WO2002000665A1 - Use of potassium channel openers for the treatment of insulitis - Google Patents

Use of potassium channel openers for the treatment of insulitis Download PDF

Info

Publication number
WO2002000665A1
WO2002000665A1 PCT/DK2001/000444 DK0100444W WO0200665A1 WO 2002000665 A1 WO2002000665 A1 WO 2002000665A1 DK 0100444 W DK0100444 W DK 0100444W WO 0200665 A1 WO0200665 A1 WO 0200665A1
Authority
WO
WIPO (PCT)
Prior art keywords
thiadiazine
thieno
chloro
dioxide
potassium channel
Prior art date
Application number
PCT/DK2001/000444
Other languages
French (fr)
Inventor
John Bondo Hansen
Anders Karlsson
Mikael Kullin
Stellan Sandler
Elisabeth BJÖRK
Zhanchun Li
Birgitte Michelsen
Jesper Svendstorp Lund
Original Assignee
Novo Nordisk A/S
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novo Nordisk A/S filed Critical Novo Nordisk A/S
Priority to AU2001265841A priority Critical patent/AU2001265841A1/en
Publication of WO2002000665A1 publication Critical patent/WO2002000665A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/12Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • 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

Definitions

  • the present invention relates to the use of potassium channel openers, which are able to protect the beta cells against toxic damage, for treating or preventing diseases related to autoimmune destruction of human beta cells, such as different types of diabetes, and methods of using these compounds.
  • BACKGROUND OF THE INVENTION Streptozotocin and alloxan are beta cell toxins.
  • the toxic effect of these compounds on rat pancreatic islets in vitro and in vivo mimics the beta-cell death associated with Type 1 and late state Type 2 diabetes.
  • the compounds of the present invention are able to inhibit strep- tozotocin and alloxan induced beta cell degeneration and death.
  • the compounds of the present invention act as activators of ATP regulated potassium channels (Katp-channels) of the beta cell and the Katp- channels of mitochondria. They may also act by antagonising the depletion of NAD induced in the islets by these toxins. Cytokines are known to reduce beta cell viability and to induce apoptosis. Cytokines have been proposed to be involved with the autoimmune degeneration of beta cells in Type 1 diabetes. The compounds of the present invention antagonize the effects of cytokines on beta cells.
  • the compounds of the present invention can be used in the treatment of insulitis associated with various forms of diabetes.
  • IDDM Type 1 or Insulin Dependent Diabetes Mellitus
  • NIDDM Non Insulin Dependent Diabetes Mellitus
  • SPIDDM slowly progressive IDDM
  • LADA latent autoimmune diabetes in adults
  • gestational diabetes due to underlying IDDM.
  • potassium channel openers are compounds disclosed in PCT Publication No. WO 97/26265 (see for instance from page 14, line 5 to page 19, line 9) and WO 99/03861 (see for instance from page 17, line 20 to page 19, line 5) as well as the following com- pounds: 3-tert-Butylamino-6-chloro-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide; 6-Chloro- 3-cyclobutylamino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 , 1 -dioxide; 6-Chloro-3-(1 , 1 -dimethyl- propylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 , 1 -dioxide; 6-Chloro-3-(1 -methylcyclo- propyl)amino-4H-thieno[3,2-e]-1 ,2,4-thiadia
  • the remaining islets showed reduced insulin content and secretion and a reduced insulin biosynthesis, amounting to 50%, 60% and 35%, respectively of control.
  • the STZ islets also displayed a lowered rate of glucose oxidation - 16% of control.
  • islets pre-incubated with diazoxide or 6-Chloro-3-isopropylamino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide maintained higher insulin content and insulin secretion compared to islets incu- bated with STZ alone.
  • K A TP channel openers can protect insulin-producing cells from being damaged by a beta-cell toxin and suggest that such an effect might be applicable in subjects with ongoing insulitis.
  • Diazoxide and other K ATP channel openers such as cromakalim and pinacidil, have been employed in experimental studies of ischemic heart. A beneficial, cardioprotective effect was observed (Garlid KD et al., Circulation Res 1997; 81 :1072-82). Although the mechanism of this phenomenon is not understood, an opening of mitochondnal potassium channels seems to be involved, resulting in dissipation of the inner mitochondnal membrane potential. This in turn leads to net oxidation of the mitochondria with an apparent reduction of energy wastage.
  • Diazoxide is known to act on K ATP channels in the plasma membrane of beta cells. It hyper- polarizes the membrane and reduces the entry of Ca 2+ , essential for the exocytosis of secre- tory granulaes. Recently, exposure of beta cells to diazoxide was found to engage also mitochondnal K AT p channels (Grimmsmann T et al., Br J Pharmacol 1998; 123:781-788). In the present study, we examined the influence of potassium channel openers on experimental beta-cell damage induced by streptozotocin, an agent known to cause energy depletion, on damage induced by alloxan, a generator of reactive oxygen species and on damage induced by cytokines.
  • Pancreata from Sprague-Dawley rats were collagenase digested and islets collected with a braking pipette as previously described (Sandier S et al., Endocrinology, 1987;121 :1424-31). Islets were precultured free floating in RPM1 1640 medium with 10 % (v/v) fetal calf serum (FCS) and 11 mM glucose for 3 days in 5% CO 2 at 37°C before experiments. Medium was changed two times during preculture. Islets were then transferred to sterile Petri dishes in KRBH (Krebs-Ringer bicarbonate with HEPES) medium with 2 mg/mL bovine serum albumin (BSA) and 5.6 mM glucose.
  • KRBH Kerata-1
  • the islets were then washed twice in KRBH and studied for morphology and insulin secretion, or cultured for 2 or 24 hours in RPMI with 10% FCS and 11 mM glucose prior to morphological and biochemical examinations. Morphology and Islet Recovery
  • Triplicates of five islets were transferred to 200 ⁇ l of KRBH with 2 mg/mL BSA and 16.7 mM glucose and incubated for 60 minutes in 5% CO 2 at 37°C. Islets from each condition were then pooled and sonicated in 200 ⁇ l of redestilled water. A 50 ⁇ l aliquot of the homogenate was mixed with 125 ⁇ l acid ethanol (0.18 M HCI in 95% ethanol) and insulin extracted over- night. Insulin concentration in the sonicate and the culture medium was determined with ra- dioimmunoassay.
  • Groups of 10 islets were transferred to glass vials with 100 ⁇ l KRBH supplemented with D- [U 14 C]glucose and nonradioactive glucose to a final concentration of 16.7 mM glucose. Triplicate samples were used. The vials were suspended in scintillation flasks, gassed with 5% CO 2 and sealed airtight. The flasks were then shaken for 90 minutes at 37°C. Metabolism was stopped by injection of 100 ⁇ l of 0.05 mM antimycin A into the center vial. Immediately thereafter 250 ⁇ l hyamine hydroxide was injected into the outer flask.
  • CO 2 was released from the incubation medium by injecting 100 ⁇ l of 0.4 M Na 2 HPO 4 solution (pH 6.0) into the center vial. To allow the CO 2 to be trapped by the hyamine hydroxide the vials were incubated for another 120 minutes at 37°C. Scintillation fluid was then added to each flask and the radio- activity counted in a liquid scintillation counter.
  • the islets exposed to Streptozotocin for 30 minutes showed degranulation, and in some islets numerous pyknotic nuclei, at the 0 hour timepoint. No signs of recovery but a further destruction and also disintegration of islets was found at 2 and 24 hours.
  • islets incubated with test compounds + STZ appeared morphologically intact at the 0 hour timepoint. During the subsequent 24 hour culture a toxic effect of STZ became noticeable. At 2 hours the surface of these islets were somewhat irregular and this was more apparent at 24 hours.
  • the numerous pyknotic nuclei as seen in the STZ group were not found in the group of islets treated with test compounds.
  • Islets examined at the 0 hour timepoint ie after a 60 minutes incubation in 5.6 mM glucose, showed a stronger stain for insulin than the islets examined after 2 and 24 hours.
  • the latter islets had been cultured in 11 mM glucose.
  • the difference in insulin staining reflects a higher stimulation of insulin secretion at 11 mM compared to 5.6 mM glucose.
  • the insulin staining of the islets treated with test compounds + streptozotocin were stronger at both 2 and 24 hours than that seen with the islets incubated with medium alone.
  • the STZ treatment also had lowered the insulin and total protein biosynthesis as well as impaired the glucose oxidation rate.
  • the inhibitory effect of the K ATP channel openers on insulin secretion was seen in islets treated with test compounds + streptozotocin at 0 and 2 hours, but not after 24 hours.
  • a partial protection of the islet function was observed when compared with islets incubated with STZ alone.
  • the proinsulin and total protein biosynthesis in the recovered STZ islets were reduced to 35% and 51 % of control, respectively.
  • the proinsulin and total protein biosynthesis did not differ from the biosynthesis found in the recovered STZ.
  • PCO compounds The effect of PCO compounds on cell viability was analysed in 51 Cr-release cytotoxicity assays using either primary islet preparations (e.g. from newborn rats) or islet tumour cell lines (e.g. mouse transgenic ⁇ -cell lines ⁇ TC-3 or Min6, or rat insulinoma lines RIN5AH or MSLG2).
  • the assay has been used to measure toxic effects of e.g. cytokines or glucose, and to address the protective effect of PCO compounds on ⁇ -cell viability, e.g. during cytokine exposure.
  • the islets were then resuspended in 10 ml Islet media and 100 ⁇ l of the islet suspension were added to each well in a flat bottom 96 well plate (approximately 35 islets in each well).
  • Mixture of cytokines and test compounds or dimethyl sulphoxide were prepared in 100 ⁇ l media in each well. All test compounds were dissolved in dimethyl sulphoxide and prepared in stock solutions at a concentration of 100mM.
  • Viability assay using rodent adherent ⁇ -cell lines e.g. RIN cells, MIN6 cells, lns-1 cells and others.
  • HBSS life tech without Ca ++ and Mg ++ Cat 14185-045
  • 1 x trypsin in HBSS was used to split the cells.
  • the cells were seeded in a flat-bottomed 96 well plate in the desired media at a density of 40000 cells/well in 100 ⁇ l media and incubated overnight to secure proper adherence.
  • 2,5 ⁇ Ci/ml Na 51 Cr (Dupont, Nez 030S) was added to the labeling media (the desired media).
  • 1 x washing of the cells with HBSS 200 ⁇ l of media with Na 51 Cr were added to each well and incubated overnight.
  • the rodent adherent ⁇ -cell lines were incubated for 24h at 37 °C and 5% CO 2 .
  • the plates were centrifuged for 5 min at 1000 rpm, and 100 ⁇ l supernatant samples were harvested from each well.
  • 100 ⁇ l 1 % triton-X were added to each well in order to lyse the cells and 100 ⁇ l were harvested to get a maximum Na 51 Cr release from the cells of each well. All the samples and the maximum samples were counted on a cobra ⁇ -counter (Packard).
  • the effects on mitochondnal Katp channels kan be evaluated as described by e.g. Grimms- mann and Rustenbeck (Br. J. Pharmacol. 1998, 123, 781-788). Routinely the effects of the compounds of the present invention can be determined measuring changes in fluorescence of the dyes JC-1 or Rhodamine 123 when incubating beta cells or pancreatic islets in a medium containing the fluorencence indicators and the test compounds.

Abstract

The present invention relates to the use of potassium channel agonists for the treatment of insulitis associated with various forms of diabetes such as IDDM, NIDDM, SPIDDM (LADA) and gestational diabetes.

Description

USE OF POTASSIUM CHANNEL OPENERS FOR THE TREATMENT OF INSULITIS
FIELD OF THE INVENTION
The present invention relates to the use of potassium channel openers, which are able to protect the beta cells against toxic damage, for treating or preventing diseases related to autoimmune destruction of human beta cells, such as different types of diabetes, and methods of using these compounds.
BACKGROUND OF THE INVENTION Streptozotocin and alloxan are beta cell toxins. The toxic effect of these compounds on rat pancreatic islets in vitro and in vivo mimics the beta-cell death associated with Type 1 and late state Type 2 diabetes.
It has now been found that the compounds of the present invention are able to inhibit strep- tozotocin and alloxan induced beta cell degeneration and death.
The compounds of the present invention, known as potassium channel openers, act as activators of ATP regulated potassium channels (Katp-channels) of the beta cell and the Katp- channels of mitochondria. They may also act by antagonising the depletion of NAD induced in the islets by these toxins. Cytokines are known to reduce beta cell viability and to induce apoptosis. Cytokines have been proposed to be involved with the autoimmune degeneration of beta cells in Type 1 diabetes. The compounds of the present invention antagonize the effects of cytokines on beta cells.
Thus, the compounds of the present invention can be used in the treatment of insulitis associated with various forms of diabetes.
Various forms of diabetes are Type 1 or Insulin Dependent Diabetes Mellitus (IDDM), Type 2 diabetes or Non Insulin Dependent Diabetes Mellitus (NIDDM), slowly progressive IDDM (SPIDDM) also referred to as latent autoimmune diabetes in adults (LADA) and gestational diabetes due to underlying IDDM.
Examples of potassium channel openers are compounds disclosed in PCT Publication No. WO 97/26265 (see for instance from page 14, line 5 to page 19, line 9) and WO 99/03861 (see for instance from page 17, line 20 to page 19, line 5) as well as the following com- pounds: 3-tert-Butylamino-6-chloro-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide; 6-Chloro- 3-cyclobutylamino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 , 1 -dioxide; 6-Chloro-3-(1 , 1 -dimethyl- propylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 , 1 -dioxide; 6-Chloro-3-(1 -methylcyclo- propyl)amino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 , 1 -dioxide; 6-Chloro-3-(2-hydroxy-1 , 1 - dimethylethylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide and 6-Chloro-3-(1 , 1 ,3,3- tetramethylbutylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1 -dioxide.
DESCRIPTION OF THE INVENTION
The influence of ATP sensitive potassium (KAγp) channel openers, diazoxide and a analogue, 6-Chloro-3-isopropylamino-4H-thieno[3,2-e]-1,2,4-thiadiazine 1,1-dioxide, has been examined on experimental beta-cell damage induced by streptozotocin (STZ), alloxan or cytokines. Rat islets were preincubated for 30 minutes with the KATP channel openers and subsequently incubated for 30 minutes following the addition of STZ. The islets were then washed and cultured for 24 hours. The STZ treatment (0.5 mM) was associated with a 40% islet loss. The remaining islets showed reduced insulin content and secretion and a reduced insulin biosynthesis, amounting to 50%, 60% and 35%, respectively of control. The STZ islets also displayed a lowered rate of glucose oxidation - 16% of control. In contrast, islets pre-incubated with diazoxide or 6-Chloro-3-isopropylamino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide maintained higher insulin content and insulin secretion compared to islets incu- bated with STZ alone. In particular following incubation with 0.3 mM 6-Chloro-3-isopropyl- amino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide +STZ, there was no islet loss. In addition to having higher insulin content and secretion, these islets also had higher insulin biosynthesis and glucose oxidation rate than islets incubated with STZ alone. We also examined the influence of these KATP channel openers on damage induced by alloxan, a generator of reactive oxygen species. In these experiments, insulin release was reduced by 31 % after treatment with 0.5 mM alloxan. This reduction was fully counteracted by simultaneous incubations with 0.3 mM 6-Chloro-3-isopropylamino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1- dioxide or 0.3 mM diazoxide. Glucose oxidation rate in islets treated with 0.5 mM alloxan was decreased after 24 hours by 51%. Islets treated with alloxan in the presence of diazox- ide had a glucose oxidation rate of 73% of control. Islets incubated with 6-Chloro-3- isopropylamino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide did not differ from control. The results demonstrate that KATP channel openers can protect insulin-producing cells from being damaged by a beta-cell toxin and suggest that such an effect might be applicable in subjects with ongoing insulitis. Diazoxide and other KATP channel openers, such as cromakalim and pinacidil, have been employed in experimental studies of ischemic heart. A beneficial, cardioprotective effect was observed (Garlid KD et al., Circulation Res 1997; 81 :1072-82). Although the mechanism of this phenomenon is not understood, an opening of mitochondnal potassium channels seems to be involved, resulting in dissipation of the inner mitochondnal membrane potential. This in turn leads to net oxidation of the mitochondria with an apparent reduction of energy wastage.
Diazoxide is known to act on KATP channels in the plasma membrane of beta cells. It hyper- polarizes the membrane and reduces the entry of Ca2+, essential for the exocytosis of secre- tory granulaes. Recently, exposure of beta cells to diazoxide was found to engage also mitochondnal KATp channels (Grimmsmann T et al., Br J Pharmacol 1998; 123:781-788). In the present study, we examined the influence of potassium channel openers on experimental beta-cell damage induced by streptozotocin, an agent known to cause energy depletion, on damage induced by alloxan, a generator of reactive oxygen species and on damage induced by cytokines.
Islet Isolation, Culture and Experimental Design
Pancreata from Sprague-Dawley rats were collagenase digested and islets collected with a braking pipette as previously described (Sandier S et al., Endocrinology, 1987;121 :1424-31). Islets were precultured free floating in RPM1 1640 medium with 10 % (v/v) fetal calf serum (FCS) and 11 mM glucose for 3 days in 5% CO2at 37°C before experiments. Medium was changed two times during preculture. Islets were then transferred to sterile Petri dishes in KRBH (Krebs-Ringer bicarbonate with HEPES) medium with 2 mg/mL bovine serum albumin (BSA) and 5.6 mM glucose.
Stock solutions of test compounds dissolved in dimethyl sulphoxide were prepared and added to the Petri dishes. Islets were incubated in 5% CO2 at 37°C for 30 minutes with or without test compounds and STZ in 0.9% NaCI was then added to a final concentration of 0.5 mM. Dry powder of alloxan was diluted to a stock of 50 mM just before the addition to the Petri dishes to a final concentration of 0.5 mM. The incubation continued for another 30 minutes and was terminated by the addition of 1 mL of cold KRBH. The islets were then washed twice in KRBH and studied for morphology and insulin secretion, or cultured for 2 or 24 hours in RPMI with 10% FCS and 11 mM glucose prior to morphological and biochemical examinations. Morphology and Islet Recovery
About 100 islets per condition were carefully transferred to a glass tube and spun down at 800 rpm for 1 minute. The medium was removed and about 200 μl left before the fixation with 8 ml of Bouin's medium, followed by dehydration in ethanol. The pellets were embedded in paraffin, cut in 5 μm sections and stained for insulin (guinea-pig anti-insulin, 1 :100 dilution, DAKO, Sweden) using the PAP method. For estimation of islet recovery, 30 islets from each condition were transferred to Petri dishes as described above and the remaining islets counted after 24 hours.
Insulin Secretion and Islet Insulin Contents
Triplicates of five islets were transferred to 200 μl of KRBH with 2 mg/mL BSA and 16.7 mM glucose and incubated for 60 minutes in 5% CO2 at 37°C. Islets from each condition were then pooled and sonicated in 200 μl of redestilled water. A 50μl aliquot of the homogenate was mixed with 125 μl acid ethanol (0.18 M HCI in 95% ethanol) and insulin extracted over- night. Insulin concentration in the sonicate and the culture medium was determined with ra- dioimmunoassay.
Proinsulin Biosynthesis and Total Protein Biosynthesis
For each condition duplicate samples of 20 islets were transferred to multiwell plates con- taining 100 μl KRBH with L-[4.5-3H]leucine (50 μCi/ml), 2 mg/mL BSA and 16.7 mM glucose and incubated for 120 minutes in 5 % CO2 at 37°C. Islets were then washed in Hanks' solution supplemented with 10 mM nonradioactive leucine and subsequently sonicated in 200 μl of redestilled water. A 50 μl fraction of the aqueous homogenate was incubated for 90 minutes with insulin antibodies coupled to Sepharose beads to separate proinsulin from other labelled proteins (15). Total protein biosynthesis was obtained by precipitating the labelled proteins with trichloroacetic acid (TCA). The antibody bound and TCA precipitable radioactivity were determined in a liquid scintillation counter.
Glucose oxidation
Groups of 10 islets were transferred to glass vials with 100 μl KRBH supplemented with D- [U14C]glucose and nonradioactive glucose to a final concentration of 16.7 mM glucose. Triplicate samples were used. The vials were suspended in scintillation flasks, gassed with 5% CO2 and sealed airtight. The flasks were then shaken for 90 minutes at 37°C. Metabolism was stopped by injection of 100 μl of 0.05 mM antimycin A into the center vial. Immediately thereafter 250 μl hyamine hydroxide was injected into the outer flask. CO2 was released from the incubation medium by injecting 100 μl of 0.4 M Na2HPO4 solution (pH 6.0) into the center vial. To allow the CO2 to be trapped by the hyamine hydroxide the vials were incubated for another 120 minutes at 37°C. Scintillation fluid was then added to each flask and the radio- activity counted in a liquid scintillation counter.
Statistics
Students' paired t-test and analysis of variance (ANOVA) were used when appropriate.
Islet Recovery and Morphology
The islets exposed to Streptozotocin for 30 minutes showed degranulation, and in some islets numerous pyknotic nuclei, at the 0 hour timepoint. No signs of recovery but a further destruction and also disintegration of islets was found at 2 and 24 hours. In contrast, islets incubated with test compounds + STZ appeared morphologically intact at the 0 hour timepoint. During the subsequent 24 hour culture a toxic effect of STZ became noticeable. At 2 hours the surface of these islets were somewhat irregular and this was more apparent at 24 hours. The numerous pyknotic nuclei as seen in the STZ group were not found in the group of islets treated with test compounds.
Islets examined at the 0 hour timepoint, ie after a 60 minutes incubation in 5.6 mM glucose, showed a stronger stain for insulin than the islets examined after 2 and 24 hours. The latter islets had been cultured in 11 mM glucose. The difference in insulin staining reflects a higher stimulation of insulin secretion at 11 mM compared to 5.6 mM glucose. The insulin staining of the islets treated with test compounds + streptozotocin were stronger at both 2 and 24 hours than that seen with the islets incubated with medium alone.
Functional Characteristics
The islets recovered 24 hours after the STZ treatment had reduced insulin content and glu- cose-stimulated insulin release. The STZ treatment also had lowered the insulin and total protein biosynthesis as well as impaired the glucose oxidation rate. An inhibition of insulin secretion was found with islets incubated with test compounds alone at 0 and 2 hours but not at 24 hours. The inhibitory effect of the KATP channel openers on insulin secretion was seen in islets treated with test compounds + streptozotocin at 0 and 2 hours, but not after 24 hours. At 24 hours following test compounds + STZ treatments, a partial protection of the islet function was observed when compared with islets incubated with STZ alone.
At 24 hours, the proinsulin and total protein biosynthesis in the recovered STZ islets were reduced to 35% and 51 % of control, respectively. The lowering of the proinsulin/total protein biosynthesis ratio, 15% compared to 23% in control islets, indicates a preferential beta-cell effect of the STZ treatment. In islets treated with test compounds + streptozotocin the proinsulin and total protein biosynthesis did not differ from the biosynthesis found in the recovered STZ.
Cytokine induced beta cell toxicity
The effect of PCO compounds on cell viability was analysed in 51Cr-release cytotoxicity assays using either primary islet preparations (e.g. from newborn rats) or islet tumour cell lines (e.g. mouse transgenic β-cell lines βTC-3 or Min6, or rat insulinoma lines RIN5AH or MSLG2). The assay has been used to measure toxic effects of e.g. cytokines or glucose, and to address the protective effect of PCO compounds on β-cell viability, e.g. during cytokine exposure.
METHODS Viability assay using primary islets:
Approximately 3500 islets were washed and resuspended in 15 ml islet media (RPMI1640 (Life tech cat 61870-010) + 10% FCS (Life cat 16000-044)) + 100 lU/ml Penicillin 100 UG/ ml streptomycin). 2,5 μCi/ml Na51Cr (Dupont, Nez 030S) was added and the suspension was transferred to a 60 mm petri dish and incubated overnight at 37 °C and 5% CO2. After incubation the islets were washed 3 times in 1 x HBSS (life tech without Ca++ and Mg++ Cat 14185-045). The islets were then resuspended in 10 ml Islet media and 100 μl of the islet suspension were added to each well in a flat bottom 96 well plate (approximately 35 islets in each well). Mixture of cytokines and test compounds or dimethyl sulphoxide were prepared in 100μl media in each well. All test compounds were dissolved in dimethyl sulphoxide and prepared in stock solutions at a concentration of 100mM. Stock solutions of 10ng/μl of cytokines (Pharmingen mrlL-1 β; 19201V, mrTNF-α; 19321T; mrlFN-γ, 19301T) dissolved in distilled H2O were prepared, and added to the wells in final concentrations ranging from 0,01 ng/ml to 100ng/ml. The islets were incubated for 48h at 37 °C and 5% CO2. The plates were centrifuged for 5 min at 1000 rpm, and 100 μl supernatant samples were harvested from each well. 100 μl 1% triton-X were added to each well in order to lyse the islets and 100 μl were harvested to obtain the total releasable Na51Cr from the islets of each well. All the samples and the maxi- mum samples were counted on a Cobra γ-counter (Packard). The release of Na51Cr was calculated for each sample, by normalizing to its own maximum and calculated by the following equation: ((Sample in %-spontaneous in %)/(100-spontaneous))%. All samples were made in quadruplicates.
Normalised sample= (Sample cpm / (sample maximum * 2)) *100%
Spontaneous release= (Untreated cells cpm / (sample maximum *2)) *100%
Viability assay using rodent adherent β-cell lines (e.g. RIN cells, MIN6 cells, lns-1 cells and others)
Cells were grown to approximately 80 % confluence. After washing once in HBSS (life tech without Ca++ and Mg++ Cat 14185-045), 1 x trypsin in HBSS was used to split the cells. The cells were seeded in a flat-bottomed 96 well plate in the desired media at a density of 40000 cells/well in 100 μl media and incubated overnight to secure proper adherence. 2,5 μCi/ml Na51Cr (Dupont, Nez 030S) was added to the labeling media (the desired media). After 1 x washing of the cells with HBSS 200 μl of media with Na51Cr were added to each well and incubated overnight. After Na51 Cr incubation cells were washed twice in HBSS, before addition of media with cytokines and PCO compounds or dimethyl sulphoxide. Mixture of these media was prepared in stocks with 200μl for each well. All PCO-compounds were dissolved in dimethyl sulphoxid and prepared in stock solutions at a concentration of 100mM. Stock solutions of 10ng/μl of cytokines (Pharmingen mrlL-1β; 19201V, mrTNF-α; 19321T; mrlFN-γ, 19301T) dissolved in distilled H2O were prepared, and added to the stocks in final concentrations ranging from 0,1 ng/ml to 100ng/ml.
The rodent adherent β-cell lines were incubated for 24h at 37 °C and 5% CO2. The plates were centrifuged for 5 min at 1000 rpm, and 100 μl supernatant samples were harvested from each well. 100 μl 1 % triton-X were added to each well in order to lyse the cells and 100 μl were harvested to get a maximum Na51Cr release from the cells of each well. All the samples and the maximum samples were counted on a cobra γ-counter (Packard). The release of Na51Cr was calculated for each sample, by normalizing to its own maximum and calcu- lated by the following equation: ((Sample in %-spontaneous in %)/(100-spontaneous))%. All samples were made in quadruplicates.
Normalised sample= (Sample cpm / (sample maximum * 2)) *100% Spontaneous release= (Untreated cells cpm / (sample maximum *2)) *100%
Effects on mitochondria.
The effects on mitochondnal Katp channels kan be evaluated as described by e.g. Grimms- mann and Rustenbeck (Br. J. Pharmacol. 1998, 123, 781-788). Routinely the effects of the compounds of the present invention can be determined measuring changes in fluorescence of the dyes JC-1 or Rhodamine 123 when incubating beta cells or pancreatic islets in a medium containing the fluorencence indicators and the test compounds.

Claims

1. The use of a potassium channel opener protecting the beta cells against toxic damage for the preparation of a pharmaceutical composition for treating or preventing diseases related to autoimmune destruction of human beta cells.
2. The use according to claim 1 wherein the protection of the beta cells is established through an opening of mitochondrial potassium channels.
3. The use according to anyone of the preceding claims wherein the diseases are related to different types of diabetes selected from the group consisting of IDDM, NIDDM, SPIDDM or LADA and gestational IDDM.
4. The use according to anyone of the preceding claims wherein the potassium chan- nel opener is selected from:
6-Chloro-3-isopropylamino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1 -dioxide, 3-tert-Butylamino-6-chloro-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1 -dioxide, 6-Chloro-3-(1 ,1-dimethylpropylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide, 6-Chloro-3-(1-methylcyclopropyl)amino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide, 6-Chloro-3-(2-hydroxy-1 , 1 -dimethylethylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1,1- dioxide,
6-Chloro-3-(1 ,1 ,3,3-tetramethylbutylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide, or other potassium channel openers as disclosed in the description.
5. The use of a potassium channel opener antagonising streptozotocin induced depletion of NAD in the pancreatic islets for the preparation of a pharmaceutical composition for treating or preventing diseases related to autoimmune destruction of human beta cells.
6. The use according to claim 5 wherein the depletion of NAD in the pancreatic islets is obtained through inhibition of poly(ADP-ribose)synthetase.
7. The use according to claim 5 or 6 wherein the diseases are related to different types of diabetes selected from the group consisting of IDDM, NIDDM, SPIDDM or LADA and gestational IDDM.
8. The use according to anyone of the preceding claims 5-7 wherein the potassium channel openers is selected from:
6-Chloro-3-isopropylamino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1 -dioxide, 3-tert-Butylamino-6-chloro-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1 -dioxide, 6-Chloro-3-(1 , 1 -dimethylpropylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1 -dioxide, 6-Chloro-3-(1-methylcyclopropyl)amino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1,1-dioxide, 6-Chloro-3-(2-hydroxy-1 , 1 -dimethylethylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1- dioxide,
6-Chloro-3-(1 , 1 ,3,3-tetramethylbutylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 , 1 -dioxide, or other potassium channel openers as disclosed in the description.
9. A method of treating or preventing diseases related to autoimmune destruction of human beta cells comprising administering an effective amount of a potassium channel op- erner protecting the beta cells against toxic damage.
10. A method according to claim 9 wherein the protection of the beta cells is established through an opening of mitochondrial potassium channels.
11. A method according to claim 9 or 10 wherein the diseases are related to different types of diabetes selected from the group consisting of: IDDM, NIDDM, SPIDDM or LADA and gestational IDDM.
12. A method according to anyone of the preceding claims 9-11 wherein the potassium channel opener is selected from:: 6-Chloro-3-isopropylamino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide, 3-tert-Butylamino-6-chloro-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1 -dioxide, 6-Chloro-3-(1 ,1-dimethylpropylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide, 6-Chloro-3-(1-methylcyclopropyl)amino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1 -dioxide, 6-Chloro-3-(2-hydroxy-1 ,1-dimethylethylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1- dioxide,
6-Chloro-3-(1 ,1 ,3,3-tetramethylbutylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1 -dioxide, or other potassium channel openers as disclosed in the description.
13. A method of treating or preventing diseases related to autoimmune destruction of human beta cells comprising administering an effective amount of a potassium channel op- erner antagonising streptozotocin induced depletion of NAD in the pancreatic islets.
14. A method according to claim 13 wherein the depletion of NAD in the pancreatic islets is obtained through inhibition of poly(ADP-ribose)synthetase.
15. A method according to claim 13 or 14 wherein the diseases are related to different types of diabetes selected from the group consisting of: IDDM, NIDDM, SPIDDM or LADA and gestational IDDM.
16. A method according to anyone of the preceding claims 13-15 wherein the potassium channel opener is selected from: 6-Chloro-3-isopropylamino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1 -dioxide, 3-tert-Butylamino-6-chloro-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide,
6-Chloro-3-(1 ,1-dimethylpropylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1-dioxide, 6-Chloro-3-(1-methylcyclopropyl)amino-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1 -dioxide, 6-Chloro-3-(2-hydroxy-1 ,1-dimethylethylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1- dioxide, 6-Chloro-3-(1 ,1 ,3,3-tetramethylbutylamino)-4H-thieno[3,2-e]-1 ,2,4-thiadiazine 1 ,1 -dioxide, or other potassium channel openers as disclosed in the description.
PCT/DK2001/000444 2000-06-26 2001-06-25 Use of potassium channel openers for the treatment of insulitis WO2002000665A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2001265841A AU2001265841A1 (en) 2000-06-26 2001-06-25 Use of potassium channel openers for the treatment of insulitis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DKPA200000988 2000-06-26
DKPA200000988 2000-06-26

Publications (1)

Publication Number Publication Date
WO2002000665A1 true WO2002000665A1 (en) 2002-01-03

Family

ID=8159578

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2001/000444 WO2002000665A1 (en) 2000-06-26 2001-06-25 Use of potassium channel openers for the treatment of insulitis

Country Status (2)

Country Link
AU (1) AU2001265841A1 (en)
WO (1) WO2002000665A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003045954A1 (en) * 2001-11-30 2003-06-05 Novo Nordisk A/S Use of selective potassium channel openers
WO2003045955A1 (en) * 2001-11-30 2003-06-05 Novo Nordisk A/S Use of selective potassium channel openers
WO2004005299A1 (en) * 2002-07-04 2004-01-15 Novo Nordisk A/S Polymorphic forms of a 4h-thieno[3,2-e]-1,2,4-thiadiazine 1,1-dioxide derivative
WO2020130836A1 (en) * 2018-12-21 2020-06-25 ACADEMISCH ZIEKENHUIS LEIDEN (h.o.d.n. LUMC) Compound for use in treatment and prevention of type i diabetes
NL2022291B1 (en) * 2018-12-21 2020-07-21 Academisch Ziekenhuis Leiden Compound for Use in Treatment and Prevention of Type I Diabetes

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1368948A (en) * 1970-11-11 1974-10-02 Manuf Prod Pharma Pyridine derivatives
EP0618209A1 (en) * 1993-03-26 1994-10-05 Adir Et Compagnie Pyridothiadiazines, processes for their preparation and pharmaceutical compositions containing them
US5889002A (en) * 1996-01-17 1999-03-30 Novo Nordisk A/S Fused 1,2,4-thiadiazine and fused 1,4-thiazine derivatives, their preparation and use
US5972894A (en) * 1997-08-07 1999-10-26 Cytran, Inc. Peptides having potassium channel opener activity
WO2000037474A1 (en) * 1998-12-18 2000-06-29 Novo Nordisk A/S Fused 1,2,4-thiadiazine derivatives, their preparation and use
US6225310B1 (en) * 1996-01-17 2001-05-01 Novo Nordisk A/S Fused 1,2,4-thiadiazine derivatives, their preparation and use

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1368948A (en) * 1970-11-11 1974-10-02 Manuf Prod Pharma Pyridine derivatives
EP0618209A1 (en) * 1993-03-26 1994-10-05 Adir Et Compagnie Pyridothiadiazines, processes for their preparation and pharmaceutical compositions containing them
US5889002A (en) * 1996-01-17 1999-03-30 Novo Nordisk A/S Fused 1,2,4-thiadiazine and fused 1,4-thiazine derivatives, their preparation and use
US6225310B1 (en) * 1996-01-17 2001-05-01 Novo Nordisk A/S Fused 1,2,4-thiadiazine derivatives, their preparation and use
US5972894A (en) * 1997-08-07 1999-10-26 Cytran, Inc. Peptides having potassium channel opener activity
WO2000037474A1 (en) * 1998-12-18 2000-06-29 Novo Nordisk A/S Fused 1,2,4-thiadiazine derivatives, their preparation and use

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
STEFAN GLOCKER ET AL: "Binding and effects of P1075, an opener of ATP-sensitive K+ channels, in the aorta from streptozotocin-treated diabetic rats", NAUNYN-SCHMIEDEBERG'S ARCH PHARMACOL, vol. 356, 1997, pages 210 - 215, XP002901885 *
T C HOHMAN ET AL: "ATP-sensitive K+ channel effects on nerve function, Na+, K+ ATPase, and glutathione in diabetic rats", EUROPEAN JOURNAL OF PHARMACOLOGY, vol. 397, 2000, pages 335 - 341, XP002901886 *
W D VLAHOS ET AL: "Diabetes prevention in BB rats by inhibition of endogenous insulin secretion", METABOLISM, vol. 40, no. 8, August 1991 (1991-08-01), pages 825 - 829, XP002901887 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003045954A1 (en) * 2001-11-30 2003-06-05 Novo Nordisk A/S Use of selective potassium channel openers
WO2003045955A1 (en) * 2001-11-30 2003-06-05 Novo Nordisk A/S Use of selective potassium channel openers
WO2004005299A1 (en) * 2002-07-04 2004-01-15 Novo Nordisk A/S Polymorphic forms of a 4h-thieno[3,2-e]-1,2,4-thiadiazine 1,1-dioxide derivative
WO2020130836A1 (en) * 2018-12-21 2020-06-25 ACADEMISCH ZIEKENHUIS LEIDEN (h.o.d.n. LUMC) Compound for use in treatment and prevention of type i diabetes
NL2022291B1 (en) * 2018-12-21 2020-07-21 Academisch Ziekenhuis Leiden Compound for Use in Treatment and Prevention of Type I Diabetes

Also Published As

Publication number Publication date
AU2001265841A1 (en) 2002-01-08

Similar Documents

Publication Publication Date Title
Miguez et al. Hepatoprotective mechanism of silymarin: no evidence for involvement of cytochrome P450 2E1
Devalia et al. Dissociation of cell death from covalent binding of paracetamol by flavones in a hepatocyte system
Katz et al. Kinetics and autoradiography of high affinity uptake of serotonin by primary astrocyte cultures
Jancso Inflammation and the inflammatory mechanisms
US6033656A (en) Method of preventing or alleviating mammalian obesity
Salhanick et al. Dexamethasone stimulates insulin receptor synthesis in cultured rat hepatocytes.
Nosek et al. Calcium stimulates ATP-Mg/Pi carrier activity in rat liver mitochondria.
BOQUIST et al. Uptake of labeled alloxan in mouse organs and mitochondria in vivo and in vitro
Rodriguez et al. Comparison of ketoconazole-and fluconazole-induced hepatotoxicity in a primary culture system of rat hepatocytes
Sundquist et al. The α1-adrenergic receptor in human erythrocyte membranes mediates interaction in vitro of epinephrine and thyroid hormone at the membrane Ca2+-ATPase
Ohkuma et al. The effects of basic substances and acidic ionophores on the digestion of exogenous and endogenous proteins in mouse peritoneal macrophages.
Lemons et al. Mediated calcium transport by isolated human fibroblast lysosomes
Kullin et al. K (ATP) channel openers protect rat islets against the toxic effect of streptozotocin.
WO2002000665A1 (en) Use of potassium channel openers for the treatment of insulitis
Brostrom et al. Alterations of glial tumor cell Ca2+ metabolism and Ca2+-dependent cAMP accumulation by phorbol myristate acetate.
US20020032193A1 (en) Use of potassium channel openers for the treatment of insulitis
De Barry et al. Molecular mechanism of the cardiotoxic action of a polypeptide neurotoxin from sea anemone on cultured embryonic cardiac cells
Urani et al. Benomyl affects the microtubule cytoskeleton and the glutathione level of mammalian primary cultured hepatocytes
Galvan et al. Calcium regulation by lens plasma membrane vesicles
Alexander et al. The influence of selenium on methyl mercury toxicity in rat hepatoma cells, human embryonic fibroblasts and human lymphocytes in culture
Kullin et al. Protection of rat pancreatic islets by potassium channel openers against alloxan, sodium nitroprusside and interleukin-1β mediated suppression—possible involvement of the mitochondrial membrane potential
De Clerck et al. Effect of flunarizine on the human red cell shape changes and calcium deposition induced by A 23187
Marchetti et al. Characterization of peripheral benzodiazepine receptors in purified large mammal pancreatic islets
Oldham et al. Primary cultures of adult rat hepatocytes—a model for the toxicity of histamine H2-receptor antagonists
Corin et al. Transport of the folate compound methotrexate decreases during differentiation of murine erythroleukemia cells.

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP