WO2020130836A1 - Compound for use in treatment and prevention of type i diabetes - Google Patents

Compound for use in treatment and prevention of type i diabetes Download PDF

Info

Publication number
WO2020130836A1
WO2020130836A1 PCT/NL2019/050872 NL2019050872W WO2020130836A1 WO 2020130836 A1 WO2020130836 A1 WO 2020130836A1 NL 2019050872 W NL2019050872 W NL 2019050872W WO 2020130836 A1 WO2020130836 A1 WO 2020130836A1
Authority
WO
WIPO (PCT)
Prior art keywords
mammal
diazoxide
diabetes
treatment
pharmaceutically acceptable
Prior art date
Application number
PCT/NL2019/050872
Other languages
French (fr)
Inventor
Michiel F. NIJHOFF
Eelco J.P. DE KONING
Original Assignee
ACADEMISCH ZIEKENHUIS LEIDEN (h.o.d.n. LUMC)
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 NL2022291A external-priority patent/NL2022291B1/en
Application filed by ACADEMISCH ZIEKENHUIS LEIDEN (h.o.d.n. LUMC) filed Critical ACADEMISCH ZIEKENHUIS LEIDEN (h.o.d.n. LUMC)
Publication of WO2020130836A1 publication Critical patent/WO2020130836A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/549Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame having two or more nitrogen atoms in the same ring, e.g. hydrochlorothiazide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics

Definitions

  • the present invention relates to diazoxide or a pharmaceutically acceptable salt thereof for use as a medicament at low doses to treat type I diabetes and to compositions containing diazoxide for use in the treatment of a mammal afflicted with, or at risk of developing type I diabetes.
  • Diabetes mellitus commonly referred to as diabetes, is a group of metabolic disorders in which a mammal's blood contains high levels of sugar, also referred to as hyperglycemia, over a prolonged period.
  • Diabetes is due to a mammal's pancreas not producing enough insulin or the mammal's somatic cells not responding properly to the insulin produced by the mammal.
  • diabetes mellitus There are three main types of diabetes mellitus:
  • Type I results from the failure of the mammal's pancreas to produce enough insulin due to loss of beta cells.
  • Type 2 begins with insulin resistance, a condition in which the mammal's cells fail to respond to insulin properly; as the disease progresses, a lack of insulin can also develop.
  • Gestational diabetes occurs when pregnant women without a previous history of diabetes develop high blood sugar levels.
  • US5284845B describes the use of diazoxide for the treatment of disorders caused by defects in glucose metabolism, including hyperglycemia and hypoglycemia and to delay or prevent onset of insulin dependency in Type I diabetic subjects.
  • EP2422787 describes the use of diazoxide for hypoglycaemia, as well as amyotrophic lateral sclerosis (ALS) and hypertension.
  • ALS amyotrophic lateral sclerosis
  • the principal use of diazoxide in diabetes therapy currently is for the treatment of hyperinsulinemia leading to
  • hypoglycaemia wherein hyperinsulinemia is characterised as excess levels of insulin circulating in blood relative to glucose.
  • the present invention relates to diazoxide or a pharmaceutically acceptable salt thereof for use as a medicament in combination with an immunosuppressive compound and/or an anti-inflammatory compound, preferably a corticosteroid, in a treatment of a mammal afflicted with Type I diabetes.
  • an immunosuppressive compound and/or an anti-inflammatory compound preferably a corticosteroid
  • the treatment substantially prevents further deterioration of beta cells in the mammal's pancreas and/or protects beta cells in the mammal's pancreas during and shortly after islet transplantation against loss due to ischemia and/or protects beta cells in the mammal's pancreas against loss due to rejection after pancreas or islet transplantation.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising diazoxide or a pharmaceutically acceptable salt thereof and an
  • immunosuppressive compound and/or an anti-inflammatory compound for use in the treatment of a mammal afflicted with, or prone to develop Type I diabetes.
  • the present invention relates to the use of diazoxide for preventing or treating the onset and progression of Type I diabetes when a large mass of beta cells is still present in the mammal's pancreas which will likely quickly deteriorate.
  • Applicants have surprisingly found that the administration of diazoxide, preferably in combination with an
  • immunosuppressive compound and/or an anti-inflammatory compound may protect beta cells from the auto-immune reaction considered as the underlying cause for this ailment.
  • the present invention in a first instance relates to a composition comprising diazoxide for the protection of beta cells.
  • the present invention relates to a process wherein diazoxide is used as a beta cell protective agent, combined with an immunosuppressive compound and/or the anti-inflammatory compound, preferably a corticosteroid.
  • the process relates to a treatment with 100 - 1600mg per day, more preferably 200 - 1200 mg per day, more preferably of from 300 to 800mg per day of diazoxide.
  • the process also relates to a treatment with one or more
  • immunosuppressive compounds and/or one or more anti-inflammatory compounds selected from the group consisting of: • a corticosteroid, particularly methylprednisolone (dose range 5 - 2000 mg per day), prednisolone (dose range 1,25 - 120 mg per day), dexamethasone (0.25 - 24 mg per day), cortisone (10 - 1000 mg per day), or hydrocortisone (10 - 1000 mg per day), or
  • a calcineurin-inhibitor particularly tacrolimus (dose range 0.5 - 24 mg per day) or cyclosporine and its derivatives (dose range 25 - 750mg per day);
  • an antimetabolite particularly azathioprine and its derivatives (dose range 25 - 1000 mg per day), mycophenolate mofetil (dose range 250 - 4000 mg per day); mycophenolic acid (dose range 180 - 2880 mg per day,) and/or methotrexate (dose range 0.5 - 20 mg per day);
  • an mTOR inhibitor particularly everolimus (dose range 0.25 - 10 mg per day) or sirolimus (dose range 0.5 - 80 mg per day);
  • an anti-lymphocyte antibody particularly alemtuzumab (dose range 0.1 - 100 mg), rituximab (200 - 2000 mg) or thymoglobulin (dose range 10 - 500 mg per day); and/or
  • an anti-cytokine antibody particularly basiliximab (dose range 10 - 100 mg), anakinra (dose range 10 - 500 mg per day), tocilizumab (dose range 8 - 1600 mg), etanercept (dose range 1 - 10 mg per day), Adalimumab (dose range 5 -640 mg), infliximab (dose range 5 - 2000 mg), golimumab (dose range 25 - 800 mg), certolizumab (dose range 50 - 1600 mg).
  • basiliximab dose range 10 - 100 mg
  • anakinra dose range 10 - 500 mg per day
  • tocilizumab dose range 8 - 1600 mg
  • etanercept dose range 1 - 10 mg per day
  • Adalimumab dose range 5 -640 mg
  • infliximab dose range 5 - 2000 mg
  • golimumab dose range 25 - 800 mg
  • certolizumab dose range 50 - 1600 mg.
  • the process also relates to a treatment with one or more immunosuppressive compounds and/or one or more anti-inflammatory compounds selected from the group consisting of:
  • a corticosteroid particularly methylprednisolone (dose range 5 - 2000 mg per day), prednisolone (dose range 1,25 - 120 mg per day), dexamethasone (0.25 - 24 mg per day), cortisone (10 - 1000 mg per day), or hydrocortisone (10 - 1000 mg per day), or
  • Fig 1 discloses the stimulation index in presence of diazoxide or a placebo compounds.
  • Fig 2 discloses the stimulation index in presence of diaozixe or a placebo compounds for Human isolated islets of Langerhans.
  • Figure 3 discloses Dynamic GSIS of islets incubated in diazoxide shows an improved insulin secretion profile in human islets incubated in diazoxide 325uM as compared to placebo)
  • Figures 4A and 4B shows that markers for ER stress are increased in hyperglycemic conditions, this increase is mitigated by addition of diazoxide.
  • FIG. 5A and 5B shows that markers for OS are increased in hyperglycemic conditions, this increase is mitigated by addition of diazoxide.
  • combination therapy may protect the beta cell from both the beta cell stress and the inflammation.
  • a first situation whereby the combination may be applied is during the onset and progression of Type I diabetes, i.e. when a majority or at least large group of beta cells is still present, but shows a fast deterioration, whereby applicants have found that diazoxide treatment ameliorates this deterioration.
  • these beta cells may be kept alive.
  • a second situation in which beta cell protection may be applied is during, and shortly after islet transplantation, since transplanted islets require several weeks to be fully vascularized and a large portion of transplanted islets are lost due to ischemia, whereby the combination therapy may advantageously be employed.
  • a third situation in which beta cell protection may be applied is in the case of a rejection after pancreas or islet transplantation.
  • a fourth situation in which beta cell protection may be applied is in the case of a person having a substantially increased risk of developing Type I diabetes, as indicated by biomarkers or genetic predisposition.
  • diazoxide or a pharmaceutically acceptable salt thereof is used as a medicament in combination with one or more immunosuppressive compounds and/or anti-inflammatory compounds, particularly one or more corticosteroids, in a treatment of a mammal afflicted with Type I diabetes or the onset thereof, and/or at risk of developing Type I diabetes.
  • a pharmaceutical composition comprising diazoxide or a pharmaceutically acceptable salt thereof and the one or more immunosuppressive compounds and/or anti inflammatory compounds, particularly one or more corticosteroids, as the active
  • corticosteroid preferably means a glucocorticoid, more preferably a natural glucocorticoid or a synthetic corticosteroid-like compound, such as betamethasone, prednisone, dexamethasone, cortisone, hydrocortisone,
  • mammal preferably means a primate, a human or a domestic or farm animal, such as a cow, horse, pig, sheep, goat, dog, cat or rodent, more preferably a human.
  • the term "pharmaceutically acceptable salt thereof” preferably means a free compound or, for example, a pharmaceutically acceptable, non-toxic organic or inorganic acid or base addition salt of diazoxide.
  • the term "mammal ... at risk of developing Type I diabetes” preferably means a mammal testing positive for one or more diabetes-associated antibodies and/or with a positive family history for Type I diabetes in one or more first-degree relatives and/or with an increased risk for Type I diabetes based on gene analysis thereof as described by: Redondo MJ, Oram RA, Steck AK. Genetic Risk Scores for Type I Diabetes Prediction and Diagnosis. Curr Diab Rep, 2017; Regnell SE, Lernmark A. Early prediction of autoimmune (Type I) diabetes. Diabetologia, 2017; Bonifacio E. Predicting Type I diabetes using biomarkers. Diabetes Care 2015.
  • diazoxide or a pharmaceutically acceptable salt thereof and an immunosuppressive compound and/or an anti-inflammatory compound, particularly a corticosteroid, or preferably a pharmaceutical composition thereof can be administered to the mammal orally, topically, intravenously, subcutaneously, buccally, rectally, dermally, nasally, tracheally, bronchially, by any other parenteral route, as an oral or nasal spray or via inhalation.
  • a pharmaceutical composition of this invention comprising diazoxide or a pharmaceutically acceptable salt thereof and the
  • immunosuppressive compound and/or anti-inflammatory compound, particularly the corticosteroid can be suitably administered.
  • this treatment substantially prevents further deterioration of beta cells in the mammal's pancreas and/or protects beta cells in the mammal's pancreas during and shortly after islet transplantation against loss due to ischemia and/or protects beta cells in the mammal's pancreas against loss due to rejection after pancreas or islet transplantation.
  • a pharmaceutical composition of this invention can be administered orally or parenterally.
  • parenterally refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion) to a mammal.
  • the pharmaceutical composition will also contain one or more pharmaceutically acceptable diluents, excipients or carriers.
  • Actual dosage levels of the pharmaceutical composition of this invention can be varied so as to obtain an amount of the active compound(s) that is effective to achieve the desired therapeutic response.
  • the selected dosage level will depend upon the route of administration, the severity of the condition being treated, and the condition and prior medical history of the mammal being treated.
  • the components of the subject composition are administered such that they develop their respective effects such that the best protection for the beta cells in the pancreas is ensured.
  • the immunosuppressive compound and/or an anti-inflammatory compound or composition is present in an amount sufficient to limit the mammal's immune response, whereas the diazoxide concentration is increased to a suitable level when the presence of the
  • an appropriate dosage of the pharmaceutical composition of this invention can generally be about 0.01 to 500 mg per kg patient body weight per day which can be administered in single or multiple doses.
  • the dosage level will preferably be about 0.1 to about 250 mg/kg per day, more preferably about 0.5 to about 100 mg/kg per day.
  • this composition can be provided in the form of tablets containing 1.0 to 1000 milligrams of each of the active ingredients, particularly 1.0, 5.0, 10.0, 15.0, 20.0, 25.0, 50.0, 75.0, 100.0, 150.0, 200.0, 250.0, 300.0, 400.0, 500.0, 600.0, 750.0, 800.0, 900.0 and 1000.0 milligrams of each active ingredient.
  • This composition can be administered on a regimen of 1 to 4 times per day, e.g. once or twice per day. The dosage regimen can be adjusted to provide the optimal therapeutic response.
  • compositions of this invention or compositions in the case where the two components are administered separately, for parenteral injection can be in the form of a pharmaceutically acceptable sterile aqueous or non-aqueous solution, dispersion, suspension or emulsion.
  • the composition can also be in the form of a sterile powder for reconstitution into a sterile injectable solution or dispersion just prior to use.
  • aqueous and non-aqueous carriers, diluents, solvents or vehicles examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol and the like), and suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • compositions of this invention can also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Inhibition of the action of microorganisms can be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol or phenol sorbic acid. It can also be desirable to include isotonic agents, such as sugars or sodium chloride. Prolonged absorption of the injectable pharmaceutical form can be effected by the inclusion of agents such as aluminium monostearate and gelatine which delay absorption.
  • composition of this invention it can be desirable to slow the absorption of the composition from subcutaneous or intramuscular injection. This can be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the composition then depends upon its rate of dissolution which, in turn, can depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered composition can be accomplished by dissolving or suspending it in an oil vehicle.
  • Injectable forms of the pharmaceutical composition of this invention can be made by forming microencapsulate matrices of the composition in biodegradable polymers, for example polylactide-polyglycolide. Depending upon the ratio of the composition to the polymer and the nature of the particular polymer employed, the rate of release of the composition can be controlled.
  • biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • Depot injectable formulations can also be prepared by entrapping the composition in a liposome or microemulsion which is compatible with body tissues of the mammal.
  • the injectable formulations can be sterilized, for example, by filtration through a bacterial retaining filter or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable media just prior to use.
  • Solid dosage forms of the pharmaceutical composition or compositions of this invention for oral administration include capsules, tablets, pills, powders and granules.
  • the composition is typically mixed with at least one inert, pharmaceutically acceptable excipient or carrier, such as sodium citrate or dicalcium phosphate and/or one or more: a) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol and silicic acid; b) binders, such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; c) humectants, such as glycerol; d) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; e) solution retarding agents, such as paraffin; f) absorption accelerators, such as quaternary ammonium compounds; g) wetting agents,
  • the solid dosage form can also comprise buffering agents.
  • the composition can also be used as a filler in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycol, for example.
  • Solid dosage forms of the pharmaceutical composition(s) of this invention for oral administration can contain a dissolution aid.
  • dissolution aids include nonionic surface active agents, such as sucrose fatty acid esters, glycerol fatty acid esters, sorbitan fatty acid esters (eg sorbitan trioleate), polyethylene glycol, polyoxyethylene hydrogenated castor oil, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene alkyl ethers, methoxypolyoxyethylene alkyl ethers, polyoxyethylene alkylphenyl ethers, polyethylene glycol fatty acid esters, polyoxyethylene alkylamines, polyoxyethylene alkyl thioethers, polyoxyethylene polyoxypropylene copolymers, polyoxyethylene glycerol fatty acid esters, pentaerythritol fatty acid esters, propylene glycol monofatty acid esters, polyoxyethylene propylene glycol monofatty acid esters, polyoxyethylene sorbitol fatty acid esters
  • Solid dosage forms of the pharmaceutical composition(s) of this invention in the form of tablets, dragees, capsules, pills, or granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art.
  • the coatings can optionally contain opacifying agents and can also be of a composition such that they release the active compound(s) only, or preferentially, in a certain part of the intestinal tract, and/or in delayed fashion.
  • embedding compositions include polymeric substances and waxes.
  • the active compounds of the pharmaceutical composition of this invention can also be in microencapsulated form with one or more of the above-mentioned excipients.
  • the active compounds can also be in finely divided form, for example, they can be micronized.
  • Liquid dosage forms of the pharmaceutical composition of this invention for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms can contain inert diluents commonly used in the art such as water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethyl formamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan and mixtures thereof.
  • the liquid dosage forms ' can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents.
  • Suspensions in addition to the active compounds, can contain suspending agents such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminium metahydroxide, bentonite, agar-agar, and tragacanth and mixtures thereof.
  • the pharmaceutical composition(s) of this invention can also be administered in the form of a liposome.
  • liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono-or multi-lamellar hydrated liquid crystals which are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolisable lipid capable of forming liposomes can be used.
  • the composition in liposome form can contain, in addition to the active compounds of the invention, stabilizers, preservatives, excipients and the like.
  • the preferred lipids are the phospholipids and the phosphatidyl cholines (lecithins), both natural and synthetic. Methods to form liposomes are known in the art, for example, Prescott, Ed., Methods in Cell Biology, Volume XIV, Academic Press, New York, N.Y. (1976), p33 et seq..
  • Formulations of the pharmaceutical composition(s) of this invention can also contain inactive components.
  • Suitable inactive components are well known in the art and are described in standard textbooks, such as Goodman and Gillman's: The Pharmacological Bases of Therapeutics, 8thEd., Gilman et al, Eds. Pergamon Press (1990), and Remington's Pharmaceutical Sciences, 17th Ed., Mack Publishing Co., Easton, Pa. (1990), both of which are incorporated by reference herein in their entirety.
  • Endoc-Beta cells were cultured for 24 hours in placebo or diazoxide (325 mM). After this, a medium change was performed and the Beta cells were then cultured for 24 hours in normoglycemic (5 mM glucose) or glycotoxic (30 mM glucose) conditions. After this a static glucose stimulated insulin secretion test was performed. When Beta cells were incubated in high glucose they suffer from secretory dysfunction (see figure). DZX co-treatment reverses this secretory dysfunction.
  • Figure 1 shows GSIS index in beta cells incubated in 5.5 mM glucose, 30 mM glucose, 30 mM glucose + 325 uM diazoxide.
  • Figures 4A and 4B show a significant reduction in markers for ER stress are increased in hyperglycemic conditions, this increase is mitigated by addition of diazoxide
  • FIGS. 5A and 5B show that markers for OS are increased in hyperglycemic conditions, this increase is mitigated by addition of diazoxide.
  • Isolated human islets are cultured in vitro for 48-72 hours in a control medium containing either methylprednisolone (2 mg/L) or diazoxide (325 pmol/L), or both diazoxide and methylprednisolone.
  • islet damage is assessed with a micro-RNA kit, GAD and PPP IRIA concentrations in the culture medium.
  • Mitochondrial function is assessed by Sea Horse or OUROBOROS.
  • islet functionality is assessed by static and dynamic glucose-stimulated insulin secretion (GSIS) according to standard procedures in the human islet isolation unit.
  • GSIS glucose-stimulated insulin secretion

Landscapes

  • Health & Medical Sciences (AREA)
  • Diabetes (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Endocrinology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Obesity (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Emergency Medicine (AREA)

Abstract

The present invention relates to diazoxide or a pharmaceutically acceptable salt thereof for use as a medicament in combination with one or more immunosuppressive and/or an anti-inflammatory corticosteroid compounds,in a treatment of a mammal afflicted with Type I diabetes, or at risk of developing Type I diabetes.

Description

Compound for Use in Treatment and Prevention of Type I Diabetes
The present invention relates to diazoxide or a pharmaceutically acceptable salt thereof for use as a medicament at low doses to treat type I diabetes and to compositions containing diazoxide for use in the treatment of a mammal afflicted with, or at risk of developing type I diabetes.
BACKGROUND OF THE INVENTION
Diabetes mellitus, commonly referred to as diabetes, is a group of metabolic disorders in which a mammal's blood contains high levels of sugar, also referred to as hyperglycemia, over a prolonged period.
Diabetes is due to a mammal's pancreas not producing enough insulin or the mammal's somatic cells not responding properly to the insulin produced by the mammal.
There are three main types of diabetes mellitus:
Type I results from the failure of the mammal's pancreas to produce enough insulin due to loss of beta cells. Type 2 begins with insulin resistance, a condition in which the mammal's cells fail to respond to insulin properly; as the disease progresses, a lack of insulin can also develop.
Gestational diabetes occurs when pregnant women without a previous history of diabetes develop high blood sugar levels.
US5284845B describes the use of diazoxide for the treatment of disorders caused by defects in glucose metabolism, including hyperglycemia and hypoglycemia and to delay or prevent onset of insulin dependency in Type I diabetic subjects.
EP2422787 describes the use of diazoxide for hypoglycaemia, as well as amyotrophic lateral sclerosis (ALS) and hypertension. In this regard, the principal use of diazoxide in diabetes therapy currently is for the treatment of hyperinsulinemia leading to
hypoglycaemia, wherein hyperinsulinemia is characterised as excess levels of insulin circulating in blood relative to glucose.
There has been an unfilled need, therefore, for a treatment of the onset and progression of Type I diabetes which will substantially prevent further deterioration of beta cells in the mammal's pancreas.
Related to this, there has been an unfilled need for a treatment to protect beta cells during and shortly after islet transplantation against loss due to ischemia and against loss due to rejection after pancreas or islet transplantation.
Summary of the Invention Accordingly, in a first aspect, the present invention relates to diazoxide or a pharmaceutically acceptable salt thereof for use as a medicament in combination with an immunosuppressive compound and/or an anti-inflammatory compound, preferably a corticosteroid, in a treatment of a mammal afflicted with Type I diabetes. Preferably, the treatment substantially prevents further deterioration of beta cells in the mammal's pancreas and/or protects beta cells in the mammal's pancreas during and shortly after islet transplantation against loss due to ischemia and/or protects beta cells in the mammal's pancreas against loss due to rejection after pancreas or islet transplantation.
In a second aspect, the present invention relates to a pharmaceutical composition comprising diazoxide or a pharmaceutically acceptable salt thereof and an
immunosuppressive compound and/or an anti-inflammatory compound, preferably a corticosteroid, for use in the treatment of a mammal afflicted with, or prone to develop Type I diabetes.
Detailed description of the invention
The present invention relates to the use of diazoxide for preventing or treating the onset and progression of Type I diabetes when a large mass of beta cells is still present in the mammal's pancreas which will likely quickly deteriorate. Applicants have surprisingly found that the administration of diazoxide, preferably in combination with an
immunosuppressive compound and/or an anti-inflammatory compound, preferably a corticosteroid, may protect beta cells from the auto-immune reaction considered as the underlying cause for this ailment.
Accordingly, the present invention in a first instance relates to a composition comprising diazoxide for the protection of beta cells.
In a second aspect, the present invention relates to a process wherein diazoxide is used as a beta cell protective agent, combined with an immunosuppressive compound and/or the anti-inflammatory compound, preferably a corticosteroid.
Preferably, the process relates to a treatment with 100 - 1600mg per day, more preferably 200 - 1200 mg per day, more preferably of from 300 to 800mg per day of diazoxide.
Preferably, the process also relates to a treatment with one or more
immunosuppressive compounds and/or one or more anti-inflammatory compounds selected from the group consisting of: • a corticosteroid, particularly methylprednisolone (dose range 5 - 2000 mg per day), prednisolone (dose range 1,25 - 120 mg per day), dexamethasone (0.25 - 24 mg per day), cortisone (10 - 1000 mg per day), or hydrocortisone (10 - 1000 mg per day), or
combinations thereof, and
• a calcineurin-inhibitor, particularly tacrolimus (dose range 0.5 - 24 mg per day) or cyclosporine and its derivatives (dose range 25 - 750mg per day);
• an antimetabolite, particularly azathioprine and its derivatives (dose range 25 - 1000 mg per day), mycophenolate mofetil (dose range 250 - 4000 mg per day); mycophenolic acid (dose range 180 - 2880 mg per day,) and/or methotrexate (dose range 0.5 - 20 mg per day);
• an mTOR inhibitor, particularly everolimus (dose range 0.25 - 10 mg per day) or sirolimus (dose range 0.5 - 80 mg per day);
• an anti-lymphocyte antibody, particularly alemtuzumab (dose range 0.1 - 100 mg), rituximab (200 - 2000 mg) or thymoglobulin (dose range 10 - 500 mg per day); and/or
• an anti-cytokine antibody, particularly basiliximab (dose range 10 - 100 mg), anakinra (dose range 10 - 500 mg per day), tocilizumab (dose range 8 - 1600 mg), etanercept (dose range 1 - 10 mg per day), Adalimumab (dose range 5 -640 mg), infliximab (dose range 5 - 2000 mg), golimumab (dose range 25 - 800 mg), certolizumab (dose range 50 - 1600 mg).
More preferably, the process also relates to a treatment with one or more immunosuppressive compounds and/or one or more anti-inflammatory compounds selected from the group consisting of:
• a corticosteroid, particularly methylprednisolone (dose range 5 - 2000 mg per day), prednisolone (dose range 1,25 - 120 mg per day), dexamethasone (0.25 - 24 mg per day), cortisone (10 - 1000 mg per day), or hydrocortisone (10 - 1000 mg per day), or
combinations thereof.
Short Description of the Figures
Fig 1 discloses the stimulation index in presence of diazoxide or a placebo compounds.
Fig 2 discloses the stimulation index in presence of diaozixe or a placebo compounds for Human isolated islets of Langerhans.
Figure 3 discloses Dynamic GSIS of islets incubated in diazoxide shows an improved insulin secretion profile in human islets incubated in diazoxide 325uM as compared to placebo) Figures 4A and 4B shows that markers for ER stress are increased in hyperglycemic conditions, this increase is mitigated by addition of diazoxide.
Figure 5A and 5B shows that markers for OS are increased in hyperglycemic conditions, this increase is mitigated by addition of diazoxide.
Detailed Description of the Invention
Without wishing to relate to any particular theory, it is believed that this
combination therapy may protect the beta cell from both the beta cell stress and the inflammation. A first situation whereby the combination may be applied is during the onset and progression of Type I diabetes, i.e. when a majority or at least large group of beta cells is still present, but shows a fast deterioration, whereby applicants have found that diazoxide treatment ameliorates this deterioration.
More preferably, by also treating the ongoing insulitis, by combining the diazoxide with an immunosuppressive compound and/or an anti-inflammatory compound, in particular a corticosteroid, these beta cells may be kept alive.
A second situation in which beta cell protection may be applied is during, and shortly after islet transplantation, since transplanted islets require several weeks to be fully vascularized and a large portion of transplanted islets are lost due to ischemia, whereby the combination therapy may advantageously be employed.
A third situation in which beta cell protection may be applied is in the case of a rejection after pancreas or islet transplantation.
A fourth situation in which beta cell protection may be applied is in the case of a person having a substantially increased risk of developing Type I diabetes, as indicated by biomarkers or genetic predisposition.
In accordance with this invention, diazoxide or a pharmaceutically acceptable salt thereof is used as a medicament in combination with one or more immunosuppressive compounds and/or anti-inflammatory compounds, particularly one or more corticosteroids, in a treatment of a mammal afflicted with Type I diabetes or the onset thereof, and/or at risk of developing Type I diabetes.
A pharmaceutical composition comprising diazoxide or a pharmaceutically acceptable salt thereof and the one or more immunosuppressive compounds and/or anti inflammatory compounds, particularly one or more corticosteroids, as the active
compounds is preferably administered to the mammal in this treatment. As used herein, the term "corticosteroid" preferably means a glucocorticoid, more preferably a natural glucocorticoid or a synthetic corticosteroid-like compound, such as betamethasone, prednisone, dexamethasone, cortisone, hydrocortisone,
methylprednisolone and prednisolone, as well as amcinonide, betamethosone
diproprionate, clobetasol, clocortolone, diflorasone, dutasteride, flumethasone pivalate, flunisolide, fluocinolone acetonide, fluocinonide, fluorometholone, fluticasone propionate, fluticasone propionate, fluticasone propionate, flurandrenolide and hydroflumethiazide
As also used herein, the term "mammal" preferably means a primate, a human or a domestic or farm animal, such as a cow, horse, pig, sheep, goat, dog, cat or rodent, more preferably a human.
As also used herein, the term "pharmaceutically acceptable salt thereof" preferably means a free compound or, for example, a pharmaceutically acceptable, non-toxic organic or inorganic acid or base addition salt of diazoxide.
As also used herein, the term "mammal ... at risk of developing Type I diabetes" preferably means a mammal testing positive for one or more diabetes-associated antibodies and/or with a positive family history for Type I diabetes in one or more first-degree relatives and/or with an increased risk for Type I diabetes based on gene analysis thereof as described by: Redondo MJ, Oram RA, Steck AK. Genetic Risk Scores for Type I Diabetes Prediction and Diagnosis. Curr Diab Rep, 2017; Regnell SE, Lernmark A. Early prediction of autoimmune (Type I) diabetes. Diabetologia, 2017; Bonifacio E. Predicting Type I diabetes using biomarkers. Diabetes Care 2015.
In accordance with this invention, diazoxide or a pharmaceutically acceptable salt thereof and an immunosuppressive compound and/or an anti-inflammatory compound, particularly a corticosteroid, or preferably a pharmaceutical composition thereof can be administered to the mammal orally, topically, intravenously, subcutaneously, buccally, rectally, dermally, nasally, tracheally, bronchially, by any other parenteral route, as an oral or nasal spray or via inhalation. Depending upon the mammal to be treated and the route of administration, varying doses of a pharmaceutical composition of this invention comprising diazoxide or a pharmaceutically acceptable salt thereof and the
immunosuppressive compound and/or anti-inflammatory compound, particularly the corticosteroid, can be suitably administered. Preferably, this treatment substantially prevents further deterioration of beta cells in the mammal's pancreas and/or protects beta cells in the mammal's pancreas during and shortly after islet transplantation against loss due to ischemia and/or protects beta cells in the mammal's pancreas against loss due to rejection after pancreas or islet transplantation.
Typically, a pharmaceutical composition of this invention can be administered orally or parenterally. The term "parenterally" as used herein, refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion) to a mammal. Generally, the pharmaceutical composition will also contain one or more pharmaceutically acceptable diluents, excipients or carriers.
Actual dosage levels of the pharmaceutical composition of this invention can be varied so as to obtain an amount of the active compound(s) that is effective to achieve the desired therapeutic response. The selected dosage level will depend upon the route of administration, the severity of the condition being treated, and the condition and prior medical history of the mammal being treated.
Preferably, the components of the subject composition are administered such that they develop their respective effects such that the best protection for the beta cells in the pancreas is ensured. Without wishing to refer to any particular theory, it is believed that the immunosuppressive compound and/or an anti-inflammatory compound or composition is present in an amount sufficient to limit the mammal's immune response, whereas the diazoxide concentration is increased to a suitable level when the presence of the
immunosuppressive compound and/or an anti-inflammatory compound is ensured. In this way, it believed that the subject composition develops a strongly synergistic effect.
In the treatment of a mammal afflicted with, or at risk of developing Type I diabetes, an appropriate dosage of the pharmaceutical composition of this invention can generally be about 0.01 to 500 mg per kg patient body weight per day which can be administered in single or multiple doses.
The dosage level will preferably be about 0.1 to about 250 mg/kg per day, more preferably about 0.5 to about 100 mg/kg per day. For oral administration, this composition can be provided in the form of tablets containing 1.0 to 1000 milligrams of each of the active ingredients, particularly 1.0, 5.0, 10.0, 15.0, 20.0, 25.0, 50.0, 75.0, 100.0, 150.0, 200.0, 250.0, 300.0, 400.0, 500.0, 600.0, 750.0, 800.0, 900.0 and 1000.0 milligrams of each active ingredient. This composition can be administered on a regimen of 1 to 4 times per day, e.g. once or twice per day. The dosage regimen can be adjusted to provide the optimal therapeutic response.
The pharmaceutical composition of this invention or compositions in the case where the two components are administered separately, for parenteral injection can be in the form of a pharmaceutically acceptable sterile aqueous or non-aqueous solution, dispersion, suspension or emulsion. The composition can also be in the form of a sterile powder for reconstitution into a sterile injectable solution or dispersion just prior to use.
Examples of suitable aqueous and non-aqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol and the like), and suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
The pharmaceutical composition or compositions of this invention can also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Inhibition of the action of microorganisms can be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol or phenol sorbic acid. It can also be desirable to include isotonic agents, such as sugars or sodium chloride. Prolonged absorption of the injectable pharmaceutical form can be effected by the inclusion of agents such as aluminium monostearate and gelatine which delay absorption.
To prolong the effects of the pharmaceutical composition of this invention, it can be desirable to slow the absorption of the composition from subcutaneous or intramuscular injection. This can be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the composition then depends upon its rate of dissolution which, in turn, can depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered composition can be accomplished by dissolving or suspending it in an oil vehicle.
Injectable forms of the pharmaceutical composition of this invention can be made by forming microencapsulate matrices of the composition in biodegradable polymers, for example polylactide-polyglycolide. Depending upon the ratio of the composition to the polymer and the nature of the particular polymer employed, the rate of release of the composition can be controlled. Examples of biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations can also be prepared by entrapping the composition in a liposome or microemulsion which is compatible with body tissues of the mammal. The injectable formulations can be sterilized, for example, by filtration through a bacterial retaining filter or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable media just prior to use.
Solid dosage forms of the pharmaceutical composition or compositions of this invention for oral administration include capsules, tablets, pills, powders and granules. In such solid dosage forms, the composition is typically mixed with at least one inert, pharmaceutically acceptable excipient or carrier, such as sodium citrate or dicalcium phosphate and/or one or more: a) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol and silicic acid; b) binders, such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; c) humectants, such as glycerol; d) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; e) solution retarding agents, such as paraffin; f) absorption accelerators, such as quaternary ammonium compounds; g) wetting agents, such as cetyl alcohol and glycerol monostearate; h) absorbents, such as kaolin and bentonite clay and i) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate and mixtures thereof. In the capsules, tablets and pills, the solid dosage form can also comprise buffering agents. The composition can also be used as a filler in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycol, for example.
Solid dosage forms of the pharmaceutical composition(s) of this invention for oral administration can contain a dissolution aid. Examples of dissolution aids include nonionic surface active agents, such as sucrose fatty acid esters, glycerol fatty acid esters, sorbitan fatty acid esters (eg sorbitan trioleate), polyethylene glycol, polyoxyethylene hydrogenated castor oil, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene alkyl ethers, methoxypolyoxyethylene alkyl ethers, polyoxyethylene alkylphenyl ethers, polyethylene glycol fatty acid esters, polyoxyethylene alkylamines, polyoxyethylene alkyl thioethers, polyoxyethylene polyoxypropylene copolymers, polyoxyethylene glycerol fatty acid esters, pentaerythritol fatty acid esters, propylene glycol monofatty acid esters, polyoxyethylene propylene glycol monofatty acid esters, polyoxyethylene sorbitol fatty acid esters, fatty acid alkylolamides, and alkyamine oxides; bile acid and salts thereof (eg chenodeoxycholic acid, cholic acid, deoxycholic acid, dehydrocholic acid and salts thereof, and glycine or taurine conjugate thereof); ionic surface active agents, such as sodium laurylsulfate, fatty acid soaps, alkylsufonates, alkylphosphates, ether phosphates, fatty acid salts of basic amino acids; triethanolamine soap, and alkyl quaternary ammonium salts; and amphoteric surface active agents, such as betaines and aminocarboxylic acid salts.
Solid dosage forms of the pharmaceutical composition(s) of this invention in the form of tablets, dragees, capsules, pills, or granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. The coatings can optionally contain opacifying agents and can also be of a composition such that they release the active compound(s) only, or preferentially, in a certain part of the intestinal tract, and/or in delayed fashion. Examples of embedding compositions include polymeric substances and waxes.
The active compounds of the pharmaceutical composition of this invention can also be in microencapsulated form with one or more of the above-mentioned excipients. The active compounds can also be in finely divided form, for example, they can be micronized.
Liquid dosage forms of the pharmaceutical composition of this invention for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms can contain inert diluents commonly used in the art such as water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethyl formamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan and mixtures thereof. Besides inert diluents, the liquid dosage forms 'can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents. Suspensions, in addition to the active compounds, can contain suspending agents such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminium metahydroxide, bentonite, agar-agar, and tragacanth and mixtures thereof.
The pharmaceutical composition(s) of this invention can also be administered in the form of a liposome. As is known in the art, liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono-or multi-lamellar hydrated liquid crystals which are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolisable lipid capable of forming liposomes can be used. The composition in liposome form can contain, in addition to the active compounds of the invention, stabilizers, preservatives, excipients and the like. The preferred lipids are the phospholipids and the phosphatidyl cholines (lecithins), both natural and synthetic. Methods to form liposomes are known in the art, for example, Prescott, Ed., Methods in Cell Biology, Volume XIV, Academic Press, New York, N.Y. (1976), p33 et seq..
Formulations of the pharmaceutical composition(s) of this invention can also contain inactive components. Suitable inactive components are well known in the art and are described in standard textbooks, such as Goodman and Gillman's: The Pharmacological Bases of Therapeutics, 8thEd., Gilman et al, Eds. Pergamon Press (1990), and Remington's Pharmaceutical Sciences, 17th Ed., Mack Publishing Co., Easton, Pa. (1990), both of which are incorporated by reference herein in their entirety.
The following, non-limiting example section illustrates the invention further.
Endoc-Beta cells were cultured for 24 hours in placebo or diazoxide (325 mM). After this, a medium change was performed and the Beta cells were then cultured for 24 hours in normoglycemic (5 mM glucose) or glycotoxic (30 mM glucose) conditions. After this a static glucose stimulated insulin secretion test was performed. When Beta cells were incubated in high glucose they suffer from secretory dysfunction (see figure). DZX co-treatment reverses this secretory dysfunction. Figure 1 shows GSIS index in beta cells incubated in 5.5 mM glucose, 30 mM glucose, 30 mM glucose + 325 uM diazoxide.
Example 2
Human isolated islets of Langerhans were cultured for 24 hours in placebo or diazoxide (325 mM). After this, a medium change was performed and the islets were then cultured for 96 hours in normoglycemic (5 mM glucose), mild hyperglycemic (11 mM glucose) or glycotoxic (20 mM glucose) conditions. After this a static glucose stimulated insulin secretion test was performed. When human islets are incubated in high glucose they suffer from secretory dysfunction. DZX co-treatment reverses this secretory dysfunction (Figure: GSIS index in human islets incubated in 5 mM glucose, 11 mM glucose, 20 mM glucose without and with 325 uM diazoxide. Example 3:
Human isolated islets of Langerhans were cultured in diazoxide (325 mM) or placebo for 48 hours, and then in normal medium for 24 hours. After this, a dynamic GSIS was performed. Figure 3 shows the effect on Dynamic GSIS of islets incubated in diazoxide shows an improved insulin secretion profile in human islets incubated in diazoxide 325uM as compared to placebo.
Figure imgf000012_0001
Human isolated islets of Langerhans were cultured for 24 hours in placebo or diazoxide (325 mM). After this, a medium change was performed and the islets were then cultured for 96 hours in normoglycemic (5 mM glucose), mild hyperglycemic (11 mM glucose) or glycotoxic (20 mM glucose) conditions. After this, markers for endoplasmic reticulum (ER) stress (ATF3, XBPls/XBPlu) were measured by qPCR.
Figures 4A and 4B show a significant reduction in markers for ER stress are increased in hyperglycemic conditions, this increase is mitigated by addition of diazoxide)
Example 5:
Human isolated islets of Langerhans were cultured for 24 hours in placebo or diazoxide (325 mM). After this, a medium change was performed and the islets were then cultured for 96 hours in normoglycemic (5 mM glucose), mild hyperglycemic (11 mM glucose) or glycotoxic (20 mM glucose) conditions. After this, markers for oxidative stress (OS) (TXIP, SOD2) were measured by qPCR.
Figure 5A and 5B show that markers for OS are increased in hyperglycemic conditions, this increase is mitigated by addition of diazoxide.
Example 6
Isolated human islets are cultured in vitro for 48-72 hours in a control medium containing either methylprednisolone (2 mg/L) or diazoxide (325 pmol/L), or both diazoxide and methylprednisolone.
After culturing, viability of the human islets is tested by FDA-PI and PrestoBlue. Islet damage is assessed with a micro-RNA kit, GAD and PPP IRIA concentrations in the culture medium. Mitochondrial function is assessed by Sea Horse or OUROBOROS. In addition, islet functionality is assessed by static and dynamic glucose-stimulated insulin secretion (GSIS) according to standard procedures in the human islet isolation unit. Human islets cultured in vitro in the control medium containing both diazoxide and methylprednisolone were shown to be significantly more viable than those culture in only diazoxide or methylprednisolone.

Claims

Claims
1. Diazoxide or a pharmaceutically acceptable salt thereof for use as a medicament in a treatment of a mammal afflicted with, or at risk of developing Type I diabetes, and/or to substantially prevent deterioration of beta cells in the mammal's pancreas.
2. Diazoxide or a pharmaceutically acceptable salt thereof according to claim 1, in
combination with one or more immunosuppressive and/or an anti-inflammatory compound, for use as a medicament in a treatment of a mammal afflicted with, or at risk of developing Type I diabetes, and/or to substantially prevent deterioration of beta cells in the mammal's pancreas.
3. Diazoxide or a pharmaceutically acceptable salt thereof according to claim 1 or claim 2, in combination with a corticosteroid compound, for use as a medicament in a treatment of a mammal afflicted with, or at risk of developing Type I diabetes, and/or to substantially prevent deterioration of beta cells in the mammal's pancreas.
4. Diazoxide or a pharmaceutically acceptable salt thereof for use as a medicament to protect beta cells in the mammal's pancreas during and shortly after islet
transplantation against loss due to ischemia.
5. Diazoxide or a pharmaceutically acceptable salt thereof according to claim 4, in combination with one or more immunosuppressive and/or an anti-inflammatory corticosteroid compounds, for use as a medicament to protect beta cells in the mammal's pancreas during and shortly after islet transplantation against loss due to ischemia.
6. Diazoxide or a pharmaceutically acceptable salt thereof for use as a medicament in combination with one or more immunosuppressive and/or an anti-inflammatory corticosteroid compound, in accordance with any one of claims 1 to 5, to protect beta cells in the mammal's pancreas against loss due to rejection after pancreas or islet transplantation.
7. A composition for use according to any one of claims 1 to 6, wherein the
immunosuppressive compound and/or anti-inflammatory compound comprises one or more corticosteroids, and one or more additional compounds.
8. A composition for use according to claim 7, wherein the additional compounds
include insulin at a concentration suitable to stabilize the plasma glucose
concentration of the mammal.
9. Composition for use according to any one of claims 1 to 8, wherein the one or more corticosteroid compounds comprises a natural glucocorticoid or a synthetic corticosteroid-like compound, preferably betamethasone, dexamethasone, cortisone, hydrocortisone, methylprednisolone and prednisolone, as well as amcinonide, betamethosone diproprionate, clobetasol, clocortolone, diflorasone, dutasteride, flumethasone pivalate, flunisolide, fluocinolone acetonide, fluocinonide, fluorometholone, fluticasone propionate, fluticasone propionate, fluticasone propionate, flurandrenolide and hydroflumethiazide, in a suitable dosage.
10. Composition for use according to any one of claims 1 to 9, wherein the
corticosteroid is methylprednisolone , at a preferred dose range of from 5 - 2000mg per day, prednisolone, preferably at a dose range of from 1,25 - 120mg per day, dexamethasone, preferably at a dose range of from 0.25 - 24mg per day, cortisone, preferably at a dose range of from 10 - lOOOmg per day, or hydrocortisone, preferably at a dose range of from 10 - lOOOmg per day.
11. Composition for use according to any one of claims 1 to 10 , wherein the components are administered sequentially, or simultaneously.
12. A pharmaceutical composition comprising diazoxide or a pharmaceutically
acceptable salt thereof and one or more immunosuppressive and/or anti inflammatory corticosteroid compounds according to any one of claims 1 to 11 for use in the treatment of a mammal afflicted with, or at risk of developing Type I diabetes.
13. Method for the treatment of a mammal afflicted with Type I diabetes, the method comprising the step of administering an effective amount of diazoxide or a pharmaceutically acceptable salt thereof and one or more immunosuppressive and/or anti-inflammatory corticosteroid compounds, according to any one of claims 1 to 12 to the mammal.
14. The method according to claim 13, wherein diazoxide is administered first, followed by administration of the one or more immunosuppressive and/or anti-inflammatory corticosteroid compounds.
15. The method according to claim 13 or 14, wherein insulin is administered to stabilize plasma glucose concentrations of the subject under treatment.
16. A composition according to any one of claims 7 to 12, for the preparation of a
medicament for the treatment or prevention of Diabetes Type I in a mammal.
PCT/NL2019/050872 2018-12-21 2019-12-21 Compound for use in treatment and prevention of type i diabetes WO2020130836A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
NL2022291 2018-12-21
NL2022291A NL2022291B1 (en) 2018-12-21 2018-12-21 Compound for Use in Treatment and Prevention of Type I Diabetes
NL2024401 2019-12-06
NL2024401 2019-12-06

Publications (1)

Publication Number Publication Date
WO2020130836A1 true WO2020130836A1 (en) 2020-06-25

Family

ID=69106163

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2019/050872 WO2020130836A1 (en) 2018-12-21 2019-12-21 Compound for use in treatment and prevention of type i diabetes

Country Status (1)

Country Link
WO (1) WO2020130836A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5284845A (en) 1991-03-14 1994-02-08 Paulsen Elsa P Use of oral diazoxide for the treatment of disorders in glucose metabolism
WO2002000665A1 (en) * 2000-06-26 2002-01-03 Novo Nordisk A/S Use of potassium channel openers for the treatment of insulitis
WO2003105896A1 (en) * 2002-06-14 2003-12-24 Novo Nordisk A/S Combined use of a modulator of cd3 and a beta cell resting compound
WO2006045799A2 (en) * 2004-10-25 2006-05-04 Solvay Pharmaceuticals Gmbh Pharmaceutical compositions comprising cb1 cannabinoid receptor antagonists and potassium channel openers for the treatment of diabetes mellitus type i, obesity and related conditions
EP2422787A1 (en) 2010-08-17 2012-02-29 Neurotec Pharma, S.L. Diazoxide for use in the treatment of amyotrophic lateral sclerosis (als)

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5284845A (en) 1991-03-14 1994-02-08 Paulsen Elsa P Use of oral diazoxide for the treatment of disorders in glucose metabolism
WO2002000665A1 (en) * 2000-06-26 2002-01-03 Novo Nordisk A/S Use of potassium channel openers for the treatment of insulitis
WO2003105896A1 (en) * 2002-06-14 2003-12-24 Novo Nordisk A/S Combined use of a modulator of cd3 and a beta cell resting compound
WO2006045799A2 (en) * 2004-10-25 2006-05-04 Solvay Pharmaceuticals Gmbh Pharmaceutical compositions comprising cb1 cannabinoid receptor antagonists and potassium channel openers for the treatment of diabetes mellitus type i, obesity and related conditions
EP2422787A1 (en) 2010-08-17 2012-02-29 Neurotec Pharma, S.L. Diazoxide for use in the treatment of amyotrophic lateral sclerosis (als)

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
"Goodman and Gillman's: The Pharmacological Bases of Therapeutics", 1990, MACK PUBLISHING CO.
"Methods in Cell Biology", vol. XIV, 1976, ACADEMIC PRESS, pages: 33
ANDREANI: "Prediction, prevention, and early intervention in insulin-dependent diabetes", DIABETES/METABOLISM REVIEWS, 7(1), 61-77, 1 January 1991 (1991-01-01), XP055672590, Retrieved from the Internet <URL:https://onlinelibrary.wiley.com/doi/pdfdirect/10.1002/dmr.5610070107> [retrieved on 20200302] *
ATHANASIOS FOUNTAS ET AL: "Severe resistant hypoglycemia in a patient with a pancreatic neuroendocrine tumor on sunitinib treatment", HORMONES, 15 December 2014 (2014-12-15), XP055607857, ISSN: 1109-3099, DOI: 10.14310/horm.2002.1560 *
BONIFACIO E: "Predicting Type I diabetes using biomarkers", DIABETES CARE, 2015
E BJORK ET AL: "Rapid Publications Diazoxide Treatment at Onset Preserves Residual Insulin Secretion in Adults with Autoimmune Diabetes", UPPSALA, AND THE DEPARTMENT OF MEDICINE, DANDERYD HOSPI-TAL, 1 January 1996 (1996-01-01), XP055672568, Retrieved from the Internet <URL:https://diabetes.diabetesjournals.org/content/diabetes/45/10/1427.full.pdf> [retrieved on 20200302] *
EVA ÖRTQVIST ET AL: "Temporary Preservation of B-Cell Function by Diazoxide Treatment in Childhood Type 1 Diabetes", DIABETES CARE 2004, 27:2191-2197, 1 January 2004 (2004-01-01), XP055672740, Retrieved from the Internet <URL:https://care.diabetesjournals.org/content/diacare/27/9/2191.full.pdf> [retrieved on 20200302] *
HIRAMATSU ET AL: "Treatment with diazoxide causes prolonged improvement of @b-cell function in rat islets transplanted to a diabetic environment", METABOLISM, CLINICAL AND EXPERIMEN, W.B. SAUNDERS CO., PHILADELPHIA, PA, US, vol. 49, no. 5, 1 May 2000 (2000-05-01), pages 657 - 661, XP005302185, ISSN: 0026-0495, DOI: 10.1016/S0026-0495(00)80044-X *
JENNIFER B MARKSF ET AL: "Clinical Review 17 Immunotherapy of Type I Diabetes Mellitus* Type I Diabetes-A Chronic Immune-Mediated Disease", JOURNAL OF CLINICAL ENDOCRINOLOGY AND METABOLISM COPYRIGHT THE ENDOCRINE SOCIETY, 1 January 1991 (1991-01-01), XP055672839, Retrieved from the Internet <URL:https://watermark.silverchair.com/jcem0003.pdf?token=AQECAHi208BE49Ooan9kkhW_Ercy7Dm3ZL_9Cf3qfKAc485ysgAAAlYwggJSBgkqhkiG9w0BBwagggJDMIICPwIBADCCAjgGCSqGSIb3DQEHATAeBglghkgBZQMEAS4wEQQMVpgmA-YT_jf5ecmoAgEQgIICCQEO7hgDRV3Ab78NCn-JhOzarVe3nJlgRkUpvJ9hKkrkCKp_m-28mYlSlN8kiOQys7-m90xS2DgwDG0JsF8QLypx35x> *
PRIYA S. GEORGE ET AL: "Diazoxide Improves Hormonal Counterregulatory Responses to Acute Hypoglycemia in Long-standing Type 1 Diabetes", DIABETES, vol. 64, no. 6, 15 January 2015 (2015-01-15), US, pages 2234 - 2241, XP055672916, ISSN: 0012-1797, DOI: 10.2337/db14-1539 *
REDONDO MJORAM RASTECK AK: "Genetic Risk Scores for Type I Diabetes Prediction and Diagnosis", CURR DIAB REP, 2017
REGNELL SELERNMARK A: "Early prediction of autoimmune (Type I) diabetes", DIABETOLOGIA, 2017
YONG WANG ET AL: "Diazoxide, a K ATP Channel Opener, Prevents Ischemia-Reperfusion Injury in Rodent Pancreatic Islets", CELL TRANSPLANTATION, vol. 24, no. 1, 1 January 2015 (2015-01-01), US, pages 25 - 36, XP055672615, ISSN: 0963-6897, DOI: 10.3727/096368913X673441 *

Similar Documents

Publication Publication Date Title
US20030232867A1 (en) Use
Ricordi et al. In vivo effect of FK506 on human pancreatic islets
Remuzzi et al. Renal vascular and thrombotic effects of cyclosporine
Ochiai et al. Studies of the effects of FK506 on renal allografting in the beagle dog
US20210052578A1 (en) Treatment of diseases
Forbes et al. Carbamazepine hepatotoxicity: another cause of the vanishing bile duct syndrome
EP1429845B1 (en) Treatment or prophylaxis of insulin-producing cell graft rejection
CZ285660B6 (en) Use of rapamycin for preparing medicament intended for treating ophthalmia in mammals
KR20150108946A (en) Methods and compositions for treating cancer
JP2002502824A (en) Costimulation blockade and mixed chimerism in allografts
EP2326332B1 (en) Method for treatment of diseases
US8450281B2 (en) Non-immunosuppressive cyclosporin for treatment of Ullrich congenital muscular dystrophy
US7659249B2 (en) Membrane-permeable NFAT inhibitory peptide
US20110142953A1 (en) Combinations of immunosuppressive agents for the treatment or prevention of graft rejections
NL2022291B1 (en) Compound for Use in Treatment and Prevention of Type I Diabetes
WO2020130836A1 (en) Compound for use in treatment and prevention of type i diabetes
KR102014349B1 (en) Immunosuppression composition comprising JAK inhibitor
US20240101671A1 (en) Method and composition for inducing tolerance
KR20070089701A (en) Treatment of graft-versus-host disease and leukemia with beclomethasone dipropionate and prednisone
WO2017209270A1 (en) Activated t cell- and/or b cell-selective cell death inducer or cell death promoter comprising as active ingredient 25-hydroxycholesterol or cholesterol analogous thereto
US20190201490A1 (en) Methods and compositions for the treatment of secretory disorders
EP1499330A1 (en) Arsenic therapy for apls-type autoimmune lymph-oproliferative syndrome in mice and humans
JP2007510721A (en) CCI-779 for treating mantle cell lymphoma
JP6878418B2 (en) How to treat graft rejection
US3591690A (en) Method of treatment of diarrhea in piglet of 2-4 weeks of age

Legal Events

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

Ref document number: 19832218

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19832218

Country of ref document: EP

Kind code of ref document: A1