Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SOLID PHARMACEUTICAL FORMULATIONS COMPRISING TICAGRELOR
Document Type and Number:
WIPO Patent Application WO/2021/080529
Kind Code:
A1
Abstract:
The present invention relates to a pharmaceutical composition, comprising ticagrelor, wherein the pharmaceutical composition is in a solid dosage form with improved pharmaceutical characteristics. Solid dosage form comprising ticagrelor and pharmaceutically acceptable excipients manufactured by using wet granulation method wherein; the composition comprises microcrystalline cellulose, and silicon dioxide together in the extragranular phase in weight ratio of microcrystalline cellulose : silicon dioxide is between 35:1 to 45:1.

Inventors:
YILDIRIM ERSIN (TR)
KANIK BAYRAM (TR)
ÖZTÜRK FATMA (TR)
AKTAŞ TANSEL (TR)
Application Number:
PCT/TR2019/050903
Publication Date:
April 29, 2021
Filing Date:
October 26, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SANTA FARMA ILAC SANAYI A S (TR)
International Classes:
A61K31/519; A61K9/20
Domestic Patent References:
WO2019170244A12019-09-12
Foreign References:
EP3332769A12018-06-13
Other References:
LI, J. Z. ET AL.: "The role of intra-and extragranular microcrystalline cellulose in tablet dissolution", PHARMACEUTICAL DEVELOPMENT AND TECHNOLOGY, vol. 1, no. 4, 1996, pages 343 - 355, XP009124869, DOI: 10.3109/10837459609031429
See also references of EP 4048276A4
Attorney, Agent or Firm:
BULUT, Pınar (TR)
Download PDF:
Claims:
CLAIMS

1. A pharmaceutical composition comprising ticagrelor and pharmaceutically acceptable excipients manufactured by using wet granulation method wherein; the composition comprises - Microcrystalline cellulose, and

- Silicon dioxide together in the extragranular phase in weight ratio of Microcrystalline cellulose : Silicon dioxide is between 35:1 to 45:1.

2. A pharmaceutical composition according to claim 1, wherein the composition is in the form of solid dosage form.

3. A pharmaceutical composition according to any one of the preceding claims, comprises crystalline ticagrelor in the form of polymorph II.

4. A pharmaceutical composition according to claim 3; comprises crystalline ticagrelor having at least X-ray powder diffraction peaks 5.5°, 13.5°, 18.3°, 22.7° and 24.3° (±0.1° 2Q). 5. A pharmaceutical composition according to any one of the preceding claims, wherein the pharmaceutically acceptable excipient is selected from fillers, diluents, disintegrants, binders and lubricants.

6. A pharmaceutical composition according to claim 5, wherein suitable filler is selected from phosphate dihydrate, polysaccharides, primarily microcrystalline cellulose, lactose, mannitol, sugars, sorbitol, sucrose, inorganic salts, primarily calcium salts and the like and mixtures thereof, preferably filler is microcrystalline cellulose.

7. A pharmaceutical composition according to claim 5, wherein a diluent is selected from silicon dioxide, magnesium oxide, glyceryl monostearate, glyceryl monooleate, glyceryl behanate, mannitol and mixtures thereof, preferably the diluent is a mixture of mannitol and silicon dioxide.

8. A pharmaceutical composition according to claim 5, wherein a disintegrant is selected from croscarmellose sodium, crospovidone, low-substituted hydroxypropyl cellulose, starch, sodium starch glycolate, carmellose and mixtures thereof, preferably the disintegrant is sodium starch glycolate.

9. A pharmaceutical composition according to claim 5, wherein binder is selected from hypromellose, cellulose or cellulose derivatives, povidone, starch, sucrose, polyethylene glycol, or mixtures thereof.

10. A pharmaceutical composition according to claim 9, wherein the binder is hydroxypropyl cellulose.

11. A pharmaceutical composition according to claim 5, wherein a lubricant is selected from sodium stearyl fumarate, magnesium stearate, calcium stearate talc, stearic acid and mixtures thereof., preferably, the lubricant is sodium stearyl fumarate, magnesium stearate or a mixture thereof, more preferably the lubricant is magnesium stearate.

12. A pharmaceutical composition according to any one of the preceding claims wherein the quantitative composition in w/w % is as stated below:

Ticagrelor 20 - 30

Mannitol 30 - 40

Microcrystalline cellulose 30 - 40

Hydroxypropyl cellulose 0.1 - 5

Sodium starch glycolate 0.1 - 5

Silicon dioxide 0.1 - 2

Magnesium stearate 0.1 - 2

Deionised water Quantity sufficient Coating agent 1 - 5 wherein together in the extragranular phase in weight ratio of Microcrystalline cellulose : Silicon dioxide is between 35:1 to 45:1.

13. A wet granulation process for preparing a pharmaceutical composition comprises crystalline Ticagrelor as polymorph II, wherein the process comprises the following steps: a. Ticagrelor, mannitol, specified amount of microcrystalline cellulose, specified amount of hydroxypropyl cellulose and sodium starch glycolate are stirred in high-shear wet granulator for 5 minutes, b. Granulation solution was prepared by dissolving specified amount of hydroxypropyl cellulose in deionized water, c. Sufficient quantity of granulation solution was added on the powder blend in step a and granulation process was performed with a 10 minute period stirring, d. Granule obtained in step c is dried at a specified temperature, e. Dried granule was sifted and remaining amounts of microcrystalline cellulose and hydroxypropyl cellulose and also silicon dioxide were added and stirred, f. Obtained granule in step e was lubricated with magnesium stearate for 4 minutes, g. Final blend in step f is subjected to tablet compression process, h. Tablets obtained in step g were coated with a proper film coating agent.

Description:
SOLID PHARMACEUTICAL FORMULATIONS COMPRISING TICAGRELOR

FIELD OF INVENTION

The present invention relates to a pharmaceutical composition, comprising ticagrelor, wherein the pharmaceutical composition is in a solid dosage form with improved pharmaceutical characteristics.

STATE OF ART

Ticagrelor is a platelet aggregation inhibitor that acts by inhibition of adenosine 5- diphosphate. Ticagrelor is a reversible, selective P2Y 12-receptor antagonist. Its chemical name is (lS,2S,3R,5S)-3-(7-{[(lR,2S)-2-(3,4-Diflourophenyl)cycloprop yl]amino}-5- (propylsulfanyl)-3H-[l,2,3]triazol[4,5-d]pyrimidine-3-yl)-5- (2-hydroxyethoxy)cyclopentane- 1,2-diol and its molecular structure is given below. Its CAS Number is 274693-27-5.

Formuia (!)

Ticagrelor is marketed by AstraZeneca under trade names BRILINTA and BRILIQUE with 60 mg and 90 mg in worldwide. Ticagrelor is indicated for the treatment or prevention of thrombotic events e.g. stroke, heart attack, acute coronary syndrome or myocardial infarction with ST elevation, other coronary artery diseases and arterial thrombosis plus other disorders related to platelet aggregation (WO 00/34283). WO99/05143 discloses a series of triazolo [4,5-d]pyrimidine compounds, including Ticagrelor. WOOl/92262 and WO2013/079589 disclose various crystalline and amorphous forms of Ticagrelor. In the pharmaceutical dosage forms of the present invention any of these forms can be used. Pharmaceutical compositions comprising ticagrelor are disclosed in WO 2008/024044 and WO 2008/024045. Oral pharmaceutical compositions which can comprise up to 50% by weight of the active ingredient are explained and they release all the active ingredient substantially. A pharmaceutical composition comprises active ingredient, a filler consisting essentially of a mixture of mannitol and dibasic calcium phosphate dihydrate, a binder consisting essentially of hydroxypropyl cellulose, a disintegrant consisting essentially of sodium starch glycolate, and one or more lubricants. Wet granulation process is preferred as the manufacturing method.

W02015001489 relates to oral pharmaceutical compositions comprising amorphous ticagrelor and processes for their preparation. TR2017/15203 discloses a method of manufacture for a formulation comprising ticagrelor, characterized in that the step of granulating the solid mixture wherein the mixture comprises the active agent ticagrelor and at least one water-soluble binder and at least one water-soluble filler that is granulated with a suitable granulation solution.

TR2016/01835 discloses a process for producing a formulation comprising ticagrelor or a pharmaceutically acceptable salt thereof, characterized in that the ticagrelor or pharmaceutically acceptable salt is dissolved and / or dispersed in a solvent or solvent mixture during manufacture.

EP3332769 relates to a solid oral pharmaceutical composition, comprising ticagrelor or a pharmaceutically acceptable salt thereof with at least one hydrophilic polymer wherein the composition is substantially free of calcium salts and their derivatives.

WO20 14059955 relates to a solid oral pharmaceutical formulation containing ticagrelor as the active pharmaceutical constituent wherein the pharmaceutical composition contains at least one the non-hygroscopic binder, at least one the non-hygroscopic filler and at least one lubricant and substantially no disintegrant. WO20 18229785 relates to oral pharmaceutical compositions comprising ticagrelor and pharmaceutically acceptable polymer in a ratio of 1:0.1 to 1:1 in the form of solid dispersion.

WO2015110952 relates to a composition comprising ticagrelor or salt thereof in an amount less than 20 % of the weight of total composition, wherein the composition is devoid of water- insoluble fillers

CN107397717 discloses a solid preparation of ticagrelor or a pharmaceutically acceptable salt thereof which comprises filler, disintegrant, lubricant, additive and high polymer. The solid preparation is prepared by mixing a filler and ticagrelor or its pharmacy or it is acceptable to salt, adding a lubricant and a disintegrant respectively and mixing the additive and the polymer to form a suitable solid preparation.

CN109700773 relates to a pharmaceutical composition comprising ticagrelor wherein the percentage the ticagrelor of weight at the formulation composition is 2%.

CN201310432438 discloses a solid dispersion comprising ticagrelor in which ticagrelor is dispersed in a carrier material to solve the problem of solubility. CN106265557 discloses a dispersible tablet formulation comprising ticagrelor in which ticagrelor is in an amount of the total tablet weight 20 to 50% by weight.

Ticagrelor is a cyclo-pentyltriazolo-pyrimidine that reversibly inhibits the P2Y12receptor which is a protein found mainly but not exclusively on the surface of blood platelets and which is an important regulator in blood clotting. Ticagrelor (BR LINTA®) is indicated to reduce the rate of cardiovascular death, myocardial infarction, and stroke in patients with acute coronary syndrome (ACS) or a history of myocardial infarction (MI). For at least the first 12 months following ACS, it is superior to clopidogrel. It also reduces the rate of stent thrombosis in patients who have been stented for treatment of ACS. In the management of ACS, ticagrelor treatment is performed with a 180 mg loading dose. During the first year after an ACS event, 180 mg dose is administered daily. After one year 120 mg dose is administered.

In the state of the art, the patent application no. EP2056832 discloses an oral pharmaceutical composition comprising ticagrelor and other excipients. The composition is presented as enhancing the bioavailability through increasing the release of the active agent due to the formulation comprising at least two fillers and one single binder. Moreover, one of these fillers is dibasic calcium phosphate dehydrate which is commonly known by its abrasive properties. Considering these undesirable properties, the use of dibasic calcium phosphate dehydrate as a tablet excipient shall increase the required amount of lubricants (Handbook of Pharmaceutical Excipients, sixth edition, page 97). However, the ratio of the lubricant given in the application, which is preferably magnesium stearate, is in the range of 0.5-1% by weight. This low ratio is likely to cause stability problems in the presence of dibasic calcium phosphate dehydrate. On the other hand, the increase of the magnesium stearate amount shall reduce the dissolution rate and the promised enhanced bioavailability accordingly (Handbook of Pharmaceutical Excipients, sixth edition, page 405).

Another patent application no. WO2011076749 discloses solid dosage forms comprising ticagrelor, characterized in that at least 90% by volume of ticagrelor particles have a particle size in the range of 1 pm to 150 pm. It further discloses that ticagrelor, being poorly soluble, presents a significant problem in the design of pharmaceutical compositions. Further, in order to exhibit good bioavailability, it is desirable to formulate dosage forms showing fast dissolution of the drug. However, it is stated that these effects are observable only if the ratio of ticagrelor is more that 50% by weight in the composition with the particle size of the drug and by the use of hydrophilic polymers/emulsifiers in the dosage form. Ticagrelor is classified as a BCS class IV compound, exhibiting low solubility and low permeability. This property leads to an undesirable dissolution profile and hence poor bioavailability. It also leads to high intra-subject and inter-subject variability following oral administration.

Ticagrelor has a low aqueous solubility (10 pg/mL in water) and this situation brings bioavailability and stability problems regarding decreases in the solubility of the tablet formulations. In the state of the art, several methods are followed, different forms of ticagrelor and different excipients are used to compose innovative formulations and to overcome some of these problems. However, the challenge is to formulate a composition, some qualities of which won't reduce, while the others are improved. Thus, there is a need in the art for a pharmaceutical composition comprising ticagrelor providing enhanced dissolution profile, stability and bioavailability at the same time. SUMMARY OF INVENTION

The object of this invention is to develop an immediate -release solid pharmaceutical formulation comprising ticagrelor or a pharmaceutically acceptable salt thereof, which has improved dissolution release and blend properties prior to tablet compression. The present invention relates to an immediate release solid oral composition comprising only ticagrelor as active substance and one or more acceptable excipients manufactured by using optimised wet granulation process.

Another object of the present invention is related to an oral dosage form composition in concurrent use of acetylsalicylic acid for the treatment of the prevention of atherothrombotic events in adult patients with

- acute coronary syndromes (ACS) or

- a history of myocardial infarction (MI) and a high risk of developing an atherothrombotic event.

Another object of the present invention is to provide a preparation method of a pharmaceutical composition of ticagrelor wherein the pharmaceutical compositions herein disclosed can be manufactured into solid dosage forms, such as tablet, film-coated tablet, capsule, sachet, having good blend characteristics and the desired dissolution profiles.

In a preferred embodiment present invention, the active agent is crystalline ticagrelor. Preferably, the active agent is ticagrelor polymorph II presenting X-ray powder diffraction pattern containing specific peaks at 5.5° (±0.1°), 6.8° (±0.1°), 10.6° (±0.1°), 13.5° (±0.1°), 14.9° (±0.1°), 18.3° (±0.1°), 19.2° (±0.1°), 22.7° (±0.1°), 24.3° (±0.1°) and 27.G (±0.1°) 2Q.

In a preferred embodiment present invention, the active agent is ticagrelor polymorph II which is also used in innovator drug product and declared that it is the most stable crystalline form of ticagrelor hat has desired solubility characteristics. Another object of the present invention is to provide a pharmaceutical compositions containing ticagrelor by using wet granulation process wherein provided for the manufacture of solid dosage form containing the active ingredient, fillers, diluents, disintegrants, binders and lubricants etc. In another object of the present invention is provide a pharmaceutical compositions containing ticagrelor wherein the total weight of the ticagrelor is less than 30% w/w by total solid dosage form weight.

In another object of the present invention is provide a pharmaceutical composition containing ticagrelor with optimized amounts of excipients manufactured by using wet granulation method.

DETAILED DESCRIPTION OF THE INVENTION

The present invention provides a solid oral dosage form comprising ticagrelor and optimized amounts of pharmaceutically acceptable excipients manufactured by using the most proper manufacturing method.

The present invention relates to an immediate release solid oral composition comprising ticagrelor and one or more acceptable excipients manufactured by using optimised wet granulation process.

Another object of the present invention is to provide a preparation method of a pharmaceutical composition of ticagrelor wherein the pharmaceutical compositions herein disclosed can be manufactured into solid dosage forms, such as tablet, film coated tablet, sachet, capsule having improved granule characteristics and the desired dissolution profiles.

Another object of the present invention is to provide a pharmaceutical compositions containing ticagrelor by using wet granulation process wherein provided for the manufacture of tablets containg the active ingredient, fillers, diluents, disintegrants, solubilizers, and lubricants etc.

In another object of the present invention is provide a pharmaceutical compositions containing ticagrelor wherein the total weight of the ticagrelor is less than 30% w/w by total tablet weight. Basicly three types of manufactruing methods are available in the prior art. Advantageous and disadvantageous of the manufacturing methods are evaluated based on the information and experimental data procured from prior art documents. Quality by Design (QbD) principles were applied to the development of the ticagrelor comprising pharmaceutical composition. The critical quality attributes that have the potential to affect product performance have been identified using the principles as outlined in ICH Q8 and Q9. Solubility of ticagrelor is very low (not ionised in the pysiological pH range) and exhibits a moderate intrinsic permeability, there is potentially a higher risk that changes in formulation and processing parameters can affect clinical performance, and this was taken into account during development. Moreover, this low aqueous solubility of ticagrelor leads to an increase of relevance of particle size. Solubility studies in human intestinal fluids, bulk-tapped density, compressibility index, Hausner ratio, flowability studies of drug substance were performed prior to beginning of the development process of pharmaceutical drug product.

As shown in table above, ticagrelor solubility is very low in pysiological pH range.

In addition, bulk density and tapped density values of ticagrelor was investigated. By using the results obtained, compressibility index, Hausner ratio and flowability of drug substance was designated.

Drug substances with poor aqueous solubility and poor dissolution in the GI fluids is a limiting factor to the in vivo bioavailability after oral administration. Therefore, in vitro dissolution has been recognized as an important element in drug development and thus increasing the dissolution rate of poorly soluble drugs and enhancing their bioavailability is an important challenge to formulation designation studies.

Ticagrelor containing pharmaceutical drug products are designed based on all investment studies explained above. Micronizing particle size values and designation of polymorphism are the main approaches to improve physicochemical and biopharmaceutical properties of the poorly soluble drugs, thereby improving their solubility. These approaches are used for ticagrelor also.

Moreover, particle size distribution analysis for ticagrelor was performed. Results are shown in the table below. Polymorphism is a direct impact on physical and chemical properties of drug substance. Based on this information XRD anaysis was performed and confirmed that the drug substance used was polymorph II as it is in the innovator drug product.

In a preferred embodiment, suitable fillers may comprise but not limited to dibasic calcium phosphate dihydrate, polysaccharides, primarily microcrystalline cellulose, lactose, mannitol, sugars, sorbitol, sucrose, inorganic salts, primarily calcium salts and the like and mixtures thereof. The most preferred filler is microcrystalline cellulose.

In a preferred embodiment, the pharmaceutical composition comprises at least a disintegrant, preferably, it is selected from croscarmellose sodium, crospovidone, low-substituted hydroxypropyl cellulose, starch, sodium starch glycolate, carmellose and mixtures thereof. More preferably, the disintegrant is sodium starch glycolate.

In a preferred embodiment, suitable diluents may comprise but not limited to dibasic calcium phosphate dihydrate, polysaccharides, primarily microcrystalline cellulose, lactose, mannitol, sugars, sorbitol, sucrose, inorganic salts, primarily calcium salts and the like and mixtures thereof. The most preferred filler is mannitol, silicon dioxide or a mixture thereof, preferably is a mixture thereof.

In a preferred embodiment, the binder may be, for example, hypromellose, cellulose or cellulose derivatives, povidone, starch, sucrose, polyethylene glycol, or mixtures thereof. The binder is dissolved in a solvent, for instance, water, an alcohol, or mixtures thereof. In particularly advantageous embodiments of the present invention, the binder solution is hypromellose dissolved in water. In a preferred embodiment, the pharmaceutical composition comprises at least a lubricant, preferably, it is selected from sodium stearyl fumarate, magnesium stearate, calcium stearate talc, stearic acid and mixtures thereof. More preferably, the lubricant is magnesium stearate.

In order to obtain a targetted product profile, quality attributes have been defined and discussed, the manufacturing process has been selected based on these aspects and a control strategy has been defined.

Beginning from the well-known prior art documents and based on experimental studies, multiple formulations were designed to improve dissolution profile and granule characteristics of pharmaceutical compositions comprising ticagrelor. In a preferred embodiment, formulation design

Manufacturing method:

1. Ticagrelor, sorbitol, dibasic calcium phosphate dihydrate, hydroxypropyl cellulose and sodium starch glycolate are stirred in high-shear wet granulator for 4 minutes.

2. Sufficient quantity of deionized water was added on the powder blend in step 1 and granulation process is performed with a 10 minute period stirring.

3. Granule obtained in step 2 is dried at a specified temperature.

4. Dried granule is milled and lubricated with magnesium stearate for 3 minutes. 5. Final blend in step 4 is subjected to tablet compression process.

6. Tablets obtained in step 5 were coated with a proper film coating agent. The formulation design and manufacturing method were very similar to that of innovator drug product. However, the problems observed in ticagrelor drug substance like flowability was poor and stickiness were laso observed in powder blend.

In addition, disintegration analysis was performed on the tablets obtained as per USP Monograph <701>. The result is about 9 to 12 minutes. This value is very high for imediate release solid dosage forms. Moreover, disintegration time result is also the indication for dissolution rate. The longer the disintegration time means the longer the drug release from the formulation. In addition, it is known from the prior arts that in vitro studies have lineer correlation with in vivo drug release. In order to overcome disintegration problem and obtain a solid pharmaceutical composition with a good chemical ve physical powder blend characteristics, another embodiment was designed.

In an another embodiment, use of mannitol was prefered instead of sorbitol. In addition, microcrystalline cellulose was replaced by dibasic calcium phosphate dihydrate not to have stickiness and flowability problem.

Manufacturing method:

1. Ticagrelor, mannitol, microcrystalline cellulose, hydroxypropyl cellulose and sodium starch glycolate are stirred in high-shear wet granulator for 5 minutes.

2. Sufficient quantity of deionized water was added on the powder blend in step 1 and granulation process is performed with a 10 minute period stirring.

3. Granule obtained in step 2 is dried at a specified temperature.

4. Dried granule is milled and lubricated with magnesium stearate for 4 minutes. 5. Final blend in step 4 is subjected to tablet compression process.

6. Tablets obtained in step 5 were coated with a proper film coating agent.

It is observed that the flowability problem was overcomed. However, stickiness problem was still ongoing. Obtained tablets were analyzed for disintegration time, the result is about 1 minute which is good for immediate release solid dosage forms. Thus, in vitro dissolution profile analysis was performed on the tablets tablets.

Dissolution conditions are performed in the following conditions: Dissolution medium: Water + 0.2% Polysorbate 80 Volume of dissolution medium: 900 ml Apparatus: Paddle Temperature: 37±0.5°C Time: 75 minutes Rotation speed: 50 rpm

Table V: Results of dissolution analysis

Dissolution results of Formulation II were higher than the dissolution release profile of innovator drug product.(Fig.1)

Comparison of dissolution profiles is shown in the graph. It was observed that the dissolution profile of formulation II presents higher dissolution profile pattern than innovator drug product. Dissoluton study is the major indicator of bioavailability study. The more similar dissolution profile to innovator drug product means closer to achieve pharmaceutical formulation wtih proper in vivo drug release profile. In another embodiment designated as Formulation III, was designed to lower the dissolution rate which ticagrelor drug substance release under specified conditions. In addition, modifications on existed critical excipients were planned to be made to overcome stickiness problem. It is known form the prior art, the amount of magnesium stearate has direct impact on dissolution rate. The solvent penetration weight decreased as the percentage of MgSt concentration increased. As a nonsoluble lubricant, MgSt does not allow water to flow through the waxy covering of the powders.

One of critical excipients in the present invention is microcrystalline cellulose for use both in extragranular and intragranular phases taht plays a critical role in compactibility and dissolution rates.

Use of more than one glidants can greatly increase hydrophobicity (the resistance of a particle to absorb water), which lowers the solvent penetration rate (speed of absorption). When a tablet does not allow solvents to penetrate, it will have a slow dissolution, or dissolving rate. Based on this information, an extra diluent silicon dioxide was considered to be used in the next embodiment.

In order to modify the previous formulation (Formulation II), changes made are listed below: Increase in Microcrystalline cellulose content o Total amount used in the formulation will be increased to get functional effect of not only filler, diluent as well to overcome stickiness problem o The amount of Microcrystalline cellulose in the intragranular phase was kept constant. Added amount which makes an increase in Microcrystalline cellulose was located in extragranular phase. It was planned to define the quantity of Microcrystalline cellulose in extragranular phase during manufacturing process in the presence of other excipients. Addition of silicon dioxide as diluent o Silicon dioxide was planned to be used in the formulation to get functional effect of diluent to overcome stickiness problem. As it is also known from the prior art, silicon dioxide is widely used in pharmaceuticals. Its small particle size and large specific surface area give it desirable flow characteristics that are exploited to improve the flow properties of dry powders in a number of processes such as tableting.

The amounts of critical excipients was considered to be adjusted during manufacturing process. Manufacturing method:

1. Ticagrelor, mannitol, specified amount of microcrystalline cellulose, specified amount of hydroxypropyl cellulose and sodium starch glycolate are stirred in high-shear wet granulator for 5 minutes.

2. Granulation solution was prepared by dissolving specified amount of hydroxypropyl cellulose in deionized water.

3. Sufficient quantity of granulation solution was added on the powder blend in step 1 and granulation process was performed with a 10 minute period stirring.

4. Granule obtained in step 3 is dried at a specified temperature.

5. Dried granule was sifted and remaining amounts of microcrystalline cellulose and hydroxypropyl cellulose and also silicon dioxide were added and stirred.

6. Obtained granule in step 5 was lubricated with magnesium stearate for 4 minutes.

7. Final blend in step 6 is subjected to tablet compression process.

8. Tablets obtained in step 7 were coated with a proper film coating agent. Flow properties were good, compression was performed succesfully. Disintegration time analysis and in vitro dissolution study was performed.

Disintegration time was found about 2 minutes. As it is observed from the Fig.2 , Formulation III presents more similar in vitro dissolution profile with respect to innovator drug product with adjustment of the amounts of critical excipients used in the formulation.

The limiting factor for the appearance of the drug in the blood could be the pure permeability through the intestinal membrane the release from the drug dosage form, or the solubility of the active ingredient. In case of solubility-limited product, the absorption could be governed by physico-chemical characteristics of the API (such as solubility, dissolution rate, particle size, crystal shape, polymorphism) and not by its permeability or formulation.

Based on this information, obtaining proper in vitro dissolution results will lead the formulation to achieve proper in vivo profile. During wet granulation manufacturing, the amount ratio of Microcrystalline cellulose used in extragranular phase is between 20-30% w/w by total weight of the composition illustrated as Formulation III.

It was surprisingly found that the use of microcrystalline cellulose together with silicon dioxide in the extragranular phase and in a specified range of amount ratio directly affects the in vitro dissolution profile of solid dosage form comprising ticagrelor as active ingredient.

The ratio of microcrystalline cellulose and silicon dioxide in Formulation III is between 35:1 to 45:1. While the invention has been described with respect to the above specific embodiments, it should be recognized that various modifications and changes may be made to the invention by those skilled in the art which also fall within the scope of the invention as defined by the appended claims.

Brief description of the drawings:

Fig.1 Dissolution results of Formulation II Fig.2 Dissolution results of Formulation III