with 10% bovine serum albumin (BSA) in Tris-buffered saline (TBS) and 0.3% Triton X- 100 (30 min at room temperature) . Sections are incubated with rabbit anti-occludin or rabbit anti-ZO- 1 ( 1 : 400 for ZO- 1 and 1 : 100 for occludin staining) for 2 h. Sections are washed three times for 10 min in TBS and probed with goat anti-rabbit fluorescein isothiocyante (FITC)-conj ugated antibodies (1 : 50) . Slides are washed three times for 10 min in TBS and mounted in mounting medium. Sections are visualised on a
fluorescence microscope. As a control, slides are incubated with serial dilutions of the primary antibody to signal extinction. Two negative controls are used : slides incubated with irrelevant antibody or without primary antibody. All the stainings are performed in duplicate in non- serial distant sections, and analysed in a double-blind manner by two different investigators.
The results show that HMOs improve gut barrier function and reduce the metabolic inflammation and insulin resistance associated with obesity, and increase release of gut peptides, such as glucagon-like peptidel and 2 (GLP1 and GLP2) .
Example 3 - Treating high fat diet induced obesity and diabetes
10-week-old C57BL/6J mice ( 100 mice) are housed in groups of five mice per cage, with free water. The mice are divided into 10 groups of 10 mice, one control group and 9 treatment groups. All of the mice are fed a high-fat (HF) diet (60 % fat and 20 % carbohydrates [kcal/100 g], or an HF diet supplemented with HMO (20 g/kg of diet) for 8 weeks. Food and water intake are recorded twice a week. The 9 treatment groups are each administered one of the following : a) 2'-FL, b) 3-FL, c) 3'-SL, d) 6'-SL, e) LNT, f) LNnT, g) LNFP-I, h) DSLNT and i) a combination of these saccharides. The control group is administered the HF diet only. Intraperitoneal or oral glucose tolerance tests are performed as follows : 6-h-fasted mice are injected with glucose into the peritoneal cavity ( 1 g/kg glucose, 20 % glucose solution) or by gavage (3 g/kg glucose, 66 % glucose solution) . Blood glucose is determined with a glucose meter (Roche Diagnostics) on 3.5 μΙ blood collected from the tip of the tail vein. A total of 20 μΙ blood is sampled 30 min before and 15 or 30 min after the glucose load to assess plasma insulin concentration.
Plasma triglyceride and cholesterol is measured from blood taken during the treatment period.
To assess intestinal permeability in vivo, the intestinal permeability of 4000 Da fluorescent dextran-FITC (DX-4000-FITC) is measured . M ice are fasted for 6 h before given DX-44-FITC by gavage (500 mg/kg body weight, 125 mg/ml) . After 1 h and 4 h, 120 ml of blood is collected from the tip of the tail vein. The blood is centrifuged at 4 °C, 12 000 g for 3 min. Plasma is diluted in an equal volume of PBS (pH 7.4) and analysed for DX- 4000-FITC concentration with a fluorescence spectrophotometer at an excitation wavelength of 485 nm and emission wavelength of 535 nm. Standard curves are obtained by diluting FITC-dextran in non-treated plasma diluted with PBS ( 1 : 3 v/v) .
Mice are anaesthetised (ketamine/xylazine, intraperineally, 100 and 10 mg/kg, respectively) after a 5 h period of fasting, blood samples and tissues are harvested for further analysis. Mice are killed by cervical dislocation. Liver, caecum (full and empty), and adipose tissues (mesenteric and corresponding lymph nodes, epididymal, subcutaneous and visceral) are precisely dissected, weighed and stored at -80 °C, for further analysis.
Total and active GLP1 are measured from blood with ELISA (M illipore, Molsheim, France) .
To assess the microbiota profile, the caecal contents collected post mortem from mice are stored at -80 °C. DNA is isolated from the caecal content samples using QIAamp DNA Stool Mini Kit. The DNA concentration of extracts is measured using NanoDrop. Aliquots of 100 ng of extracted DNA are subjected to PCR using the 16S rDNA universal heteroduplex analysis (HDA) primers HDA1-GC 50-CGCCCGGGGCGCGCCCCGGGCGGG- GCGGGGGCACGGGGGGACTCCTACGGGAGGCAGCAGT-30 and HDA2 50- TTACCGCGGCTGCTGGCA-30 (both primers are disclosed in Walter et al. Appl. Environ.
Microbiol. 66 , 297 (2000)) at 56 °C for strand annealing. Initial denaturation at 94 ° C for 4 min is followed by thirty cycles of 30 s at 94 °C, 30 s at 56 °C and 1 min at 72 °C. The quality of PCR products is verified by agarose gel electrophoresis. Amplified 16S rDNA fragments are separated by denaturing gradient gel electrophoresis (DGGE) using an
INGENYphorU system equipped with 6% polyacrylamide gels with a denaturant in the range of 30-55 %, where 100 % denaturant is equivalent to 7M-urea and 40 % formarmide.
Electrophoresis is carried out at 130 V for 4-5 hours at 60 °C. Polyacrylamide gels are stained with GelRede nucleic acid stain for 45 min, destained in ultrapure water and viewed under UV light. Bands of interest are excised from gels and lysed in ultrapure water. Extracted DNA is re-amplified using the same primers and PCR conditions. To purify the bacterial DNA, PCR products are reloaded on a denaturant gradient gel followed by excision and lysis of selected bands. DNA samples recovered from lysed bands of the second DGGE are re-amplified by PCR before purification using the QIAquick PCR Purification Kit and sequenced. Species
identification is done using the Ribosomal Microbiome Database Project Classifier tool.
Because of the limited sensitivity of DGGE to quantify microbial diversity, the microbial composition of DNA samples is also analysed using high-throughput sequencing . The V5-V6 region of 16S rRNA from caecal content DNA samples is amplified using the primers 784F 50- AGGATTAGATACCCT- GGTA-30 and 1061R 50-CRRCACGAGCTGACGAC-30 3640 (both primers are disclosed in Andersson et al. PloS ONE 3 , e2836 (2008)) . Amplicons are pyrosequenced using a Roche 454 GS-FLX system. Sequences of at least 240 nucleotides and containing no more than two undetermined bases are retained for taxonomic assignment. The QIIME software is used for chimera check and the Greengenes database is used for classification. Bacterial diversity is determined at the phylum, family and genus levels.
The results show that HMOs are able to change the intestinal microbiota by increasing the abundance of bifidobacteria . Additionally, HMO supplementation reduces cholesterol, body weight, fat accumulation and glucose tolerance.
Example 4 - Treating obesity induced dia betes
Six-week-old ob/ob mice ( 120 mice) on C57BL/6 background are housed in a controlled environment ( 12 h daylight cycle) in groups of 2 mice/cage, and kept with free access to food and drinking water. The mice are separated into 10 groups of 10 mice, one control group and 9 treatment groups. One group is fed a control diet, and the 9 treatment groups each receive a control diets containing one of the following HMOs (20 g/kg of diet) for five weeks: a) 2'-FL, b) 3-FL, c) 3'-SL, d) 6'-SL, e) LNT, f) LNnT, g) LNFP-I, h) DSLNT, and i) a combination of these saccharides. Fresh food is given daily.
Experiments to show impact of HMOs on glucose tolerance, intestinal permeability plasma LPS, cytokines and gut hormones, caecal microbiota profile and bacterial translocation are performed as described under Example 2.
Example 5 - Human trial in overweight and obese children
A total of 75 male and female patients, enrolled to a childhood obesity treatment program, are recruited to participate in the study. Patients are randomized into three groups, each of 25 patients, with 2 groups receiving different investigational products and one group receiving a placebo product for 8 weeks. The investigational products contain 4.5 grams of either 2'-FL alone or a combination of 2'-FL and LNnT in a 4: 1 ratio while the placebo product contains 4.5 grams of glucose. All products are in powder form in a unit dosage container.
The patients are eligible to participate if: they are between 5 and 12 years of age, have a BMI SDS (Standard Deviation Score) of ≥ 2.0 and are enrolled in the childhood obesity treatment program at the Children's Obesity Clinic. All recruited patients and their representatives are able and willing to understand and comply with the study procedures. Patients are excluded if: they have participated in a clinical study one month prior to the screening visit and throughout the study; have any gastrointestinal disease(s) that may cause symptoms or may interfere with the trial outcome; have other severe disease(s) such as malignancy, kidney disease or neurological disease; have psychiatric disease; have used highly dosed probiotic supplements (yoghurt allowed) 3 months prior to screening and throughout the study; have consumed antibiotic drugs 3 months prior to screening and throughout the study; and consume on a regular basis medication that might interfere with symptom evaluation 2 weeks prior to screening and throughout the study.
At the initial visit (screening) patients and their representatives are given both oral and written information about the study; the children are asked for informed assent and their representatives to sign an informed consent form.
Eligibility criteria are checked and for children who are enrolled to the study, medical history and concomitant medication are registered. A physical examination is done and pubertal staging is determined. Blood pressure, pulse rate, height and bodyweight are measured, and body composition is determined by a DXA (dual energy x-ray absorptiometry)-scan and bioimpedance. BMI SDS is calculated, waist and hip circumferences measured and food intake registered. Fasting blood samples are collected for safety and biomarker studies and for biobanking.
The serum from the blood samples is transferred to cryotubes and stored at -80° C. The following biomarkers are measured; Lipopolysaccharides (LPS), hsCRP, free fatty acids, total cholesterol, HDL, LDL, HbAlc, glucose, insulin, triglycerides, TNF-a, IL-Ιβ, IL-6, IL-8, IL-10, GLP1, GLP2, Adiponectin, and Zonulin.
Equipment for collecting faecal samples is distributed. The faecal samples are stored at -80° C until analysis. Microbiological analysis is performed on the faecal samples using 16S rRNA gene sequencing.
The Gastrointestinal Symptom Rating Scale (GSRS) questionnaire is completed on site by the participating child's representative(s), and the Bristol Stool Form Scales (BSFS) is distributed to the participant's representative(s) with instructions to assess the stool consistency during the study and at each faecal sampling point using the BSFS.
At the second visit (randomization), patients and their representatives are asked about adverse events, faecal samples are collected and equipment for collection of new samples is distributed. BSFS is collected and new BSFS is distributed. Study products are distributed together with a compliance form (diary). Patients and their representatives are reminded to follow the healthy dietary habits.
The study runs for 8 weeks with the patients consuming either a placebo or one of two investigational products daily. Patients are instructed to consume the products in the morning with breakfast. Compliance is monitored via a compliance form (diary) to be filled in daily.
Four weeks after commencement there is an intermediate check. Patients and their representatives are asked about adverse events and any changes in the patient's usual medication. Faecal samples are collected and equipment for collection of new samples is distributed. Blood pressure, pulse rate, waist and hip circumference, height and bodyweight are measured and BMI SDS calculated. The GSRS is completed on site by the participating child's representative. The BSFS is collected and new BSFS is distributed to the participant's representative(s) with instructions to assess the stool consistency at each faecal sampling point using the BSFS. Patients and their representatives are reminded to follow the healthy dietary habits.
At the end of intervention (8 weeks), each patient has a visit with the medical team. Patients and their representatives are asked about adverse events and any changes in the patient's usual medication. Study products and compliance forms are collected to check compliance. BSFS and faecal samples are collected and equipment for collection of new samples is distributed. A physical examination is done and pubertal staging is determined. Blood pressure, pulse rate, height and bodyweight are measured, and body composition is determined by a DXA (dual energy x-ray absorptiometry)-scan and bioimpedance. BMI SDS is calculated, waist and hip circumferences measured and food intake registered. Fasting blood samples are collected for safety and biomarker studies and for biobanking, and equipment for collecting faecal samples is distributed. The GSRS questionnaire is completed on site by the participating child's representative(s).
To examine potential long term effects of the intervention, an un-blinded follow-up period follows with a visit 8 weeks after end of intervention. A physical examination is done and pubertal staging is determined. Blood pressure, pulse rate, height and bodyweight are measured, and body composition is determined by a DXA (dual energy x-ray
absorptiometry)-scan and bioimpedance. BMI SDS is calculated, waist and hip circumferences measured and food intake registered. Fasting blood samples are collected for safety and biomarker studies and for biobanking. Faecal samples are collected.
The results show that oral ingestion of HMOs modulate the intestinal microbiota, and specifically stimulate the growth of bifidobacteria. The blood biomarker analysis indicates that the patients given the investigational products have increased levels of GLP1. The level of GLP1 correlated positively with the abundance of bifidobacteria. Collectively, HMOs are able to increase bifidobacteria and change the intestinal environment, and by this, increasing the level of GLP1 in obese children. The results also show that oral ingestion of HMOs modulate the intestinal microbiota, and specifically stimulate the growth of bifidobacteria, particularly species belonging to the B. adolescentis phylogenetic group, and change the SCFA profile. The blood biomarker analysis indicated that the patients given the investigational products have a lipid profile with lower triglyceride levels and higher high-density lipoprotein cholesterol. Additionally, the blood pressure and body composition is decreased . The abundance of bifidobacteria correlates negatively with the level of low-density lipoprotein cholesterol and positively with the level of high-density lipoprotein cholesterol. Collectively, HMOs are able to increase bifidobacteria and change the intestinal environment, and by this, improve the lipid profile, hypertension and body composition, all incidence reducing the risk of CVD.
The patients receiving the intervention product have lower HOMA-IR scores. Further, the patients given the investigational products show a greater reduction of body fat, body weight and BMI SDS as compared to the placebo group. The blood biomarker analysis indicates that the patients given the investigational products have increased levels of GLP1 and GLP2, reduced levels of metabolic endotoxemia and inflammatory markers and reduced gut permeability indicating an improved mucosal barrier compared to the placebo. The faecal analysis indicates that the patients given the investigational products have reduced bacterial dysbiosis and a higher level of bifidobacteria compared to the placebo, particularly
Bifidobacterium pseudocatenulatum .
Example 6 - Nutritional composition
A ready to feed nutritional composition is prepared from water, maltodextrin, milk protein concentrate, Sucromalt, glycerine, cocoa powder, soy protein isolate, fructose, high oleic safflower oil, soy oil, canola oil, plant sterol esters, HMOs, soy lecithin, magnesium chloride, calcium phosphate, carrageenan, sodium ascorbate, potassium citrate, sodium phosphate, calcium citrate, choline chloride, potassium chloride, sodium citrate, magnesium oxide, taurine, L-carnitine, alpha-tocopheryl acetate, zinc sulphate, ferrous sulphate, niacinamide, calcium pantothenate, vitamin A palmitate, citric acid, manganese sulphate, pyridoxine hydrochloride, vitamin D3, copper sulphate, thiamine mononitrate, riboflavin, beta carotene, folic acid, biotin, potassium iodide, chromium chloride, sodium selenate, sodium molybdate, phytonadione, vitamin B12.
The composition has an energy density of 0.8 kcal/ml with an energy distribution (% of kcal) as follows : protein : 20 %, carbohydrate : 48 %, fat: 32 %.
Example 7 - Tablet composition
A tablet is prepared from HMO, hydroxypropyl methylcellulose, sodium alginate, gum, microcrystalline cellulose, colloidal silicon dioxide, and magnesium stearate. All raw materials except the magnesium stearate are placed into a high shear granulator and premixed . Water is sprayed onto the premix while continuing to mix at 300 rpm. The granulate is transferred to a fluidised bed drier and dried at 75 °C. The dried powder is sieved and sized using a mill. The resulting powder is then lubricated with magnesium stearate and pressed into tablets. The tablets each contain 325 mg of HMO. The tablets each have a weight of 750 mg.
Example 8 - Capsule composition
A capsule is prepared by filling about 1 g of HMO into a 000 gelatine capsule using a filing machine. The capsules are then closed. The HMO are in free flowing, powder form.

Claims

CLAI MS
1. One or more human milk oligosaccharides (HMOs) for use in
- stabilising or reducing insulin resistance in a human individual having an obesity-related metabolic disorder,
- regulating satiety in a human having a metabolic disorder,
- reducing propensity to obesity in a human having a metabolic disorder,
- reducing the propensity of a cardiovascular disease (CVD) and/or a CVD- associated pathological condition or disease in a human, preferably, in an overweight or obese human individual,
- for preventing development of a cardiovascular disease (CVD) and/or a CVD- associated pathological condition or disease in a human, preferably, in an overweight or obese human individual, and/or
- treating a cardiovascular disease (CVD) and/or a CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual.
2. The one or more HMOs for the use according to claim 1, wherein the HMO is a
fucosylated or a non-fucosylated neutral HMO.
3. The one or more HMOs for the use according to claim 1 or 2, which is a mixture of at least a fucosylated and at least a non-fucosylated neutral HMO.
4. The one or more HMOs for the use according to any of the claims 1 to 3, wherein the fucosylated HMO is selected from 2'-FL, 3-FL and DFL, and the non-fucosylated neutral HMO is selected from LNT and LNnT.
5. The mixture of at least a fucosylated and at least a non-fucosylated neutral HMO for the use according to claim 3 or 4, in which the HMOs comprise or essentially consist of any of the combinations of 2'-FL and LNnT, 2'-FL and LNT, or 2'-FL, LNT and LNnT.
6. The one or more HMOs for the use according to claim 1, wherein the HMO is lacto-N- tetraose (LNT), lacto-N-neotetraose (LNnT), 2'-fucosyllactose (2'-FL), lacto-N- fucopentaose I (LNFP-I), 3-fucosyllactose (3-FL), difucosyllactose (DFL), 3'- sialyllactose (3'-SL) or 6'-sialyllactose (6'-SL), or a mixture thereof.
7. The one or more HMOs for the use according to any of the claims 1 to 6 effective to increase the abundance of bifidobacteria in the gastrointestinal tract of the human.
8. The one or more HMOs for the use according to claim 7, wherein the abundance of bifidobacteria is the relative abundance of bifidobacteria of the phylogenetic
Bifidobacterium adolescentis group, preferably Bifidobacterium adolescentis and/or Bifidobacterium pseudocatenulatum .
9. The one or more HMOs for the use according to claim 1, wherein the use is stabilising or reducing insulin resistance in a human individual having an obesity-related metabolic disorder.
10. The one or more HMOs for the use according to claim 9, wherein the obesity-related metabolic disorder is obesity, obesity induced pre-diabetes or obesity induced type 2 diabetes.
11. The one or more HMOs for the use according to claim 9 or 10, wherein the human individual is an obese paediatric patient.
12. The one or more HMOs for the use according to claim 1, wherein the use is:
- regulating satiety, and/or
- reducing propensity to obesity in a human having a metabolic disorder in a human having a metabolic disorder.
13. The one or more HMOs for the use according to claim 12, the human having a
metabolic disorder is obese or has obesity induced pre-diabetes or type 2 diabetes.
14. The one or more HMOs for the use according to claims 9 to 13, which is a mixture of 2'-FL and LNnT in a mass ratio of 5: 1 to 1 : 1.
15. The one or more HMOs for the use according to claim 1, wherein the use is:
- reducing the propensity of,
- preventing development of, and/or
- treating
a cardiovascular disease (CVD) and/or a CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual.
16. The one or more HMOs for the use according to claim 15, wherein the HMO is a single HMO or a mixture of two to five HMOs.
17. The one or more HMOs for the use according to claim 15, wherein the CVD-associated pathologic condition or disease is selected from heart failure, heart attack, stroke, pulmonary embolism, cardiac arrest and peripheral artery disease (PAD) .
18. The one or more HMOs for the use according to any of the claims 15 to 17, which is a mixture of 2'-FL and LNnT in a mass ratio of 1.5: 1 to 4: 1.
19. A synthetic composition for use in
- regulating satiety in a human having a metabolic disorder,
- reducing propensity to obesity in a human having a metabolic disorder, - stabilising or reducing insulin resistance in a human individual having an obesity-related metabolic disorder,
- reducing the propensity of a cardiovascular disease (CVD) and/or a CVD- associated pathological condition or disease in a human, preferably, in an overweight or obese human individual,
- for preventing development of a cardiovascular disease (CVD) and/or a CVD- associated pathological condition or disease in a human, preferably, in an overweight or obese human individual, and/or
- treating a cardiovascular disease (CVD) and/or a CVD-associated pathological condition or disease in a human, preferably, in an overweight or obese human individual,
wherein the composition contains an effective amount of one or more human milk oligosaccharides (HMOs), preferably those specified in any of the claims 2 to 6.
The synthetic composition for the use according to claim 19, which is a nutritional composition.
The synthetic composition for the use according to claim 19 or 20, which is in a unit dosage form.
The one or more HMOs for the use according to any of the claims 1 to 18, or the synthetic composition for the use according to any of the claims 19 to 21, which is administered to the human in need enterally.
One or more human milk oligosaccharides (HMOs) selected from the group consisting of fucosylated HMOs and non-fucosylated HMOs, preferably a mixture of one or more fucosylated HMOs and one or more non-fucosylated neutral HMOs, for use in increasing the abundance of bifidobacteria in a human, preferably in an overweight or obese human having a propensity of, or diagnosed with a cardiovascular disease (CVD).
A synthetic composition for use in increasing the abundance of bifidobacteria in a human, preferably, in an overweight or obese human having a propensity of, or diagnosed with a cardiovascular disease (CVD), the synthetic composition comprising one or more human milk oligosaccharides (HMOs) selected from the group consisting of fucosylated HMOs and non-fucosylated neutral HMOs, preferably a mixture of one or more fucosylated HMOs and one or more non-fucosylated neutral HMOs.
EP17812794.0A 2016-06-15 2017-06-15 Synthetic compositions comprising human milk oligosaccharides for use the prevention and treatment of disorders Withdrawn EP3471562A4 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US15/183,391 US10835544B2 (en) 2014-12-08 2016-06-15 Synthetic composition for regulating satiety
US15/183,431 US10987368B2 (en) 2014-12-08 2016-06-15 Synthetic composition for preventing or treating CVD
US15/183,404 US10881674B2 (en) 2014-12-08 2016-06-15 Synthetic composition for treating metabolic disorders
PCT/DK2017/050198 WO2017215721A1 (en) 2016-06-15 2017-06-15 Synthetic compositions comprising human milk oligosaccharides for use γν the prevention and treatment of disorders

Publications (2)

Publication Number Publication Date
EP3471562A1 true EP3471562A1 (en) 2019-04-24
EP3471562A4 EP3471562A4 (en) 2020-07-29

Family

ID=60663363

Family Applications (1)

Application Number Title Priority Date Filing Date
EP17812794.0A Withdrawn EP3471562A4 (en) 2016-06-15 2017-06-15 Synthetic compositions comprising human milk oligosaccharides for use the prevention and treatment of disorders

Country Status (2)

Country Link
EP (1) EP3471562A4 (en)
WO (1) WO2017215721A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114728015A (en) * 2019-11-14 2022-07-08 格礼卡姆股份公司 Synthetic composition for balancing intestinal bile acid spectrum
WO2021123253A1 (en) * 2019-12-19 2021-06-24 Basf Se Composition for the treatment of (pre)hyperglycemia and/or associated morbidities thereto
WO2021152176A1 (en) * 2020-01-31 2021-08-05 N.V. Nutricia Nutritional composition for use in gut maturation
CN115335063A (en) * 2020-03-25 2022-11-11 菲仕兰坎皮纳荷兰公司 Compositions comprising one or more HMOs having a LacNAc-Lac core
TW202233201A (en) * 2020-12-16 2022-09-01 荷蘭商弗里斯蘭康必奶荷蘭有限公司 Synthetic compositions comprising lnfp iii and lsta
CN114145353A (en) * 2021-11-30 2022-03-08 内蒙古伊利实业集团股份有限公司 Formula milk powder capable of improving intestinal microenvironment health and preparation method and application thereof

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5906982A (en) * 1997-03-31 1999-05-25 Abbott Laboratories Nutritional formulations containing Lacto-N-neoTetraose
RU2444210C2 (en) * 2005-12-23 2012-03-10 Н.В. Нютрисиа Baby nutritional compositions intended for prevention of adiposity
EP2072052A1 (en) * 2007-12-17 2009-06-24 Nestec S.A. Prevention of opportunistic infections in immune-compromised subjects
PL2234505T3 (en) * 2007-12-21 2013-06-28 Nutricia Nv Use of sphingomyelin and non-digestible carbohydrates for improving intestinal microbiota
WO2011119023A1 (en) * 2010-03-26 2011-09-29 N.V. Nutricia Low protein infant formula with increased essential amino acids
CN103889240B (en) * 2011-10-18 2016-09-14 雀巢产品技术援助有限公司 For increasing insulin sensitivity and/or reducing the compositions of insulin resistance
ES2891731T3 (en) * 2013-05-22 2022-01-31 Glycom As Synthetic mixture of oligosaccharides for the treatment of the microbiota of a mammal
US20150305385A1 (en) * 2014-04-25 2015-10-29 Mead Johnson Nutrition Company Pediatric nutritional composition with human milk oligosaccahrides, prebiotics and probiotics
CN115350200A (en) * 2014-12-08 2022-11-18 格礼卡姆股份公司 Synthetic compositions for the treatment of metabolic disorders
RU2018130329A (en) * 2016-01-26 2020-02-27 Сосьете Де Продюи Нестле С.А. COMPOSITIONS CONTAINING 2FL AND LNNT FOR USE IN INFANTS OR CHILDREN FOR PREVENTION OF OBESITY IN THE FURTHER LIFE OR RELATED COMORBIDITY
AU2017213083A1 (en) * 2016-01-26 2018-05-10 Société des Produits Nestlé S.A. Compositions comprising sialylated oligosaccharides for use in infants or young children to prevent later in life obesity or related comorbidities and promote a healthy growth
US20210205341A1 (en) * 2016-01-26 2021-07-08 Nestec S.A. Compositions comprising human milk oligosaccharides for use in infants or young children to prevent or treat a health disorder by increasing glp-1 secretion
CN108495636A (en) * 2016-01-26 2018-09-04 雀巢产品技术援助有限公司 The human milk oligosaccharides beneficial to health by preventing fat rebound too early

Also Published As

Publication number Publication date
WO2017215721A1 (en) 2017-12-21
EP3471562A4 (en) 2020-07-29

Similar Documents

Publication Publication Date Title
US11529364B2 (en) Synthetic composition for treating metabolic disorders
JP7301093B2 (en) Synthetic compositions and methods for treatment of irritable bowel syndrome
US10987368B2 (en) Synthetic composition for preventing or treating CVD
EP3471562A1 (en) Synthetic compositions comprising human milk oligosaccharides for use the prevention and treatment of disorders
WO2017046711A1 (en) Synthetic composition for microbiota modulation
US11291677B2 (en) Synthetic composition for microbiota modulation
US11890293B2 (en) Synthetic composition for treating metabolic disorders
BR112020010843A2 (en) human milk oligosaccharides and synthetic compositions for modulation of the microbiota
EP3458073A1 (en) Synthetic composition
EP3493686B1 (en) Nutritional compositions and infant formulas comprising a mix of oligosaccharides and optionally bifidobacterium lactis for preventing, treating or reducing the severity of non-rotavirus-associated diarrhoea
EP3589139A1 (en) Synthetic composition for microbiota modulation
CA3081496A1 (en) Mixture of hmos for reducing or preventing fatigue
EP3630123A1 (en) Synthetic composition comprising one or more human milk oligosaccharides (hmos)
EP4058031A1 (en) Synthetic composition for balancing the bile acid profile in the intestine
US10835544B2 (en) Synthetic composition for regulating satiety
EP3716984A1 (en) Mixture of hmos for treating wheat sensitivity
JP2022535718A (en) Human milk oligosaccharides for use in treating symptoms in patients suffering from non-celiac wheat and/or gluten sensitivity

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20190115

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 31/702 20060101ALI20200123BHEP

Ipc: A61P 3/04 20060101ALN20200123BHEP

Ipc: A23L 33/125 20160101AFI20200123BHEP

Ipc: A61P 3/10 20060101ALN20200123BHEP

Ipc: A61P 9/10 20060101ALN20200123BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20200626

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 9/10 20060101ALN20200622BHEP

Ipc: A61P 3/04 20060101ALN20200622BHEP

Ipc: A61P 3/10 20060101ALN20200622BHEP

Ipc: A23L 33/125 20160101AFI20200622BHEP

Ipc: A61K 31/702 20060101ALI20200622BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20230922

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20240203