Abstract: The present invention discloses a Dasatinib formulation composition in the form of self-nano emulsifying drug delivery system by spontaneously formation of nano- emulsion with immediate contact of gastric fluids. The pharmaceutical composition of Dasatinib shows an improved solubility, dissolution rate, enhanced drug loading ability, excellent drug release and bio availability of poorly soluble drugs along with 10 high stability. The composition comprises of an effective amount of a Dasatinib, an oil phase, a surfactant and a cosurfactant.
Technical Field of the Invention
The present invention relates to a Dasatinib formulation composition in the form of self-nano emulsifying drug delivery system and the method of preparation thereof.
The pharmaceutical composition of the present invention shows an improved solubility, enhanced drug loading ability, excellent drug release and highly stable.
Background of the Invention
Poor bioavailability of drugs has been a major limitation in the successful utilization of many therapeutic effective molecules. As it happens, most of these molecules are lipophilic in nature and tend to be poorly absorbed in the aqueous medium present in the gastrointestinal (GI) tract. The problem of poor bioavailability is at times further compounded by a faster elimination rate which further reduces the efficiency of such molecules being used as a drug target of choice.
Dasatinib is an ATP-competitive protein tyrosine kinase inhibitor. The main targets of Dasatinib are BCR/Abl (the "Philadelphia chromosome"), Src, c-Kit, ephrin receptors, and several other tyrosine kinases. Strong inhibition of the activated BCR-ABL kinase distinguishes Dasatinib from other CML treatments, such as imatinib and nilotinib. Although Dasatinib only hasaplasma half-life of three to five hours, the strong binding to BCR-ABL1 results in a longer duration of action. The structure of Dasatinib is based on a chemical scaffold different from that of imatinib, and has a 325-fold greater potency, with the ability to bind both the inactive and active conformations of the ABL kinase domain.
Figure 1: Structure of Dasatinib
Dasatinib has been approved for the treatment of patients with chronic phase disease at a recommended dose of 100 mg once daily (regardless food assumption), and for patients with accelerated phase and blast phase at a recommended dose of 140 mg once daily. It is possible to assume the drug regardless food assumption. This review summarizes the relevant clinical data from several trials using Dasatinib first-line and second- line in patients with chronic myeloid leukemia (CML).
In a press release on January 2, 2019 “Bristol-Myers Squibb Company announced the U.S. Food and Drug Administration (FDA) has expanded the indication for Sprycel ® (dasatinib) tablets to include the treatment of pediatric patients one year of age and older with newly diagnosed Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) in combination with chemotherapy. Sprycel is the only second-generation tyrosine kinase inhibitor approved for this patient population. The approval, which was granted following priority review by the FDA, is based on data from the Phase 2 study, CA180-372 (NCT01460160). “We recognize the urgency around developing and delivering the rapies for children and young adults living with cancer, and today’s approval is an important example of our commitment to pediatric oncology,” said Jeffrey Jackson, Ph.D., development lead, hematology, Bristol-Myers Squibb. “Building on our previous indication for children with Ph+ chronic myeloid leukemia in chronic phase, we’re pleased to bring Sprycel tablets to a second type of pediatric leukemia. This approval will give physicians another treatment option to offer appropriate pediatric patients with Ph+ ALL.” The announcement indicates and analysis the future opportunities and forecast the market demand, size, share and growth for dasatinib formulation development.
Oral administration is regarded as the preferred route of drug intake offering numerous advantages including convenience, ease of compliance, potential for availability to large patent population and cost effectiveness. Thus oral bioavailability is a key factor in lead selection and development of new drugs.
Poor oral bioavailability affects the drugs performance and leads to inter and intra patient variability. A number of chemotherapeutic as well as chemo preventive agents suffer from poor oral bio availability rendering the mun suitable for oral delivery. Oral bio availability depends primarily on Drug permeability, aqueous solubility, dissolution rate, presystemic metabolism, first-pass metabolism and susceptibility to efflux mechanisms. Of these low permeability and poor solubility are the most common causes of poor oral bioavailability. The advances in understanding the cause of poor bioavailability have led to significant improvements in the design of technologies to combat these deficiencies. The strategies to
improve oral bioavailability can be grouped into three main groups comprising: Pro-drugs and drug conjugates, Medicinal chemistry, and Formulation design. The present invention proposes the application of formulation design to enhance the solubility there by drug release of selected drug candidate.
Formulation Design is often the route of choice for modifying the oral bioavailability of drugs as it offers a low cost and rapid solution to these problems particularly for drug already in the market. As opposed to pro-drug and medicinal chemistry approaches, Formulation Design does not require chemical modification of the drug or creation of new chemical entities. This provides considerable advantage in terms of reduced cost and development timeline. Poor aqueous solubility and dissolution rate frequently affect the oral performance of drugs. This issue has been successfully addressed in the art by using techniques such as, co-solvents, micronization, solid dispersions, surfactants, nano-suspensions, micro emulsions and self-emulsifying drug delivery systems (SEDDS), pre-formed emulsions/phospholipid complexescontainingthelipophilicentityhavebeenatriedandtestedmethodtoachievea
better solubility and absorption. This generally involves forming ‘Lipids micelles’ of the lipophilic entity with the help of suitable surfactant(s). Such micelles are then delivered as such at the absorption site. US Patent Publication
A study by M. Yasmin begum et al, (formulation and evaluation of Dasatinib loaded solid lipid nanoparticle) concluded that SLNs are used as drug carriers for lipophilic drugs to
enhance the bioavailability of poorly watersoluble drugs through nanoparticles, as a drug delivery system.
The advent of the Self emulsifying drug delivery systems (SEDDS) technique witnesses a marked improvement in the bioavailability of the lipophilic moieties. SEDDS comprise of an isotropicmixture of drug,oil,surfactantand/orco-solventswhichuponoraladministration gets emulsified in the aqueous media in the GI tract. The distinguishing feature of SEDDS is its ability to emulsify spontaneously to produce fine oil-in-water emulsions when introduced into an aqueous phase under gentle agitation. The resulting oil-in-water emulsion is thermodynamically stable due to the relatively small volume of the dispersed oil phase, the narrow range of drop let sized is tribution and the polarity of the oildroplets (GrovesMJ, Degalindez D A, The self-emulsifying action of mixed surfactants in oil, Acta Pharm Suec, 13, 1976, 361-372). The oil-in-water emulsion shows higher absorption in the GI tract. This approach has found a general acceptance for the lipophilic drugs that suffer frompoor absorption rates. The SEDDS approach is being successfully followed in commercially available formulations containing cyclosporin A, ritonavir andsquinavir.The success of the
self emulsifying technique in increasing the bioavailability of the drug depends on the oil-surfactant pair, surfactant concentration and the temperature at which self emulsification occurs. It is also widely understood that the droplet size also plays a key role indeterminingtheabsorptionrateandhencetheoverallbioavailabilityofthedrugmolecule, as a small droplet size provides a large interfacial area for its absorption.
With the recent developments in nanotechnology, oil droplets of nano dimensions have been achieved. These nano sized oil droplets are more effective in increasing the bioavailability of the drug molecule, courtesy their size. These drug delivery systems are called Self Nano Emulsifying Drug Delivery System (SNEEDS). SEDDS now represent abroad category typically encompassing emulsions with a droplet size ranging between a few nanometers to several microns, while SNEDDS is used specifically where oil droplets are below 100 nm in size.
Itis, therefore, aneed in theart to develop aself-emulsifying Dasatinib composition which has an enhanced drug loading ability as well as an increased bioavailability and better stability. Further, a self-emulsifying Dasatinib composition of such nature must also be able to address the lacunas present in the existing art, specifically as elaborated in the preceding paragraphs. Further, it would be desirable that such a composition is based on creating a nano emulsion at the absorptions iteto markedly increase the absorption efficiency. Compared to conventional dosage forms SNEDDS are advantageous as it involves ease of manufacture and scale-up, selective targeting of drug(s) toward specific absorption window in GIT, enhanced oral bioavailability by increasing solubility and reducing the dose, thereby promoting efficient drug transport, quick onset of action and also SNEDDS has a much larger surface area and free energy. Thus, there is aneed for highly efficient composition of dasatinib with anticancer activity. Dasatinib, an anticancer agent with poor aqueous solubility is best suitable for the development of SNEDDS.
Brief Summary of the Invention
Accordingly, it is an object of the invention to provide a pharmaceutical formulation composition of Dasatinib having high drug delivery system by spontaneously emulsified in situ when exposed to gastrointestinal tract (GIT) fluids, forming oil-in-water nano-emulsions.
It is another object of the invention to provide a drug delivery system for a Dasatinib based formulation composition that has a high drug loading capacity and increased oral bioavailability along with increased solubility.
Another object of the invention to provide a stable drug delivery system for a Dasatinib based formulation composition that does not require the usage of a polymeric molecular aggregation inhibitor to maintain the stability of the composition.
Yet another object of the invention to provide a stable drug delivery system for a Dasatinib based formulation composition that does not result in precipitation out of aqueous medium once the nano emulsion is formed, even in the presence of surfactant and co-surfactants. In an exemplary embodiment of the present invention is to provide a process for the preparation of formulation composition of Dasatinib for self-nano emulsifying drug delivery system with immediate contact of gastric fluids.
In another exemplary embodiment of the present invention is to provide a process for the preparation of formulation composition of Dasatinibfor self-nano emulsifying drug delivery system that are typically used for prevention of cancer disorders.
Brief Description of the Drawings
The present invention will be more readily understood from the detailed description of
embodiments thereof made in conjunction with the accompanying drawings of which:
Fig 1: Structure of Dasatinib
Fig 2: Solubility of Dasatinib drug various oils, surfactants and co-surfactants
Fig 3: Phase diagram with emulsification region.
Fig 4: Phase diagram for 70mg loadeddrug
Fig 5: Phase diagram for 100mg loaded drug
Fig 6: Phase diagram for 140mg loaded drug
Fig 7: Dissolution profile of Dasatinib Pure drug and formulations F1to F15
Fig 8: FTIR Spectroscopy of Dasatinib puredrug
Fig 9: FTIR Spectroscopy of Dasatinib optimized formulation F12
Fig 10: particle size of Dasatinib optimized SNEDDS formulation F12
Fig 11: Zeta potential of the optimized formulation F12
Fig 12: Scanning Electron Microscopy of Dasatinib optimized formulation (F12)
Detailed Description of the invention
The present invention relates to provide a pharmaceutical formulation composition of Dasatinib having high drug delivery system by spontaneously emulsified in situ when exposed to gastrointestinal tract (GIT) fluids, forming oil-in-wate nano-emulsions. The said formulation composition that has a high drug loading capacity and increased oral bioavailability along with increased solubility.
Main object of the invention to provide a process for the preparation of formulation composition of Dasatinib for self-nano emulsifying drug delivery system with immediate contact of gastric fluids. Wherein the stable drug delivery system does not require the usage of a polymeric molecular aggregation inhibitor to maintain the stability of the composition. TheDasatinibformulationcompositiondoesnotresultinprecipitationoutofaqueous
medium once the nano emulsion is formed, even in the presence of surfactant and co-surfactants.
In another exemplary embodiment of the present invention is to provide a process for the preparation of formulation composition of Dasatinib for self-nano emulsifying drug delivery system that are typically used for prevention of cancer disorders.
The present invention describes a pharmaceutical composition comprising Dasatinib as a self-nano emulsifying drug delivery formulation with an improved solubility, enhanced drug loading ability, greater drug release and better stability.
In an embodiment, includes a said formulation composition with effective amount of Dasatinib, an oil phase, a surfactant and a co-surfactant, preferably Dasatinib 100-150 mg and 20-80% (w/w) of the oil phase; 15-57% (w/w) of the surfactant, and 5-19% (w/w) of the co surfactant.
The Hydrophilic-Lipophilic Balance (HLB) of the oil phase is between 2 and 10, more preferably between 4 and 8. The HLB value provides a means for ranking surfactants based on the balance between the hydrophilic and lipophilic portions of the surfactant or emulsifying agent and the higher the HLB number, the more hydrophilic the surfactant or emulsify ingagent.
In an embodiment, the surfactants of the composition of the present invention are selected from ionic or non-ionic surfactants.
In a preferred embodiment, the surfactants used in the present invention comprise non-ionic surfactants. In yet another embodiment, the surfactant has an HLB between 9 and 18. The surfactant is capable of forming a stable emulsion, preferably a fine emulsion and more preferably a Nano emulsion of the present composition when it is brought into contact with aqueous fluid, such as in the G.I. tract. The surfactant of the present invention does not precipitate out the active ingredient Dasatinib from the emulsion, and hence offers better stability of the formulation than the existing art. It also does away with the use of polymeric molecular aggregation inhibitors, as used in the existing art, to avoid the formation of Dasatinib precipitation.
In an embodiment, the co-surfactants of the present invention is selected from a group comprising polyethylene glycol chain lengths preferably having a molecular weight of 200 to 600), propylene glycol derivatives. In an embodiment, the co-surfactants of the present invention also act as solubilizers in the resulting composition.
The Dasatinib composition of the present invention forms an oil/water Nano emulsion instantaneously when brought into contact with the aqueous medium of the GI tract with mild agitation provided by gastric mobility in the tract region. The formation of nano emulsion leads to a superior absorption and enhanced bioavailability of the Dasatinib enabling reduction in dose, more consistent temporal profiles of drug absorption, and protection of drugs from the hostile environment ingut.
In a preferred embodiment, the droplet size of the composition of the present invention is 35-45 nm. The composition of the present invention, by virtue of its choice of surfactants, does not lead to any precipitation of the active ingredient, and subsequently also does away with the use of any polymeric molecular aggression inhibitors, thus providing a composition that is effective and stable over a wide period of time. Further, a stable self-nano emulsifying composition with a Dasatinib loading ability of 100mg, hitherto unachieved, is a marked advancement over the existing art and paves way for further research and development on Dasatinib as a better, safer and efficient drug of choice.
The Dasatinib composition of the present invention is ideal for oral delivery systems, since they are homogeneous, thermodynamically stable, have uniform droplet sizes, and are optically clear. The Dasatinib composition of the present invention can be administered in the form of liquid or solid dosage form. In an embodiment, when administered as a liquid dosage
form, the composition is filled in hard or soft gelatin capsules. Oral unit dosage forms in accordance with the present invention will suitably comprise from 100 mg of the Dasatinib. The dosage of the drug and the number of times it is administered to the patient will vary depending on several factors including but not limited to the age of the patient, the severity of the condition of the patient, past medical history, among other factors, and will be determined by the physician in his sound discretion.
The composition of the present invention is preferably administered to mammals, such as dog, cat, horse, pig, mice, rat and especially humans. When the composition of the present invention is prepared in the form of a soft or hard be apparent to those skilled in the art. Such changes and modifications may be made without departing from the scope of the invention.
Solubility
The solubility of Dasatinib in various oils (Miglyol 810, Capryol 90, Triacetin, Caprylic acid, Imwitor, Castoroil, AkomedE, NeobeeM5, Oleicacid and cornoil), Surfact ants(brij35, tween 20, Cremophor EL, Labrasol, Cremophor RH 40, Tween 80), Co-surfactants (PEG-400, PEG-600, Propylene glycol, lauro glycol) was studied by adding excess amount of Dasatinib in 2 ml vehicle each in vial and heated in hot water bath at 40ᵒC for solubilising the drug. The samples were subjected to further mixing using vortex mixer and kept aside for 48hours in orbital shaker at room temperature to attain equilibrium solubility. Later these vials were
centrifuged at 3000 rpm for 10mins, the excess drug gets settled and from the supernatant the drug concentration is quantified by using UV spectrophotometry at 315nm. The results of the solubility test are provided in table 1and figure 2.
From the solubility studies it was evident that medium chain triglycerides were highly influencing solubility of the drug; these are obtained from vegetable source and are resistant to oxidation. Triacetin being a medium chain triglyceride with high solvent capacity was showing high degree of solubility for Dasatinib. The water solubility of Dasatinib is only very low 0.0035mg/ml. Surfactant and Co-Surfactants with high HLB values were useful in reducing the surface tension between oil and fluids in which drug is solubilized. Based on results from the solubility study, Triacetin, brij 35 and propylene glycol were showing high solubilizing efficiency for Dasatinib.
Construction of Pseudo ternary phase diagrams
For the construction of phase diagram and an Oil, surfactant and co-surfactant chosen from solubility studies were mixed in different ratios of oil and Smix (surfactant: co-surfactant) ranging from 1:9 to 9:1 were prepared and filled invials and thoroughly shaken. The Smix ratios chosen were 1:1, 2:1, 3:1 and 4:1. Measured quantities of the vehicles were filled into 2ml vials using micro-pipette and shaken thoroughly. From this mixture one ml is taken and 100ml of water was added and gently agitated, the ratios which spontaneously formed Nano-emulsion upon gentle agitation with water, and showed no phase separation and no turbidity were separated and checked for transmittance values, the mixtures with transmittance value greater than 80 were noted, these ratios were used to plot ternary phase diagram using CHEMIX software. The apexes of the triangle were labelled representing each of the vehicles Varying ratios of oil, surfactant and co-surfactant mixed and agitated with water and checked for no phase separation and no turbidity, the ratios showing no phase separation and clear in appearance were separated and checked for transmittance. The ratios exhibitingtransmittance more than 80% were considered for constructing pseudo ternary phase diagram. All the values were fed in CHEMIX software and connected and the region was shaded. The shaded region in Pseudo ternary phase diagram represents the miscibility region where the oil, surfactant and co-surfactant are completely miscible with each other and spontaneously form nano emulsion on simple agitation with GI fluids (table 2 and figure3).
Effect of Dasatinib loading
The drug loading has considerable influence on the globule size and phase behavior of the spontaneously emulsifying systems. In the view of this, effect of Dasatinib loading on the transmittance, phase behavior and area of nanoemulsion formation was studied selected oil-surfactant and co-surfactant compositions. (Triacetin oil – brij 35–propylene glycol compositions as mentioned above).
Seventeen compositions of varying ratios of triacetin oil–brij35–propyleneglycolwere taken and in 1ml composition of each ratio were incorporated with 70 mg, 100 mg and140 mg of Dasatinib (i.e. 15*3=45 formulations). Required amount of Dasatinib was added to the screw capped glass vials containing required amount of surfactant and co-surfactant. Drug was solubilized using a vortex mixer or by heating at 40° C in a water-bath wherever necessary. Finally required amount of oil was added to the vialsandvortex mixed for 2min for proper mixing. The transmittance of the resulting dispersions up on diluting 25 mg of the formulations with 50 mL distilled water was measured using UV spectrophotometer at 550nm. The area of Nano emulsification region was identified as described above by constructing pseudo-ternary phase diagrams, it was found that 100mg loaded ratios of SNEDDS formulation showed maximum emulsification area indicating spontaneous and clear emulsion formation compared to 70mg and 140 mg. High drug loading of 140mg may have led to precipitation of the drug thus showed less Nano emulsification region.
Development of Dasatinib SNEDDS formulations
Fifteen ratios were considered for formulating Dasatinib SNEDDS from the emulsification region of pseudo ternary phase diagram. Based on solubility studies conducted for choosing vehicles for the formulation, triacetin as oil phase, Brij 35 and propylene glycol as surfactant and co-surfactant respectively were screened for formulation of Dasatinib SNEDDS. Dasatinib(100mg) added too ilin to the glass via land heated hot water that40ºCuntil drug solubilized. Then to this oily mixture were added surfactant and co-surfactant and sonicated for 15min. The quantity of oil, surfactant and co-surfactant used are mentioned in table5.
A series of SNEDDS (F1- F15, the composition was shown in table 3) which showed transmittance values more than 90) were selected from 100 mg loaded Dasatinib system and prepared as described above. About 1ml of the formulation (equivalent to 100 mg of the Dasatinib) was filled in size ‘00’ hard gelatin capsules, sealed and stored at ambient temperature (25° C) until used. These SNEDDS were evaluated for visual observations, turbidity, effect of pH of the dispersion media on globule size and zeta potential, robustness to dilution and invitro dissolution study and were optimized.
EVALUATIONS
Visual Observations
To assess the self-emulsification properties, Dasatinib SNEDDS (25 mg) was introduced into 50mLofdistilledwaterinaglassErlenmeyerflaskat37°Candthecontentsweregently
stirred manually. After equilibrium, time of self-emulsification, dispersibility and appearance were observed and rated according to grading system (table 4).
It was observed from the results that Visual observations indicated that at higher levels of surfactant, the spontaneity of the self-emulsification process was increased. This may be due to excess penetration of water into the bulk oil causing massive interfacial disruption and ejection of droplets into the bulk of aqueous phase. When a co-surfactant, propylene glycol was added to the system, it further lowered the interfacial tension between the o/w interfaces and also influenced the interfacial film curvature (table5).
Turbidity Measurement
TurbidityoftheprepareddispersionswasmeasuredusingNepheloturbiditymeterusing30 mL of the dispersion. The Nephelo turbidity meter was carefully calibrated with formazin standard solution before the measurements. It is seen from the results that turbidity values (NTU) have been reported to be of use in SNEDDS characterization. From these results it can be generalized that the formulations that have low turbidity (<20) gave a transmittance values of more than 90 indicating rapid and spontaneous emulsification within 1min, hence it givesa good correlation between transmittance and turbidity values (table 5).
Robustness to Dilution
Robustness of Dasatinib SNEDDS to dilution was studied by diluting 25 mg of SNEDDS with 50, 100 and 1000 mL of distilled water, 0.1N HCl, pH 4.5 acetate buffer and pH 6.8 phosphate buffers. The diluted nanoemulsions were stored for 24 hours and observed for any signs of phase separation or drug precipitation. Nanoemulsions resulting from the dispersion of Dasatinib SNEDDS (F1-F15) with distilled
water, 0.1N HCl, pH 4.5 acetate buffer and pH 6.8 phosphate buffer were found to be robust to all dilutions and no separation or drug precipitation was observed even after 24 hours of storage.
Percentage drug content
All the formulations of Dasatinib - loaded SNEDDS were subjected to assay analysis in order to determine their percentage drug content. Accurately weighed samples were dissolved individually in 10mL of methanol and stirred by vortex mixer for a period of 10 min. Each of
the solutions was filtered, and the drug content of each filtrate was estimated UV spectrophotometrically against blank at 315 nm.
The drug content of all formulations ranged between 91.36 to 99.98% with maximum value exhibited by F12 (table 6).
Entrapment efficiency
A known quantity of Dasatinib loaded SNEDDS mixed with 100mL phosphate buffer (pH 6.8) and kept in dark for 24h.the contents filtered, filtrate diluted and analyzed for drug content by UV for the drug content at 315nm.Entrapmentefficiencywas calculated by formula
Experimental drug content x 100
Drug entrapment efficiency =
Theoretical drug content The entrapment efficiency of all formulations vary between 91.73 to 99.72% with maximum value displayed by F12 (table6).
Thermodynamic stability test
Thermodynamic stability studies were carried out for the all formulations. SNEDDS were subjected to centrifugation at 18,000 rpm at 4 °C for 30 min. The stable formulations with no phase separation were further subjected to 6 heating and cooling cycles by incubating them for 48 h at 45 °C and 4 °C, respectively. The formulations that remained stable at former conditions were proceeded to 3 freeze–thaw cycles between -21 °C and 25 °C, and monitored for the time-dependent physical changes like drug precipitation
Fifteen formulations of Dasatinib SNEDDS were subjected to thermodynamic stability studies and checked for any phase separation, all the fifteen formulations passed the centrifugation test by showing no phase separation of phases. The formulations were also subjected to freeze thawing for 2days at -20ᵒC and +40ᵒC for 2days, all the formulations were stable and passed the stability test.
In vitro dissolution studies of Dasatinib SNEDDS formulations
In vitro dissolution test was carried out by using USP type II (paddle) apparatus. The liquid SNEDDS whose weight equivalent to 100mg of Dasatinib were filled into hard gelatin capsulesandaddedto900mlofacetatebufferpH4with1%tritonX-100wasusedas dissolution medium and the paddle was rotated at 60 rpm at temperature (37°C ± 0.5°C). In specified time intervals (0,5,10,15,20,25,30,35,40,45min) an aliquot of 5ml samples of the solution were withdrawn from the dissolution apparatus and with replacement of fresh fluid to dissolution medium. The samples were filtered through filter paper of 0.45 µm. Absorbance of these solutions were measured at λ max 315nm using a UV/Visible Spectrophotometer (ELICO164). The drug release was plotted against time to determine the release profile of various batches.
Fifteen formulations with 100mg Dasatinib drug loaded SNEDDS were formulated and in-vitro dissolution studies were conducted, as it is a solubility enhancement technique all the fifteen formulation were showing good drug release compared to pure drug (30.25%) which indicates formation of Nano-emulsion. The formulation when comes in contact with the dissolution media gets self emulsifies and form an oil in water emulsion stabilized by surfactants and co-surfactants used in the formulation. Surfactants and co-surfactants act by reducing interfacial tension between oil and water, and also in general as the particle size is small with more surface area, more is the absorption and hence more is the drug release, and out of all formulation F12 was showing maximum drug release of 99.96% at the end of 60minutes with more Smix in the ratio compared to other formulations (table 8 and figure 7).
Accelerated stability studies
All formulations filled in hard gelatin capsules were packed in HDPE screw capped bottles and kept in humidity chambers maintained at 40 ± 2°C/ 75 ± 5% RH as per ICH guidelines for Zone III and stored for 6 months. Samples were evaluated for drug
content and drug release (ICH Harmonized Tripartite guideline on “Stability Testing of New Drug Substances and Products Q1A (R2)”, 6 February2003).
Stability testing is to provide evidence on how the quality of a drug substance or drug product varies with time under the influence of a variety of environmental factors such as temperature, humidity and light and enables recommend storage conditions.
No visible physical changes were observed in all the formulations withdrawn from the humidity chambers. The samples were assayed for %entrapment efficiency, % drug content and in-vitro drug release and the results are shown in Table 20. No significantdifferencewasobservedafterstorageatacceleratedconditionsat40±2° C/75±5% RH for a period of six months (table 9).
FTIRstudies
An FTIR-8400S Spectrophotometer (Shimadzu, Japan) equipped with attenuated total reflectance (ATR) accessory was used to obtain the infrared spectra of drug in the isotropic mixtures of excipients. Analysis of pure drug i.e., Dasatinib and physical
mixtures of the drug with the excipients were carried out using diffuse reflectance spectroscopy (DRS)-FTIR with KBr disc. All the samples were dried under vacuum prior to obtaining any spectra to remove the influence of residual moisture. For each the spectrum, 8 scans were obtained at a resolution of 4 cm-1 from a frequency range of 400-4000 cm-1.
FTIR spectrums are mainly used to identify any interactions between the pure drug (Figure 8) and any of the excipients used. The presence of all these peaks gives conformation about purity of the drug. From the IR study the major peak of Dasatinib (Fig.8) were found to 1629.90 cm-1 (NH bending), 1454.38 cm-1 (C-Hstretching), 1126.47 cm-1(C-C stretching), 1287.77 cm-1 (O-H stretching groups), 774.89 cm-1 (N-H stretching), at 1292.35 cm-1 which is associated with O-H stretching, 1587.47 cm-1(stretching of N-H group). The FTIR spectra of optimized formulation (Figure 9) were having similar fundamental peaks and pattern as seen in pure drug Dasatinibandalsoitisobservedthatfromthereisappearanceoffunctionalgroups related to the excipients used in the formulation, carbonyl stretch of triacetin at 1770.98 cm-1, c-c stretching of triacetin at 1060.34 cm-1, 2859.17 cm-1corresponds to C-H stretch of CH2 groups of brij 35 and 1045.62 corresponds to -C-OH stretching of propyleneglycol.
From the FTIR studies it can be concluded that the peaks of drug remains intact and no interaction was found that the drug and excipients and were compatible. Frequencies of functional groups and unique absorption bands of pure drug remained intact in optimized formulation containing different excipient. Hence, there was no major interaction between the drug and excipients used in thestudy.
Particle size distribution
One gram of Dasatinib SNEDDS Formulations (F12) was diluted in 200 ml of 0.1N HCl. The droplet size/distribution of the prepared solution was determined using MALVERN Particle Size Analyzer (Model zetasizer Ver. 6.01, Malvern Instruments,
UK). The average Particle size was determined to be 44.3 nm with Z-Average of 30.4 nm indicating all the particles were in the nanometer range (figure 10).
Zeta potential of SNEDDS
The emulsion stability is directly related to the magnitude of the surface charge. In conventional SNEDDS, the charge on an oil droplet is negative because of the presence of free fatty acids. Zeta-potential of the resulting Nano emulsion was determined simultaneously while measuring particle size using a zetasizer Zeta potential is responsible for the degree of repulsion between adjacent, similarly charged, dispersed droplets. A zeta potential value of ±30 mV is sufficient for the stability of a micro emulsion. The zeta potential of the optimized SNEDDS formulation (F12) was found to be -26.8 mV which comply with the requirement of the zeta potential for stability. (Figure 11)
Scanning electron microscopy (SEM) for Dasatinib SNEDDS
Shape and surface morphology of microspheres was studied using scanning electron
microscopy (SEM). The SNEDDS after converting to emulsion were mounted on
metal stubs and the stub was then coated with conductive gold with sputter coater
5 attached to the instrument HITACHI, S-3700N (Ruan et al., 2003).
Scanning electron microscope studies of optimized formulation of Dasatinib (F12) revealed oval shaped globules. The size is within nanometers. There are clear liquid droplets without any pores (Figure 12A, 12B).
A B
Figure 12: Scanning electron microscopy of Dasatinib optimized formulation (F12)
15
Page 24 of 28
4. CLAIMS We Claim
1. A pharmaceutical composition of a self-nanoemulsifying drug delivery system
comprising:
a. an effective amount of Dasatinib or its pharmaceutically acceptable salts or
solvates or hydrates of about 100 to 150 mg by weight of composition,
b. an oil phase of about 20 to 80% by weight of composition,
c. a non-ionic surfactant of 15 to 60% by weight of composition,
d. a co-surfactant of about 5 to 19% by weight of composition,
wherein the composition is spontaneously emulsified in situ when exposed to gastrointestinal tract (GIT) fluids, forming oil-in-water nano-emulsions, which is substantially free of polymeric molecular aggregation inhibitor.
2. ThepharmaceuticalcompositionofSelf-NanoemulsifyingDrugDeliverySystem (SNEDDS) as claimed in claim 1, is characterized by an improved oral bio availability of Dasatinib high drug delivery system both In Vitro and In Vivo Evaluation.
3. The pharmaceutical composition of Self-Nanoemulsifying Drug Delivery System (SNEDDS) as claimed in claim 1, wherein oil phase is selected from the group consisting of Miglyol 810, Capryol 90, Triacetin, Caprylic acid, Imwitor, Castor oil, Akomed E, Neobee M5, Oleic acid and corn oil.
4. The pharmaceutical composition of Self-Nanoemulsifying Drug Delivery System (SNEDDS) as claimed in claim 1, wherein oil phase is preferably Triacetinoil.
5. The pharmaceutical composition of Self-Nanoemulsifying Drug Delivery System (SNEDDS) as claimed in claim 1, wherein the non-ionic surfactant is selected from the group consisting of brij 35, Tween 20, Cremophor EL, Labrasol, Cremophor RH 40, Tween 80, preferably brij35.
6. The pharmaceutical composition of Self-Nanoemulsifying Drug Delivery System (SNEDDS) as claimed in claim 1, wherein the co-surfactant is selected from the group consisting of PEG-400, PEG-600, Propylene glycol, lauro glycol, preferably propyleneglycol.
7. The pharmaceutical composition of Self-Nanoemulsifying Drug Delivery System (SNEDDS) as claimed in claim 1, wherein the composition comprises surfactant and co-surfactant that does not allow precipitation of Dasatinib from the emulsion thereby provides high stability of the formulation.
8. The pharmaceutical composition of Self-Nanoemulsifying Drug Delivery System (SNEDDS) as claimed in claim 1, wherein the composition comprises the higher drug loading of about 150 mg of Dasatinib that does not allow precipitation of Dasatinib.
9. The pharmaceutical composition of Self-Nanoemulsifying Drug Delivery System (SNEDDS) as claimed in claim 1, wherein the composition is administered in liquid dosage form, that is delivered through hard or soft gelatin capsules.
10. The pharmaceutical composition of Self-Nanoemulsifying Drug Delivery System (SNEDDS) as claimed in claim 1, wherein the said composition is stable at 40° C and 75 relative humidity for at least about 6months.
| # | Name | Date |
|---|---|---|
| 1 | 202041041569-Correspondence_Online Submission_ 03-11-2020.pdf | 2020-11-03 |
| 1 | 202041041569-FORM 1 [24-09-2020(online)].pdf | 2020-09-24 |
| 2 | 202041041569-DRAWINGS [24-09-2020(online)].pdf | 2020-09-24 |
| 2 | 202041041569-ENDORSEMENT BY INVENTORS [05-10-2020(online)].pdf | 2020-10-05 |
| 3 | 202041041569-COMPLETE SPECIFICATION [24-09-2020(online)].pdf | 2020-09-24 |
| 3 | 202041041569-FORM 3 [05-10-2020(online)].pdf | 2020-10-05 |
| 4 | 202041041569-FORM-26 [05-10-2020(online)].pdf | 2020-10-05 |
| 4 | 202041041569-Proof of Right [05-10-2020(online)].pdf | 2020-10-05 |
| 5 | 202041041569-FORM-9 [05-10-2020(online)].pdf | 2020-10-05 |
| 6 | 202041041569-FORM-26 [05-10-2020(online)].pdf | 2020-10-05 |
| 6 | 202041041569-Proof of Right [05-10-2020(online)].pdf | 2020-10-05 |
| 7 | 202041041569-COMPLETE SPECIFICATION [24-09-2020(online)].pdf | 2020-09-24 |
| 7 | 202041041569-FORM 3 [05-10-2020(online)].pdf | 2020-10-05 |
| 8 | 202041041569-DRAWINGS [24-09-2020(online)].pdf | 2020-09-24 |
| 8 | 202041041569-ENDORSEMENT BY INVENTORS [05-10-2020(online)].pdf | 2020-10-05 |
| 9 | 202041041569-Correspondence_Online Submission_ 03-11-2020.pdf | 2020-11-03 |
| 9 | 202041041569-FORM 1 [24-09-2020(online)].pdf | 2020-09-24 |