OA19279A - Treatment for Parkinson's disease. - Google Patents

Treatment for Parkinson's disease. Download PDF

Info

Publication number
OA19279A
OA19279A OA1201800475 OA19279A OA 19279 A OA19279 A OA 19279A OA 1201800475 OA1201800475 OA 1201800475 OA 19279 A OA19279 A OA 19279A
Authority
OA
OAPI
Prior art keywords
compound
formula
alkyl
hydrogen
halogen
Prior art date
Application number
OA1201800475
Inventor
Nitin Krishnaji Damle
Sanjay Nandlalji Mandhane
Manoj Atmaramji Upadhya
Sameer Vishwanath Mehetre
Gajanan Uttamrao Chidrewar
Prabal Sengupta
Trinadha Rao Chitturi
Original Assignee
Sun Pharma Advanced Research Company Limited
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 Sun Pharma Advanced Research Company Limited filed Critical Sun Pharma Advanced Research Company Limited
Publication of OA19279A publication Critical patent/OA19279A/en

Links

Abstract

The invention relates to a method of treating or preventing Parkinson's disease in a subject comprising administering a compound of Formula I wherein, R1 is –NHC(O) C3-6 cycloalkyl and R2 is hydrogen; or R1 and R2 along with the carbon atoms to which they are attached form a six membered aromatic ring, wherein the ring is substituted with one or more groups selected from hydrogen, halogen and C1-6 alkyl; R3 and R4 are independently selected from group comprising hydrogen, halogen, C1-3 alkyl, OC1- 3 alkyl, NO2, SC1-3 alkyl, C1-3 haloalkyl, OC1-3 haloalkyl, and SC1-3 haloalkyl; or a pharmaceutically acceptable salt thereof.

Description

RELATED APPLICATIONS
This application daims the benefit of Indian Patent Application no. IN 201621019087 filed on June 02, 2016 and IN 201621019185 filed on June 02, 2016; which is hereby incorporated by reference . ,
FIELD OF THE INVENTION
The invention relates to a method of treating or preventing Parkinson’s disease in a subject comprising administering a compound of Formula I
Formula I wherein, Ri is -NHC(O) C3.6 cycloalkyl and R2 is hydrogen;
or Rt and R2 along with the carbon atoms to which they are attached form a six membered aromatic ring, wherein the ring is substituted with one or more groups selected from hydrogen, halogen and Ci.6 alkyl;
R3 and R4 are independently selected from group comprising hydrogen, halogen, Ον3 alkyl, OC^ alkyl, NO2, SC^ alkyl, ΟΊ.3 haloalkyl, OC^ haloalkyl, and SC^ haloalkyl; or a pharmaceutically acceptable sait thereof.
BACKGROUND OF THE INVENTION c-Abl is a non-receptor protein tyrosine kinase that is implicated in various cellular processes which include régulation of cell survival, growth and motility. c-Abl kinase inhibitors such as imatinib (Gleevec®), nilotinib (Tasigna®), dasatinib (Sprycel®) and ponatinib (Iclusig® ) hâve been developed and marketed for clinicai use in the treatment of chronic myeloid ieukemia.
Recent studies hâve demonstrated that c-Abl plays an important rôle in oxidative stressinduced neuronal cell death (Wu et al., Cell Death Differ., 2016; 23:542-552). It has been reported that c-Abl is involved in Parkinson’s disease (Gonfloni et al., Int. J. Cell Biol., 2012; 2012:1-7). Also, c-Abl is known to be activated by dopaminergic stress and by dopaminergic neurotoxins viz. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP, converted enzymatically in vivo to its active form MPP+) causes tyrosine phosphorylation of parkin leading to loss of parkin’s E3 ligase activity. This results in the accumulation of various substrates of parkin and ultimately to death of dopaminergic neurons (Ko et al., PNAS, 2010; 107:16691-16696).
Parkinson disease (PD) is a common neurodegenerative disease characterized by protein accumulation in intracellular inclusions designated as Lewy bodies and Lewy neuritis and subséquent loss of dopaminergic neurons. Rare familial mutations hâve provided insight into this chronic, progressive neurodegenerative disease like mutation in α-synuclein and LRRK2 cause autosomal-dominant PD, whereas mutations in DJ-1, PINK1 and parkin results in autosomal-recessive PD. Parkin is an E3 ubiquitin ligase, and familial mutations are thought to impair E3 ligase activity of parkin (Ko et al., PNAS, 2010; 107:16691-16696).
c-Abl has been shown to regulate the dégradation of two proteins implicated in the pathogenesis of PD viz. parkin and α-synuclein (Mahul-Mellier et al., Hum. Mol. Genet., 2014; 23:2858-2879). c-Abl phosphorylâtes parkin on tyrosine 143. This phosphorylation inhibits parkin’s E3 ubiquitin ligase activity, leading to accumulation of AIMP2 and FBP1 (parkin substrates) and loss of parkin’s cytoprotective function resulting in cell death (Ko et al., PNAS, 2010; 107:16691-16696; Imam étal., J. Neurosci., 2011; 31:157-163). Further, cAbl régulâtes clearance of α-synuclein, a synaptic protein that has been strongly implicated in the pathogenesis of PD. A bi-directional relationship in vivo between α-synuclein and c-Abl has been described wherein an increase in α-synuclein expression facilitâtes its phosphorylation and subséquent activation of c-Abl. Conversely, an increase in the c-Abl expression and activation results in α-synuclein accumulation and aggregation, suggesting that inhibition of c-Abl might constitute a viable strategy for protecting dopaminergic neurons from accumulated α-synuclein toxicity in PD (Hébron et al., Hum. Mol. Genet., 2013; 22:3315-3328; Hébron et al., Autophagy, 2013; 9:1249-1250).
c-Abl inhibitors like nilotinib are known to cross the blood-brain barrier and protect dopaminergic neurons in a mouse model of PD induced with 1-methyl-4-phenyl-1,2,3,6tetrahydropyridine (herein referred to as MPTP) (Karuppagounder et al., Sci. Rep. 2014; 4:4874). Nilotinib has been shown to increase α-synuclein clearance via the autophagy pathway and protects against α-synuclein accumulation-induced loss of dopaminergic neurons in this mouse model of PD (Hébron et al., Hum. Mol. Genet., 2013; 22:3315-3328; Imam et al., J. Neurosci., 2011; 31:157-163). Further, US20150087653 discloses method of treating neurodegenerative diseases comprising of administering tyrosine kinase inhibitors such as nilotinib. However, nilotinib has several major drug associated adverse effects. USFDA has issued a boxed warning for Tasigna® capsules since its treatment is associated with potentially severe cardiac side effects (QT prolongation) and sudden deaths in patients. Dasatinib is known to cause pleural effusion and hemorrhage. Ponatinib is also associated with severe adverse effects which include thromboembolism and vascular occlusion. Imatinib is not a potent inhibitor of Abl kinase. Moreover, both imatinib and dasatinib being Pglycoprotein (p-gp) substrate, show poor brain concentration. . Thus, there is a need for a potent Abl kinase inhibitor which can cross the blood-brain barrier and does not cause cardiovascular side effects.
SUMMARY OF THE INVENTION
The invention provides method of treating or preventing Parkinson’s disease in a subject comprising administering a therapeutically effective amount of a compound of Formula I,
Formula I wherein, Rt is -NHC(O) C3.6 cycloalkyl and R2 is hydrogen;
or Rt and R2 along with the carbon atoms to which they are attached form a six membered aromatic ring, wherein the ring is substituted with one or more groups selected from hydrogen, halogen and C^e alkyl;
R3 and R4 are independently selected from group comprising hydrogen, halogen, C^ alkyl, OC^ alkyl, NO2, SC^s alkyl, 0^3 haloalkyl, 0Cv3 haloalkyl, and SC-|.3 haloalkyl; or its pharmaceutically acceptable sait thereof.
DESCRIPTION OF THE FIGURES
Fig.1. Photomicrographs from coronal sections showing tyrosine hydroxylase (TH)-positive neurons, showing prévention of neurodegeneration by the compound of Formula l.a.
Fig.2. Percentage area of TH-positive neurons on administration of the compound of Formula l.a.
Fig.3. Integrated density of TH-immunoreactivity on administration of the compound of Formula l.a.
Fig.4. Photomicrographs from coronal sections showing tyrosine hydroxylase (TH)-positive neurons, showing prévention of neurodegeneration by the compound of Formula l.b.
Fig.5. Percentage area of TH-positive neurons on administration of the compound of Formula l.b.
Fig.6. Integrated density of TH-immunoreactivity on administration of the compound of Formula l.b.
DETAILED DESCRIPTION OF THE INVENTION
In one aspect, the présent invention provides a method of treating or preventing Parkinson’s disease in a subject comprising administering a therapeutically effective amount of the compound of Formula I,
Formula I wherein, Rt is -NHC(O) C3.6 cycloalkyl and R2 is hydrogen;
or Rt and R2 along with the carbon atoms to which they are attached form a six membered aromatic ring, wherein the ring is substituted with one or more groups selected from hydrogen, halogen and Ον6 alkyl;
R3 and R4 are independently selected from group comprising hydrogen, halogen, Ci.3 alkyl, OCi-3 alkyl, NO2, SC^ alkyl, C1.3 haloalkyl, 00^3 haloalkyl, and SCi.3 haloalkyl; or its pharmaceutically acceptable sait thereof.
In another aspect, the présent invention provides a method of treating or preventing Parkinson’s disease in a subject, comprising selecting a subject suffering from Parkinson’s disease or at risk of developing Parkinson’s disease and administering a therapeutically effective amount of a compound of Formula I.
The phrase “therapeutically effective amount of compound of Formula I” as used herein refers to amount of the compound of Formula I that elicit the therapeutic effect for which it is administered.
The term alkyP refers to a saturated hydrocarbon chain radical that includes solely carbon and hydrogen atoms in the backbone, either linear or branched and which is attached to the rest of the molécule by a single bond, e.g., methyl, ethyl, n-propyl, 1-methylethyl (isopropyl), n-butyl and n-pentyl.
The numerical in phrases like “Cve alkyl”, refers that there are 1 to 6 carbon atoms in the alky chain.
The term “C3.6 cycloalkyf refers to a non-aromatic mono-cyclic ring System of 3 to 6 carbon atoms. Monocyclic rings include cylcopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
The term “haloalkyf refers to alkyl chain substituted with one or more halogen radical selected from chloride, bromide, iodide and fluoride.
In one embodiment, the présent invention provides method of treating or preventing Parkinson’s disease comprising administering a therapeutically effective amount of a compound of Formula I wherein FL in the compound of Formula I is -NHC(O) cyclopropyl and R2 is hydrogen.
In another embodiment, the présent invention provides a method of treating or preventing 5 Parkmson’s disease comprising administering a therapeutically effective amount of a compound of Formula I wherein, Rd and R2 in the compound of Formula I, along with the carbon atoms to which they are attached form a six membered aromatic ring, wherein the ring is substituted with one or more groups selected from hydrogen, halogen and Ον6 alkyl. Preferably, the aromatic ring is substituted with hydrogen i.e. unsubstituted.
In another embodiment, the présent invention provides a method of treating or preventing Parkmson’s disease comprising administering a therapeutically effective amount of a compound of Formula I wherein, Rt and R2 in the compound of Formula I, along with the carbon atoms to which they are attached form a six membered aromatic ring, wherein the 15 ring is substituted with hydrogen i.e. unsubstituted; and R3 is chloro and R4 is methyl and are présent as a substituent at 2 and 6 position in the ring.
In another embodiment, the présent invention provides a method of treating or preventing Parkmson’s disease comprising administering a therapeutically effective amount of a 20 compound of Formula I wherein, R3 and R4 in the compound of Formula I are selected from halogen and alkyl. In a preferred embodiment, R3 and R4 are halogen and methyl and are présent as a substituent at 2 and 6 position in the ring.
The Chemical name of some of the preferred compounds of Formula I are provided below in 25 Table 1.
Table 1:
Compound No. Chemical name
La A/-(2-Chloro-6-methylbenzoyl)-4-methyl-3-[2-(3-quinolyl)ethynyl]benzo hydrazide
l.b Cyclopropanecarboxylic acid (5-{5-[N'-(2-chloro-6- methylbenzoyl)hydrazine carbonyl]-2-methyl-phenylethynyl}-pyridin-2-
yl)amide
l.c Cyclohexanecarboxylic acid (5-{5-[N'-(2-chloro-6- methylbenzoyl)hydrazine carbonyl]-2-methyl-phenylethynyl}-pyridin-2yl)amide
l.d Cyclobutanecarboxyhc acid (5-{5-[N'-(2-chloro-6-methylbenzoyl) hydrazine carbonyl]-2-methyl-phenylethynyl}-pyridin-2-yl)amide
Le A/-(2-Chloro-6-methylbenzoyl)-4-methyl-3-[2-(6-chloro-3quinolyl)ethynyl]-benzohydrazide
l.f /V-(2-Chloro-6-methylbenzoyl)-4-methyl-3-[2-(6-methyl-3-quinolyl) ethynyl]-benzohydrazide
ig A/-(2-Chloro-6-methylbenzoyl)-4-methyl-3-[2-(6-fluoro-3quinolyl)ethynyl]-benzohydrazide
Suitable pharmaceutically acceptable salts of the compound of the invention may be salts of inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, and the like or of organic acids such as, for example, acetic acid, benzenesulfonic acid, methanesulfonic 5 acid, benzoic acid, citric acid, glycolic acid, lactic acid, fumaric acid, succinic acid, adipic acid, pimelic acid, suberic acid, azelaic acid, malic acid, tartartic acid, or amino acids, such as glutamic acid or aspartic acid, and the like. One or more hydrogen atoms of the compound of Formula I may be deuteriated i.e. substituted with a deuterium atom.
WIPO publication WO2012098416 (the ‘416 publication) discloses a markush group of compounds active as c-Abl kinase inhibitors and their usefulness for the treatment of cancers like chronic myelogenous leukemia (CML). Compounds of Formula I of the présent invention may be prepared by the processes described in WO2012098416 and WO2016185490 which are incorporated herein by reference.
The inventors hâve found that the compound of Formula I of présent invention, are potent Abl kinase inhibitors and advantageously cross the blood-brain barrier effectively resulting in a high ratio of brain to plasma concentration for the compound of Formula I and a high therapeutic index.
Particularly, the inventors hâve found that the compound of Formula I, at therapeutically effective dose, is devoid of cardiovascular side effects when tested for its in vitro effect on hERG channel and its in vivo effect on ECG parameters like QT interval, QTC interval, QTcf interval and heart rate in conscious beagle dogs and guinea pig. The compound of Formula I was found to be safe as they did not show any undue effect on ECG parameters and heart rate as described herein in examples.
The compound of Formula I can be administered orally in the form of a suitable dosage form. A suitable dosage form may include tablet, pellets, capsule, sachet, pellets in sachet, pellets in capsule, powder, granules and the like. The compound of Formula I may be formulated in oral dosage form which may include pharmaceutically acceptable excipients which are in common knowledge of a person skilled in the art. Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses pharmaceutically acceptable carriers which can be used for préparation of a suitable dosage form.
The following examples serve to illustrate the invention without limiting the invention in its scope.
EXAMPLE 1
Abl Kinase inhibition
In a final reaction volume of 25 pL, Abl (human) (5-10 mU) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 50 μΜ EAIYAAPFAKKK, 10 mM Mg(OAc)2 and [γ-33Ρ-ΑΤΡ] [spécifie activity approx. 500 cpm/pmol, concentration as required). The reaction is initiated by the addition of the MgATP mix. After incubation for 40 minutes at room température, the reaction is stopped by the addition of 5 pL of a 3 % phosphoric acid solution. 10 pL of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in methanol priorto drying and scintillation counting.
Results for the représentative compounds of Formula I are provided in Table-2.
Table 2: Comparative potencies of tyrosine kinase inhibitors in c-Abl
IC50 (nM)
La l.b Nilotinib Ponatinib Dasatinib Imatinib
0.9 0.6 18 0.4 0.27 190
EXAMPLE 2
Brain/plasma pharmacokinetic studies
To détermine whetherthe compound of Formula La crosses the blood-brain barrier, C57BL6 5 mice were administered orally at 30 mg/kg of the compound of Formula La, 100 mg/kg of nilotinib or 30 mg/kg of dasatinib. At 1, 4 & 8 hour time points post treatment, mice were anesthetized with isoflurane and a 0.4 mL of blood was withdrawn from retro-orbital plexus into eppendorf tubes containing 8 pL sodium heparin as an anticoagulant (100 lU/ml) and transferred to ice containers. Blood samples were centrifuged immediately for 7 min at 8500 10 rpm, 4 °C. Plasma was separated in the pre-labeled eppendorf tubes and stored at -70 °C till further analysis. Immediately thereafter, the mice were sacrificed, whole brain was removed and rinsed with ice-cold phosphate buffered saline (PBS) to remove extraneous blood and blot-dried. Brain tissue samples were weighed and homogenized in 1:2 volume of PBS using a tissue homogenizer and stored in labeled vials at -70 °C until further analysis.
Concentrations of the compound of Formula La in brain and plasma were determined using the LC-MS technique.
Brain to plasma ratio was found to be significantly higher for the compound of Formula La as compared to that for nilotinib or dasatinib (see Table 3).
Table 3: Concentration of compounds in Brain and Plasma.
Compoun ds Treatment * (mg/kg) Time point s (hr) Plasma conc. (ng of compound/m L of plasma) Brain conc. (ng of compound/g of brain tissue) Ratio of brain conc./plas ma conc.
Compoun d of Formula La 30 1 4798 ± 858 1901 ±959 0.40
4 3167 ±50 510 ±367 0.16
8 2715 + 379 435 ±157 0.16
Nilotinib 100 1 31683 ± 7958 380 ± 70 0.01
4 38813 ±11635 487±126 0.01
8 16988 ±2133 180 ±46 0.01
Dasatinib 30 1 553 ± 550 23.4 ± 0 0.04
4 222 ±122 25 ±5 0.11
8 BQL 24 ±4 -
bUL-beluw the Ιιιηιί ot quantification
EXAMPLE 3
Efficacy in animal model of Parkinson’s disease for compound of Formula l.a
C57BL/6 mice (6-8 weeks old, 25-30 g body weight) were administered orally with vehicle, the compounds of Formula l.a (10 or 30 mg/kg, once a day) for 7 days. On day 7, these animais received four intraperitoneal injections of a neurotoxin, 1-methyl-4-phenyl-1,2,3,6tetrahydropyridine (MPTP-HCI; 17 mg/kg free base; Sigma) in saline at 2 hour intervals. Daily dosing with compound continued for 7 additional days after the last injection of MPTP. Briefly, the compound was administered 6 days prior to MPTP administration, on the day of MPTP administration and 7 days after MPTP administration. Ail animais were sacrificed 7 days after the MPTP administration and brain tissues were processed for immunohistochemistry évaluation. The standard avidin-biotin method (Benno et al., Brain Res., 1982, 246.225-236) was used for the immunohistochemical détection of tyrosine hydroxylase (TH). Initially, the brains were sectioned at the substantia nigra pars compacta (SNPc) level using a cryostat and mounted on glass slides. The slides with brain sections were rinsed 3x5 minutes in 0.1 M PBS. The sections were first incubated for 30 minutes at room température with normal blocking sérum in 0.1 M PBS with 0.3 % Triton X-100 and for additional 2 hours with a rabbit polyclonal antibody to mouse tyrosine hydroxylase (Invitrogen) diluted 1:1000 in 0.1 M PBS with normal blocking sérum. The slides with sections were further rinsed 3x5 minutes in 0.1 M PBS and incubated for 1 hour at room température with a biotinylated anti-rabbit antibody (Vectastain® ABC kit). The sections were rinsed 3x5 minutes in 0.1 M PBS and incubated for 1 hour at room température with A and B solutions (Vectastain® ABC kit) diluted 1:50 in 0.1 M PBS. After 1 hour the sections were rinsed 3x5 minutes in 0.1 M PBS and incubated in the 3,3’-diaminobenzidine (DAB)/H2O2 solution (Sigma) for approximately 5-8 minutes (the development process was checked under microscope for an optimal signal-to-noise ratio). The sections were first rinsed 2x5 minutes in 0.1 M PBS and then with 3x5 minutes in Milli-Q water. The slides were cover shpped in glycerol-gelatin solution and kept at room température overnight for drying after which images of the stained brain sections were captured using a caméra attached to a microscope and anaslyzed using NIH ImageJ® software (NIH, Bethesda, MD). Images were analyzed by converting to 8-bit resolution and optimal brightness/contrast. Background and 5 pseudo signais on individual images were removed and area covered by cell bodies and fibers was measured. Percentage area compared to total image area and integrated density was calculated. Results obtained were compared for any inter-group or intra-group variation and statistical significance using GraphPad Prism® 6.0. Oral administration of the compound of Formula La significantly prevented the neurodegeneration induced by MPTP as seen from photomicrographs from coronal sections showing TH-positive neurons (Fig. 1), the détermination of percentage area of TH-positive neurons (Fig. 2) and integrated density of TH-immunoreactivity (Fig. 3)
EXAMPLE 4
Efficacy in animal model of Parkinson’s disease for compound of Formula l.b
Efficacy of compound of Formula l.b for treating the Parkinson’s disease was tested by the same method as described for the compound of Formula La in Example 3 above.
The results obtained were compared for any inter-group or intra-group variation and 20 statistical significance using GraphPad Prism® 6.0. Oral administration of the compound of
Formula Lb significantly prevented the neurodegeneration induced by MPTP as seen from photomicrographs from coronal sections showing TH-positive neurons (Fig. 4), the détermination of percentage area of TH-positive neurons (Fig. 5) and integrated density of TH-immunoreactivity (Fig. 6)
EXAMPLE 5
Electrophysiological Procedures- Effect on hERG K* Channel
The compound of Formula La and Lb were tested for cardiovascular safety in an in vitro test to détermine inhibition of hERG K+ channel. The compounds of Formula La and Lb did not 30 show any significant inhibition of hERG current at the concentration tested.
The in vitro effect of the compounds of Formula La and Lb on ionic currents in voltageclamped human embryonic kidney cells (HEK293) that stably express the human ether-à-go11 go-related gene (hERG) was examined. Two concentrations of the compounds of Formula La and Lb (1 and 10 μΜ) were tested at near physiological température.
Cells were transferred to the recording chamber and superfused with vehicle control solution 5 (HEPES-buffered physiological saline solution+1 % dimethylsulfoxide + 1 % Bovine sérum albumin). Micropipette solution for whole cell patch clamp recordings was composed of : potassium aspartate, 130 mM; MgCI2, 5 mM; EGTA, 5 mM; ATP, 4 mM; HEPES, 10 mM; pH adjusted to 7.2 with 10N KOH. Micropipette solution was prepared in batches, aliquoted, stored frozen and a fresh aliquot thawed each day. The recording was performed at a 10 température of 33 to 35 °C using a combination of in4ine solution pre-heater, chamber heater and feedback température controller. Température was measured using a thermistor probe in the recording chamber. Micropipettes for patch clamp recording were made from glass capillary tubing using a P-97 micropipette puller (Sutter Instruments, Novato, CA). A commercial patch clamp amplifier was used for whole cell recordings. Before digitization, 15 current records were low-pass filtered at one-fifth of the sampling frequency.
Cells stably expressing hERG were held at -80 mV. Onset and steady State inhibition of hERG potassium current due to the test compounds (Formula La and Lb) were measured using a puise pattern with fixed amplitudes (conditioning prepulse +20 mV for 1 s; 20 repolanzing test ramp to -80 mV (-0.5 V/s) repeated at 5 s intervals). Each recording ended with a final application of a supra-maximal concentration of the reference substance (E-4031, 500 nM) to assess the contribution of endogenous currents. The remaining uninhibited current was subtracted off-line digitally from the data to détermine the potency of the test compounds for hERG inhibition.
The compound of Formula La inhibited hERG current by (Mean ± SEM) 2.1 ± 0.4 % at 1 μΜ (n = 3) and 9.7 ± 0.4 % at 10 μΜ (n = 3) compared to 1.0 ± 0.6 % (n = 3) in vehicle control (see Table 4). Concentrations of the compound of Formula La greater than 10 μΜ were not tested due to the solubility limit of the compound in the vehicle control. Under similar 30 conditions, the positive control (terfenadine, 60 nM) inhibited hERG potassium current by (Mean ± SD; n = 2) 82.8 ± 1.2 %, confirming a slight effect of the compound of Formula La at its very high concentration in hERG K+ channeL In comparison nilotinib shows nearly 90 % inhibition at 1 μΜ concentration (Table 5).
Table 4: Mean percent inhibition of hERG current at each concentration of the compound of Formula La and l.b.
Compound Concentratio η (μΜ) Mean SD SEM N
Compound l.a 0 1.0 % 1.0 % 0.6 % 3
1 2.1 % 0.7 % 0.4 % 3
10 9.7 % 0.7 % 0.4 % 3
Compound l.b 0 1.6 % 0.3 % 0.2 % 3
0.3 5.6 % 1.3% 0.8 % 3
1 12.9 %* 4.1 % 2.4 % 3
3 25.0 %* 6.0 % 3.4 % 3
value is statistically different from vehicle alone.
# I
Table 5: percentage inhibition of hERG current by nilotinib#
Conc. (pM) 0.03 0.1 0.3 1
% Inhibition 14.9 ±2 43.9 ±0.7 70.4 ± 2.5 89.7 ±1.6
. 22-068 (Tasigna® Capsule) pg No.
review
EXAMPLE 6
Effect of compound of Formula l.a on QT, QTC intervals, and heart rate in conscious beagle dogs
The compound of Formula l.a was subjected to in vivo test to détermine its effect on the QT, QTcb & QTCf intervals, and heart rate in conscious beagle dogs.
Compound of Formula l.a was administered via pérorai (p.o.) route at three dose levels: 5, 15, and 30 mg/kg in conscious telemetered male and female beagle dogs. Emesis occurred in two animais (1 male and 1 female) treated with the 30 mg/kg dose group of the compound of Formula l.a at 14 min following dosing. Hence, the group treated with 30 mg/kg dose was not considered for the data analysis. No emesis was observed with 5 and 15 mg/kg doses.
Measurements of ECG parameters (QT interval, QTcb interval, QTcf interval) and heart rate were carried out and recorded over a period of 2 hour prior to drug administration (baseline), and continuously up to 24 hour post-administration. Each animal received both vehicle (placebo) and three different doses of the compound of Formula l.a on different days in a 5 Latin square design, with a washout period of a minimum of 96 hr. On a separate day, one additional oral dosing of compound of Formula l.a was performed to assess plasma concentrations of the compound of Formula l.a at different time points, to dérivé pharmacokinetic (PK) parameters.
Data of ECG parameters (QT interval, QTcb interval, QTcf interval,) and heart rate were statistically compared as follows: Data of the placebo group at 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12 and 24 hour time points were compared with baseline data of the same group. Data of different groups treated with the compound of Formula l.a were compared with the corresponding data of the placebo group.
Analysis of ECG based on pooled data of male and female dogs showed that as compared to baseline, placebo treatment had no statistically significant effect on any of the ECG parameters (QT interval, QTcb interval, QTcf interval) and heart rate. Compound of Formula La at 5 and 15 mg/kg doses had no effects on QT, QTcb, QTcf, durations compared with 20 placebo. In addition, other parameter such as heart rate remained unchanged.
The PK analysis showed a dose-dependent systemic exposure of the compound of Formula l.a to animais administered with different doses of the compound of Formula l.a. AUC0.inf was in the range of 1925 to 4824, 6776 to 12756, and 25927 to 46749 hrxng/mL with 5, 15, and 25 30 mg/kg doses of the compound of Formula l.a, respectively. Cmax was in the range of 1218 to 2033, 2723 to 5323, and 9825 to 9967 ng/mL with 5, 15, and 30 mg/kg doses of the compound of Formula l.a, respectively.
The results of this study showed that single oral administration of the compound of Formula La up to 15 mg/kg dose has no effect on cardiovascular functions in conscious beagle dogs.
There was no change in ECG morphology at any dose levels of the compound of Formula l.a. There were no gender différences in the treatment effect on ECG parameters and heart rate.
EXAMPLE 7
Effect of compound of Formula l.b on QT, QTc intervals, and heart rate in anesthetized guinea pig
The effect of compound of Formula l.b on QT, QTc intervals, and heart rate was studied in anesthetized guinea pig.
Male Dunkm-Hartley guinea pigs of body weight range 300-400 g were anaesthetized by intraperitoneal injection of urethane (1.5 g/kg). A tracheotomy was performed and animal was allowed to breath spontaneously throughout the experiment. The left jugular vein was catheterized using polyethylene cathéter filled with heparinized saline solution (100 IU/ mL) for drug administration. Electrodes were placed in lead II position and baseline ECG was recorded.
For different animais, according to body weight, doses of the compound of Formula l.b were weighed and prepared in 0.5 mL of DMSO and administered via a slow intravenous infusion administered over a period of 10 min. ECGs were recorded during and after completion of infusion up to 0.5 hr. QT and RR intervals were measured, using PowerLab 4SP® software. The data were analyzed at 1, 5, and 10 min during infusion and 5, 10, 15 and 30 min after infusion taking mean of 9 beats.
There was no change in QT intervals with administration of 1 mg/kg intravenous dose of the comund °f Formula | b

Claims (8)

1. Use of a compound of Formula R1 ^N.
R3 R4
Formula I wherein, FL is -NHC(O) C3.6 cycloalkyl and R2 is hydrogen;
or and R2 along with the carbon atoms to which they are attached form a six membered aromatic ring, wherein the ring is substituted with one or more groups selected from hydrogen, halogen and C^e alkyl;
R3 and R4 are independently selected from group comprising hydrogen, halogen, Cv3 alkyl, OC^ alkyl, NO2, SC^ alkyl, haloalkyl, OC^ haloalkyl, and SC^s haloalkyl, in the manufacture of a médicament for use in a method of treating or preventing Parkinson’s disease.
2. The use as claimed in claim 1, wherein R3 and R4 in the compound of Formula I are selected from halogen and alkyl.
3. The use as claimed in either of daims 1 and 2, wherein Rt in the compound of Formula I is -NHC(O) cyclopropyl and R2 is hydrogen.
4. The use as claimed in any one of the preceding daims, wherein Rt and R2 in the compound of Formula I, along with the carbon atoms to which they are attached form a six membered aromatic ring, wherein the ring is substituted with one or more groups selected from hydrogen, halogen and C^e alkyl.
5. A compound of Formula I
R3 R4
Formula I wherein, R-, is -NHC(O) C3.6 cycloalkyl and R2 is hydrogen;
or Rt and R2 along with the carbon atoms to which they are attached form a six membered aromatic ring, wherein the ring is substituted with one or more groups selected from hydrogen, halogen and alkyl;
R3 and R4 are independently selected from group comprising hydrogen, halogen, alkyl, OC^ alkyl, NO2, SC^ alkyl, haloalkyl, OC^a haloalkyl, and SCi.3 haloalkyl, for use in a method of treating or preventing Parkinson’s disease.
6. The compound as claimed in claim 5, wherein R3 and R4 in the compound of Formula I are selected from halogen and alkyl.
7 The compound as claimed in either of daims 5 and 6, wherein Rt in the compound of Formula I is -NHC(O) cyclopropyl and R2 is hydrogen.
8. The compound as claimed in any one of daims 5 to 7, wherein Rt and R2 in the compound of Formula I, along with the carbon atoms to which they are attached form a six membered aromatic ring, wherein the ring is substituted with one or more groups selected from hydrogen, halogen and alkyl.
OA1201800475 2016-06-02 2017-06-02 Treatment for Parkinson's disease. OA19279A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN201621019087 2016-06-02
IN201621019185 2016-06-02

Publications (1)

Publication Number Publication Date
OA19279A true OA19279A (en) 2020-06-05

Family

ID=

Similar Documents

Publication Publication Date Title
US11813252B2 (en) Treatment for Parkinson's disease
JP6509386B2 (en) Compounds for treating mTOR pathway related diseases
NO332772B1 (en) Use of receptor tyrosine kinase inhibitor for the preparation of a unit dose drug for the treatment of cancer
WO2020259528A1 (en) Method for treating idiopathic pulmonary fibrosis
JP2009521470A (en) Treatment of synuclein disorder
JP2004529916A (en) Adenosine A2A receptor antagonist in combination with a neurotrophic active compound in the treatment of Parkinson's disease
US20200397777A1 (en) Prevention and treatment of organ fibrosis
JP2020525446A (en) Modulators of cystic fibrosis transmembrane conductance regulator for treating autosomal dominant polycystic kidney disease
JP2010531854A (en) Pirenzepine and its derivatives as anti-amyloid agents
OA19279A (en) Treatment for Parkinson's disease.
JP2010531854A5 (en)
CZ289533B6 (en) Use of dihydropyridine derivatives with annellated carbon or heterocyclic rings for the preparation of pharmaceutical preparations intended for protection of brain tissue
EA037697B1 (en) Treatment for parkinson's disease
US20030166673A1 (en) Medicines for the prevention and treatment of neurodegenerative diseases
US20100331363A1 (en) Treatment of mitochondrial disorders using a farnesyl transferase inhibitor
US20210308138A1 (en) Tissue transglutaminase modulators for medicinal use
JP2022530890A (en) How to treat a cough by using a diaminopyrimidine compound
WO2018203559A1 (en) Pharmaceutical compositions for polyglutamine diseases
ITMI20110208A1 (en) HETEROCYCLES WITH ANTI-HYPERTENSIVE ACTIVITY
JP2000505425A (en) Methods of treating conditions associated with beta-amyloid peptide using TRH