Abstract
Fixed-dose combination (FDC) products represent a novel, safe, and cost-effective formulation. Combined use of anticoagulant and antiplatelet medications is common among comorbid cardiovascular patients. This study aimed to formulate FDC tablets for Apixaban and Clopidogrel, as prophylaxis and treatment of thrombo-embolic events. FDC tablets were developed by combining small tablets of Immediate-Release Clopidogrel 75 mg and Extend-Release Apixaban 5 mg through direct compression and wet granulation. Particularly, Apixaban tablets were developed using design expert software, and various types and concentrations of polymers were entered. For Clopidogrel tablets, various diluents were used to develop the formulation. Then, the dissolution profile for each formula was studied. Finally, the optimized formulations were encapsulated within hard gelatin capsules. Apixaban formulation followed zero-order with super case Ⅱ transport mechanism as the dominant mechanism of drug release. The Apixaban drug release rate was affected by the type and concentration of the polymers in the formulation (P < 0.05). As the HPMC concentration was increased, Apixaban release was retarded. For, Clopidogrel, the formulated tablets with spray-dried lactose filler and sodium stearyl fumarate lubricant were found to be stable with good properties. In conclusion, the optimum formulation yielded Clopidogrel and extended-release Apixaban for 24 h with the desired in vitro drug dissolution.
Keywords: Fixed-dose combination tablets, Apixaban, Clopidogrel, Extended-release, Factorial design, In vitro dissolution
1. Introduction
Cardiovascular disease (CVD) is a major cause of morbidity and mortality globally, consequently, the rising cost of health care. The yearly number of new cases increased and the total number of prevalent cases doubled between 1990–2019 (Brodmann et al., 2020). Anti-thrombotic agents are used in single, dual, or triple therapy based on patient diagnosis and risk factors as prophylaxis for cardiovascular events and complications. However, using multi-antithrombotic agents simultaneously increases the risk of bleeding, as the main side effect, and hospitalization. Many reviews were performed to evaluate these regimens and revealed that Clopidogrel (CLOP) is the preferable agent from the P2Y12 receptor antagonist due to its lower adverse medication effect of bleeding. Furthermore, in a clinical trial, dual therapy, including CLOP and Apixaban (APX), was superior to triple therapy with Aspirin or dual therapy with warfarin due to reduced medication-related reversed effects and less hospitalization (Andreou et al., 2018, Barnes, 2020, Bergmark et al., 2022, Lip, 2020, Lupercio et al., 2020, Virginia et al., 2021). These patients have a therapy regimen composed of multi-drug components that can be formulated as fixed-dose combination (FDC) therapy.
Apixaban (Eliquis®) is a direct oral anticoagulant agent, that highly selective active site inhibitor of factor Xa, administered as 5 mg twice daily and reduced to 2.5 mg in certain cases, and its biopharmaceutics classification system (BCS) is class Ⅲ, as good soluble poor permeable substances (Bell et al., 2009, EMC, 2022, Lexicomp, , 2022, Lexicomp, , 2022). The chemical structure of APX is shown in Fig. 1 A (Rxlist, 2022). The Food and Drug Administration (FDA) approved APX to be marketed on December 28, 2012, by Bristol-Myers Squibb/ Pfizer (FDA, 2012). After that, many studies were performed and patents were registered to develop APX in different dosage forms; immediate release (IR) tablets or capsules (Gambhire et al., 2017, Jia et al., 2017, Kumar Shaha and Mehdi Hasan, 2018), IR crushed tablet (Song et al., 2015), dispersible tablets (Vadaliwala et al., 2019), and sublingual film (Shah et al., 2022). In addition, other studies formulated APX for prolonged-release transdermal patches (Patel et al., 2021), transdermal nano-emulsion (Abdulbaqi and Rajab, 2021), and a patent formulated it as extended-release (ER) tablets using only hydroxypropyl methylcellulose (HPMC) (马玉国, 2016). APX as ER tablets is characterized to cause lower gastrointestinal irritation, reduce the bleeding risk, stabilize the drug release, and decrease administration frequencies (马玉国, 2016). Furthermore, clinical studies were performed to assess the blood concentration after administration of an IR tablet of 10 mg once a day and revealed that more fluctuation in blood concentration was observed compared to a dose of 5 mg twice a day. So it is favored to be administered twice a day (Frost et al., 2013, Lassen et al., 2007).
Fig. 1.
Chemical structure of A) Apixaban B) Clopidogrel hydrogen sulfate.
Clopidogrel (Plavix®) is an anti-platelet agent, P2Y12 antagonist, administered as 75 mg once daily, and its BCS is class Ⅱ, as poor soluble good permeable substances (FDA, 1997, Lexicomp, , 2022, Lexicomp, , 2022). Fig. 1 B shows the chemical structure of Clopidogrel (CLOP) Hydrogen Sulfate salt (Merck, n.d.). The FDA approved Sanofi to market CLOP in 1997 (FDA, 1997). Many formulations were developed later in different dosage forms to enhance CLOP solubility and bioavailability. A literature search revealed formulating CLOP as IR tablets (Kavya et al., 2014, Kumar et al., 2013), gastro retentive system as floated tablets (Rao and Lakshmi, 2014), high-density tablets (Desai and Purohit, 2017) or floated osmotic capsules (Shah and Prajapati, 2019), liquisolid compact (Ali et al., 2021, Mohammed and Mohammed, 2018), micro-emulsion (Patel et al., 2010), nano-suspension (Jassim and Hussein, 2014), oral disintegrating tablets (Mahrous et al., 2016), or fast disintegrating films (Alladi Saritha, 2018), and as FDC system (Huang et al., 2011, Seo and Han, 2019).
A single-pill combination, known as FDC, is a system formulated to deliver two or more active pharmaceutical ingredients as one unit. These products are increasing and represent an important segment of the global pharmaceutical market (Kaushal, 2017). The number of FDC approvals significantly increased in the 2000 s and represents an important segment of the global pharmaceutical market (Hao et al., 2015, Kaushal, 2017). Formulation of FD multidrug therapy may give a synergistic and sustained therapeutic effect associated with positive therapeutic outcomes, greater efficacy, safety improvement, and fewer side effects compared to a single maximum dose. Better patient control was also achieved due to fewer frequent and missed doses (Bell, 2013, Kavanagh et al., 2018). Add that patient adherence increased due to less dosing burden and lower cost (Gambhire et al., 2017, Kim and Weon, 2021).
However, these benefits are combined with potential challenges associated with FDC formulations, including drug solubility, dissolution profile, dose, possible drug interaction, and individual drug characteristics. After FDC usage, other challenges may be faced; lack of inclusion in treatment protocol, less dose flexibility to fit individual medications, and the ability to determine the source of side effects (Kavanagh et al., 2018, Kim and Weon, 2021).
Multi-drug therapy can be formulated using different technologies, including monolithic, multilayer, multi-particulate, or 3D-printing systems, depending on the compatibilities between the active constituents and the dissolution profile for each (Kaushal, 2017).
APX and CLOP have different solubility and permeability, so it is better to formulate the FDC as a multi-particulate system.
The present study aimed to develop novel FDC tablets of multi-tablet system and evaluate their in vitro dissolution profile. The FDC consisted of one CLOP (75 mg) tablet and one or two APX (5 mg each) tablets, matrix type, encapsulated within translucent gelatin shell capsules (0 size).
2. Materials and methods
2.1. Materials and reagents
A pharmaceutical grade APX and CLOP hydrogen sulfate salt were received as gifts from Pharmacare PLC (Palestine). Tablet excipients including hydroxypropyl methylcellulose, 28–30 % methoxyl, 7–12 % hydroxypropyl, viscosity (2 % aq. soln., 20 °C) 7500–14000 mPa.s (HPMC) and hydroxypropyl cellulose (HPC) were purchased from Alfa Aesar (by Thermos Fisher Scientific, United Kingdom). Methocel, sodium laurilsulfate (SLS), spray-dried lactose, mannitol, microcrystalline cellulose 112 (MCC 112), colloidal silica, sodium stearyl fumarate (SSF), magnesium stearate (Mg. stearate), and anhydrous ethanol were donated from Pharmacare PLC (Palestine). For buffer preparation, sodium hydroxide pellets, sodium phosphate tribasic dodecahydrate, 98 %, and hydrochloric acid were analytical grades and purchased from Carlo Erba reagents, Acros organics, and Merck Serono respectively. HPLC grade solvents for analysis including; acetonitrile (ACN), trifluoroacetic acid (TFA), and triethylamine (TEA) were purchased from Merck (Merck Serono Amman, Jordan). Water was obtained by filtration using a cellulose nitrate filter (0.45) micron manufactured by Sartorius stedim biotech company (Jordan). Translucent hard gelatin shell capsules (0 size) were donated from Jerusalem Pharmaceuticals Co. Ltd. (Palestine).
2.2. Instruments
APX and CLOP tablets were formulated on a manual single-punch tablet compression machine. Pharma- test® (Germany) instruments were used to perform the friability, hardness, disintegration, and dissolution tests. Copley® tapped density tester, Ohaus® electronic balance, Ohaus® loss on drying (LOD) tester, pH/orp meter, manual sieves, digital caliper, Elma® bath sonicator, and DHG- 9023A dry oven were used during formulation and analysis. Compatibility study experiments were conducted using a Bruker Fourier transform infrared (FT-IR) vacuum spectrometer, equipped with a platinum ATR unit with single reflection diamond crystal (Bruker Optik GmbH, Rosenheim, Germany), and the obtained spectra were compared using OPUS viewer software. Quantitative analysis was performed using an Agilent HPLC 1200 series with an AS thermostat (Santa Clara, USA), and UV/ VIS detector. The used column was BDS Hypersil C18, (4.6*150 mm), 5 μm, thermo-scientific part # 28105–154630. PerkinElmer double beam UV/ VIS spectrometer lambda 25, sonicator, and vacuum filter pump were also used during the analysis. For stability studies, samples were kept in a climate chamber (Binder, Tuttlingen, Germany).
2.3. Compatibility study
A list of expected excipients including polymers, fillers, binders, disintegrants, and lubricants was chosen. A binary mixture of active pharmaceutical ingredients (APIs) and each excipient was prepared in a 1:1 ratio and stored in a stability chamber at room temperature and 40 ± 2 °C/ 75 ± 2 % relative humidity (RH) for four weeks, while other samples were stored in a dry oven at 60 °C for two weeks. These different stress conditions were defined by the international council for harmonization of technical requirements for pharmaceuticals for human use (ICH) guidelines (ICH, 2003). The samples were tested for their physical appearance, and their compatibility was evaluated using FT-IR Spectroscopy. The FT-IR peak matching method spectrum was used for compassion. The samples were scanned in the wavelength range of 4000––400 cm−1.
2.4. Tablets formulation
2.4.1. Preparation of Clopidogrel tablets
The proposed formulas of CLOP tablets were formulated using direct compression (DC) technology. The proposed formulas are listed in Table 1. All ingredients were weighed for 150 tablets per batch for each of the three proposed formulas. All ingredients, except glidant and lubricant, were blended manually for 5 min using a mortar and a pestle. The glidant was then added and blended manually in a polypropylene bag for 2 min, followed by the lubricant, which was added and blended manually in a polypropylene bag for 2 min. Finally, the blends were directly compressed with a manual single-punch tablet machine using 6 mm biconcave punches.
Table 1.
Clopidogrel formulated batches.
| Ingredients | Function | Formulation code (mg/ tablet) |
||
|---|---|---|---|---|
| CLOP 1 | CLOP 2 | CLOP 3 | ||
| Clopidogrel hydrogen sulfate | API | 98 | 98 | 98 |
| Lactose spray dried | Diluent | 42.9 | 0 | 0 |
| Mannitol | Diluent | 0 | 42.9 | 0 |
| MCC 112 | Diluent | 0 | 0 | 42.9 |
| Klucel | Binder and disintegrant | 7.5 | 7.5 | 7.5 |
| Sodium stearyl fumarate | Lubricant | 1.5 | 1.5 | 1.5 |
| Colloidal silica | Glidant | 0.15 | 0.15 | 0.15 |
| Total | 150.05 | 150.05 | 150.05 | |
2.4.2. Preparation of Apixaban tablets
2.4.2.1. Formulation development
APX formulation development aimed to obtain stable sustained-release tablets that were released completely within 24 hrs. The experiment included various manufacturing technologies, including direct compression, wet granulation with water, or wet granulation with absolute ethanol. Dissolution experiments were performed on three tablets of each formula to discriminate between formulas and identify the minimum and maximum amounts of excipients required to produce tablets with the intended characteristics. Eleven formulas (ER1- ER11), listed in Table 2, were prepared until we clearly understood the excipients' behavior and the appropriate manufacturing process.
Table 2.
Formulation trials of Apixaban extended-release tablets:
| Ingredient | ER 1 | ER 2 | ER 3 | ER 4 | ER 5 | ER 6 | ER 7 | ER 8 | ER 9 | ER 10 | ER 11 |
|---|---|---|---|---|---|---|---|---|---|---|---|
| Apixaban | 5 | 5 | 5 | 5 | 5 | 5 | 5 | 5 | 5 | 5 | 5 |
| HPMC | 35 | 35 | 35 | 20 | 25 | 50 | 0 | 0 | 20 | 20 | 20 |
| HPMC E5 | 0 | 0 | 0 | 0 | 0 | 0 | 20 | 50 | 30 | 0 | 25 |
| HPC | 35 | 35 | 35 | 30 | 45 | 0 | 30 | 0 | 0 | 0 | 25 |
| Avicel 101 | 0 | 0 | 0 | 20 | 0 | 20 | 20 | 20 | 20 | 50 | 0 |
| SLS | 0.5 | 0.5 | 0.5 | 0.5 | 0.5 | 0.5 | 0.5 | 0.5 | 0.5 | 0.5 | 0.5 |
| Mg stearate | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 |
| Total | 75.75 mg | ||||||||||
| Manufacturing process | D.C* | WG. DW** | WG. Et-OH*** | ||||||||
D.C.: direct compression.
WG. DW: wet granulation with distilled water.
WG. Et-OH: wet granulation with absolute ethanol.
2.4.2.2. Factorial design
For the determined excipients, a full factorial design (24), D-optimal level, was employed using design expert software version 6.0.4.1 (STAT- EASE), and 4 factors were evaluated. One replicate was run, and then the experimental trials were performed at the sixteen resulting combinations. The concentrations of HPMC (X1), HPC (X2), Methocel E5 (X3), and SLS (X4) were selected as independent variables (Table 3). At the same time, the three dissolution parameters, mean dissolution time (MDT), times 25 and 90 % of the drug released (T25% and T90%), were identified as responses. The data were subjected to 3D response surface methodology to determine the influence of the three polymers and SLS on the dependent variables.
Table 3.
Experimental design plan generated by software:
| Component | Code | Minimum | Maximum | Coded low | Coded High |
|---|---|---|---|---|---|
| A. HPMC | X 1 | 20 | 35 | +0 | +0.495868 |
| B. Methocel E5 | X 2 | 0.25 | 25 | +0 | +0.818182 |
| C. HPC | X 3 | 20 | 35 | +0 | +0.495868 |
| D. SLS | X 4 | 0 | 0.25 | +0 | +0.0082F6446 |
All the formulations contained the same quantities of APX and Mg. stearate (5 mg and 0.025 mg, respectively), but varying quantities of polymers and SLS, and the total tablet weight was 75.75 mg (Table 4). Apixaban ER tablets were formulated by wet granulation technology. All excipients were weighed separately for 150 tablets per batch for each formula. First, excipients, except SLS and Mg stearate, were added in the geometric method. Next, polymers and APX were blended manually for 5 min in a mortar and a pestle, and absolute ethanol was sprayed to formulate granules. The formulated granules passed through mesh size #12, dried in a dry oven at 40 °C for 30 min, and then crossed into mesh size #16. Finally, SLS and lubricant were manually added and blended in a polypropylene bag for 2 min. The final blend was compressed with a manual single-punch tablet machine using 6 mm biconcave punches.
Table 4.
Ingredients of Apixaban ER tablets based on factorial design (APX1- APX16):
| Formula code |
Ingredient (mg/ tab.) |
||||||
|---|---|---|---|---|---|---|---|
|
Apixaban |
HPMC |
Methocel E5 |
HPC |
SLS |
Mg stearate |
Total |
|
| APX 1 | 5 | 35 | 0.5 | 35 | 0 | 0.25 | 75.75 |
| APX 2 | 5 | 27.5 | 7.87 | 35 | 0.12 | 0.25 | 75.74 |
| APX 3 | 5 | 35 | 0.25 | 35 | 0.25 | 0.25 | 75.75 |
| APX 4 | 5 | 20 | 15.5 | 35 | 0 | 0.25 | 75.75 |
| APX 5 | 5 | 20 | 15.25 | 35 | 0.25 | 0.25 | 75.75 |
| APX 6 | 5 | 20 | 25 | 25.37 | 0.12 | 0.25 | 75.74 |
| APX 7 | 5 | 25.5 | 25 | 20 | 0 | 0.25 | 75.75 |
| APX 8 | 5 | 27.05 | 16.15 | 27.05 | 0.25 | 0.25 | 75.75 |
| APX 9 | 5 | 25.25 | 25 | 20 | 0.25 | 0.25 | 75.75 |
| APX 10 | 5 | 35 | 15.37 | 20 | 0.12 | 0.25 | 75.74 |
| APX 11 | 5 | 35 | 7.87 | 27.5 | 0.12 | 0.25 | 75.74 |
| APX 12 | 5 | 30.12 | 20.12 | 20 | 0.25 | 0.25 | 75.74 |
| APX 13 | 5 | 20 | 20.12 | 30.12 | 0.25 | 0.25 | 75.74 |
| APX 14 | 5 | 31.04 | 12.05 | 27.29 | 0.12 | 0.25 | 75.75 |
| APX 15 | 5 | 24.85 | 20.61 | 24.85 | 0.19 | 0.25 | 75.75 |
| APX 16 | 5 | 25.5 | 25 | 20 | 0 | 0.25 | 75.75 |
The final FDC consisted of one CLOP (75 mg) tablet and one or two APX (5 mg each) tablets encapsulated within translucent hard gelatin shell capsules (0 size).
2.5. Evaluation of blends
Various parameters, such as angle of repose and bulk/ tapped density were determined to characterize the final blend flow and compressibility. In addition, Carr's Index (CI%) and Hausner's ratio (HR) were also calculated as per USP General Chapter 〈6 1 6〉 and < 1174> (United States Pharmacopeial, 2011, USPC, 2014).
2.6. Evaluation of FDC tablets
2.6.1. Physical parameters
Ten tablets were randomly selected for each of the 16 formulas of APX and CLOP batch to perform weight variation, thickness, and hardness test. The friability test of IR CLOP and ER APX formulated tablets were evaluated using the pharm-test friabilator. The disintegration times of the IR CLOP formulated tablets were evaluated using a pharm-test disintegrator that operated at 37 ± 2 °C. Six tablets of CLOP were placed into the six cells of the rack, one per cell, and then immersed in water. For all evaluations, the means and standard deviations were calculated.
2.6.2. Assay
Ten tablets of each formula were powdered in a mortar and a pestle. The weight of one tablet (75.75 mg and 150.05 mg of APX and CLOP, respectively) was dissolved in 100 ml ACN, leftover a night to ensure complete dissolution, 5 ml of solution was filtered in a 50 ml volumetric flask through a 0.45 μm syringe filter, and the volume was made up using purified water. The drug content assay was evaluated by chromatographic separation of the APIs on a BDS Hypersil C18, (4.6*150 mm, 5 μm), with ACN and TFA in the ratio 48:52 (v/v) as a mobile phase, at a flow rate of 0.9 ml/ min., column temperature 45 °C, and injection volume of 5 μL. The calibration curve was developed for each API and the correlation coefficient R2 was more than 0.999 (Al-Shami et al., 2024).
2.6.3. In vitro drug release study
2.6.3.1. In vitro dissolution studies
For CLOP, in vitro drug release studies were carried out in 750 ml of 0.1 N HCL for 2 hrs. For APX, in vitro, drug release studies were carried out in 900 ml of 0.05 M Sodium phosphate buffer with 0.05 % SLS for 24 hrs. A USP type II dissolution apparatus (paddle type) was used to test both at 75 rpm and 37 ± 0.5 °C. All dissolution studies were performed on three tablets of each formula.
The in vitro drug release study for the final FDC, two strengths, was carried out using the USP delayed release method A (United States Pharmacopeial Convention, 2011). The study was performed in 750 ml of 0.1 N HCL. Samples were withdrawn at (5, 10, 20, 30, 45, 60, and 120 min.). After 2 hrs., 250 ml of 0.2 M tribasic sodium phosphate equilibrate 37 ± 0.5 °C was added to the dissolution media, and the pH of the media was adjusted to (6.8 ± 0.05). The apparatus continued to run for 24 hrs. A sample of 5 ml was withdrawn from the dissolution media in a specified period (4, 8, 12, 16, 20, and 24 hrs.). All of the experiments were tested using a USP type II dissolution apparatus (paddle type) at 75 rpm and 37 ± 0.5 °C. All dissolution studies were performed on six tablets of each formula.
A sample of 5 ml was withdrawn from the dissolution media using an auto-sampler, and no volume correction was made. The samples were filtered using a 0.45 μm Clarify® syringe filter. The absorbance of the samples was measured using a spectrophotometric method at 210 nm using HPLC developed method(Al-Shami et al., 2024) and the % cumulative release (% CR) was plotted using calculated mean values of cumulative drug release versus time.
2.6.3.2. Kinetic analysis of dissolution data
The release data were fitted to five kinetic models including; zero-order, first-order, Higuchi, Hixon-Crowell, and Korsmeyer-Peppas to determine the drug release mechanism with the aid of DD Solver add-in software. The drug release mechanism was considered according to the coefficient of determination; R2 values (Grassi and Grassi, 2014).
2.7. Swelling and erosion studies
The swelling nature of tablets was studied by water gain on 3 tablets. The swelling index study was performed using USP dissolution apparatus-Ⅱ in the final dissolution media at 37 ± 0.5 °C, rotated at 75 rpm. At predetermined intervals for 24 hrs., the tablets were withdrawn using a predetermined weight mesh, and excess dissolution media was removed with absorbent tissue, and then weighed. The percentage swelling of the tablet was determined according to the following equation (Gerogiannis et al., 1993):
where Wt is the mass in the swollen state at time t, and W0 is the initial tablet weight.
The matrix erosion was determined on the same tablets, at the same time intervals. After weighing the hydrated tablets, they were dried in an oven at 60 °C for 24 hrs., and the remaining dried were weighed. The percentage erosion (% mass loss) was determined according to the following equation (Ravi et al., 2007):
where Wo was the initial tablet weight, Wr was the dry weight after time t.
The remaining percentage was determined using the following equation:
2.8. Stability study
Forty capsules of the optimized batch were packed in a screw-capped amber glass bottle and kept for accelerated stability study at 40 °C/ 75 % R.H. in a climate chamber (BINDER, Tuttlingen, Germany) for 90 days. Accelerated stability study samples were analyzed at 0 and 90 days for physical appearance and drug content concerning the initial results of the same batch.
2.9. Statistical analysis
To investigate the significance of the differences between the results from the studied formulations, a one-way analysis of variance (ANOVA) test was used. The significance level was set at (α < 0.05). Design Expert software version 6.0.4.1 (STAT- EASE) was used for analysis.
3. Results
3.1. Compatibility study
The FT-IR spectra were recorded for pure API and powder mixtures of API and each excipient to assess any possible chemical interactions between API and the excipients (Dave et al., 2015). Table 5 reflects the FT-IR peak values (cm−1) for each functional group. Fig. 2 demonstrates the FT-IR spectra of pure APX, CLOP, and their physical mixture upon preparation and after exposure to stress conditions, Fig. 3, Fig. 4 show each API with the physical mixture with each excipient. No physical changes were observed, and the obtained spectra for pure APIs correlated well with that of the APIs mixture and with excipients and showed all the characteristic peaks with no major changes. Hence, no significant chemical interaction occurs in the solid state at zero time. In addition, no shifting was observed for peaks of each mixture at zero time and after exposure to stress conditions. This finding concludes that the APX and CLOP are compatible with each other, and each API was compatible with its formulation components.
Table 5.
FT-IR data of Apixaban and Clopidogrel hydrogen sulfate.
| API | Functional groups | IR values (cm−1) |
|---|---|---|
| Apixaban | N–H stretch | 3482 |
| N–H stretch | 3308 | |
| C–H stretch | 2902 | |
| C = O stretch | 1679 | |
| Clopidogrel hydrogen sulfate | N–H stretch | 2501 |
| C-S-C stretch | 2345 | |
| C = O stretch | 1751 | |
| C = C stretch | 1438 | |
| C-O stretch | 1062, 1152, 1185 |
Fig. 2.
FT-IR spectra of pure APIs and their physical mixture in different conditions.
Fig. 3.
FT-IR spectra of pure APX and a physical mixture of APX with each excipient. APX.: Apixaban, HPMC: Hydroxypropyl methylcellulose, HPC.: Hydroxypropyl cellulose, SLS.: Sodium lauryl sulfate, Mg. ST.: Magnesium Stearate.
Fig. 4.
FT-IR spectra of pure CLOP and a physical mixture of CLOP with each excipient. CLOP: Clopidogrel Hydrogen sulfate, Lac. S.D.: spray dried lactose, Aerosil: Colloidal silica, SSF.: Sodium stearyl fumarate.
3.2. Formulation development
Based on the excipients compatibility studies and revising the reference listed drug (RLD) product, different laboratory scale experiments were prepared and tested to obtain the appropriate dissolution profile. The experiment included various manufacturing technologies, including direct compression, wet granulation with water, or absolute ethanol.
3.2.1. Development of IR CLOP tablets
Three formulations of CLOP (CLOP 1- CLOP 3) were formulated using different diluents including mannitol, MCC 112, and spray dried lactose. Observation of the characteristics of the powder bed showed that the formula with mannitol (CLOP 2) tended to stick with punches during the compression process. Further, the formula with MCC 112 (CLOP 3) showed a longer disintegration time (within 10 min) than CLOP 1 and CLOP 2 disintegrated within 4 and 7 min, respectively. The dissolution test results revealed that CLOP 1 and 2 were released completely (% CR > 96 %) within the first 30 min of the test, while 76 % of CLOP 3 was released within the first 30 min. Compared to mannitol and lactose, the formula with spray dried lactose (CLOP 1) was chosen as an optimum formula to scale up.
3.2.1.1. Development of ER APX tablets
As preliminary (scanning) experiments, eleven APX formulas were investigated (ER 1 − ER 11), with each formula containing different quantities and types of polymers, as well as different manufacturing technologies. Table 2 shows the tested formulas' composition and their manufacturing process.
The pre-formulation results revealed that the wet granulation technology with absolute ethanol that was sprayed on the powder bed and mixed was the optimum method of formulation. Subsequently, the quantities of polymers that give formulas with extended release within 24 hrs., were determined to be within the range of 20 to 35 mg/ tablet for HPMC and HPC, and less than 25 mg/ tablet for Methocel E5. Furthermore, several Apixaban ER formulas (APX 1 – APX 16) were developed and evaluated utilizing these findings and a factorial approach. Table 4 shows the sixteen developed formulas using design expert software.
3.3. Evaluation of blend
Table 6 presents the findings of various evaluations of the blend characteristics investigation for formulation APX 1–16. The results of bulk densities for all formulations of APX were in the range of 0.19–0.35 g/ ml and 0.33 g/ ml for CLOP. The findings of tapped density for APX formulations were in the range of 0.24–0.40 g/ ml and 0.38 g/ ml for CLOP. The angle of repose values were between 29° − 41° for APX and 34° for CLOP. Carr’s Index and Hausner’s ratio were between 7.1–17.6 % and 1.0–1.2, respectively for APX, and 13.3 % and 1.15 for CLOP. These results revealed excellent to good flow for all the formulas of APX except formulas APX 8, which showed fair flow and APX 4 which showed passable flow. These results indicated that all blends have acceptable flow properties and compressibility, and all pre-compression parameters were within the acceptable ranges.
Table 6.
Evaluation of blend characteristics (n = 3).
| Formulation code | Bulk Density | Tapped density | Angle of repose ⊖ | Flow Property | Carr’s index | Hausner’s ratio | Scale of flowability |
|---|---|---|---|---|---|---|---|
| APX 1 | 0.31 | 0.333846 | 32 | Good | 7.142857 | 1.076923 | Excellent |
| APX 2 | 0.250915 | 0.294825 | 35 | Good | 14.89362 | 1.175 | Good |
| APX 3 | 0.246926 | 0.272122 | 35 | Good | 9.259259 | 1.102041 | Excellent |
| APX 4 | 0.282214 | 0.348618 | 41 | Passable | 19.04762 | 1.235294 | Fair |
| APX 5 | 0.278918 | 0.310614 | 35 | Good | 10.20408 | 1.113636 | Excellent |
| APX 6 | 0.305957 | 0.343268 | 34 | Good | 10.86957 | 1.121951 | Good |
| APX 7 | 0.357852 | 0.402583 | 32 | Good | 11.11111 | 1.125 | Good |
| APX 8 | 0.297844 | 0.339316 | 39 | Fair | 12.22222 | 1.139241 | Good |
| APX 9 | 0.302717 | 0.343827 | 31 | Good | 11.95652 | 1.135802 | Good |
| APX 10 | 0.300867 | 0.347154 | 29 | Excellent | 13.33333 | 1.153846 | Good |
| APX 11 | 0.2462 | 0.284077 | 32 | Good | 13.33333 | 1.153846 | Good |
| APX 12 | 0.335605 | 0.390027 | 31 | Good | 13.95349 | 1.162162 | Good |
| APX 13 | 0.310667 | 0.362444 | 35 | Good | 14.28571 | 1.166667 | Good |
| APX 14 | 0.198118 | 0.240571 | 30 | Excellent | 17.64706 | 1.214286 | Fair |
| APX 15 | 0.3032 | 0.36384 | 30 | Excellent | 16.66667 | 1.2 | Fair |
| APX 16 | 0.331286 | 0.3865 | 31 | Good | 14.28571 | 1.166667 | Good |
| CLOP 1 | 0.3267 | 0.3769 | 34 | Good | 13.3333 | 1.1538 | Good |
3.4. Evaluation of tablets
The evaluation of the resulting tablets of the sixteen APX formulations and CLOP formulation is presented in Table 7. Among APX and CLOP batches the results revealed that all batches showed a total weight loss of less than 1 % after the friability test, had a uniform tablet thickness, whereas the hardness of tablets was variate in the range of 4.13–5.7 kilopond (kp) for APX formulations. The hardness and thickness values showed sufficient mechanical resistance in all the patches. Furthermore, a disintegration test was performed on IR CLOP 1 tablets, and the disintegration time range was between 3–4 min, within the accepted limits. For the assay test, APX tablets assay were between 95–102 %, which indicates getting accuracy in dosing. All evaluation parameter values were within acceptable limits according to the USP 38-NF 33 and European Pharmacopoeia.
Table 7.
Evaluation of formulation batches of tablets.
| Formula code | Weight variation | Thickness | Hardness | Friability (%loss) | Moisture content (%LOD) | Assay (%) |
|---|---|---|---|---|---|---|
| APX 1 | 76.09 ± 2.9 | 2.99 ± 0.07 | 5.69 ± 0.49 | 0.06 | 4.68 | 95.33 |
| APX 2 | 75.5 ± 2.4 | 2.99 ± 0.07 | 5.3 ± 0.416 | 0.097 | 3.95 | 95.72 |
| APX 3 | 75.8 ± 2.37 | 2.97 ± 0.07 | 5.03 ± 0.37 | 0.04 | 4.12 | 97.04 |
| APX 4 | 75.18 ± 2.11 | 2.92 ± 0.06 | 4.75 ± 0.26 | 0.6 | 4.26 | 96.1 |
| APX 5 | 75.04 ± 2.15 | 2.89 ± 0.06 | 4.82 ± 0.43 | 0.2 | 3.99 | 99.36 |
| APX 6 | 75.81 ± 2.19 | 2.92 ± 0.07 | 4.76 ± 0.43 | 0.4 | 3.84 | 100.89 |
| APX 7 | 75.31 ± 2.12 | 2.92 ± 0.07 | 4.22 ± 0.3 | 0.4 | 3.51 | 101 |
| APX 8 | 74.47 ± 2.08 | 2.9 ± 0.05 | 4.13 ± 0.5 | 0.02 | 3.9 | 98.22 |
| APX 9 | 75.38 ± 1.72 | 2.9 ± 0.08 | 4.92 ± 0.15 | 0.22 | 3.99 | 99.64 |
| APX 10 | 74.49 ± 2.4 | 2.89 ± 0.08 | 4.77 ± 0.31 | 0.14 | 4.05 | 101.76 |
| APX 11 | 75.48 ± 2.26 | 2.92 ± 0.06 | 5.38 ± 0.85 | 0.04 | 4.43 | 95.77 |
| APX 12 | 76.49 ± 1.86 | 2.95 ± 0.07 | 4.48 ± 0.31 | 0.16 | 3.85 | 98.76 |
| APX 13 | 76.52 ± 2.19 | 2.9 ± 0.08 | 4.89 ± 0.45 | 0.18 | 3.39 | 96.21 |
| APX 14 | 75.69 ± 3.13 | 2.91 ± 0.09 | 5.29 ± 0.47 | 0.03 | 4.64 | 95.15 |
| APX 15 | 75.51 ± 2.64 | 2.92 ± 0.07 | 4.42 ± 0.36 | 0.04 | 4.65 | 95.08 |
| APX 16 | 76.86 ± 2.27 | 3.02 ± 0.04 | 4.65 ± 0.42 | 0.09 | 4.12 | 102.23 |
| CLOP 1 | 148.9 ± 3.56 | 4.36 ± 0.08 | 6.63 ± 0.61 | 0.65 | 2.9 | 99.8 |
The upper limit is 83.3 mg for APX and 161.3 for CLOP.
The lower limit is 68.2 mg for APX and 138.8 for CLOP.
3.5. Experimental design and response surface analysis
A full factorial design 24 was selected as it helps in understanding the effects of polymers and SLS concentrations on response parameters. Based on the preliminary studies, the quantities of polymers were determined. Changes on the concentration of the three polymers (HPMC, HPC, Methocel E5) and SLS quantity were spontaneously applied, since a single response optimization is thought to yield misleading results (Anderson et al., 2017).
The in vitro dissolution parameters and APX release profiles of the sixteen formulations (APX 1–16) are presented in Fig. 5 and Table 8. APX release was affected by the polymer type and amount used in each formula. The cumulative percent released after 24 hrs. of each of the APX formulations was complete except for APX 10, that only 80 % of the APX were released. The results illustrated that there is a relationship between the concentration of each polymer and its dissolution profile. Formulation with HPMC concentrations between (39.6–46.2 %) APX 1, APX 3, APX 10, APX 11, APX 12, and APX 14 showed the lowest cumulative amount of APX release within 20 h. (91.5, 89.7, 68.4, 84.0, 75.8, and 87.2 %, respectively). When the HPMC concentrations were between 32.8 – 33.7 % (APX 2, APX 7, APX 8, APX 9, and APX 15), a complete release was achieved after 20 hrs. of dissolution. While formulas with HPMC concentrations of 26.4 % showed complete release after 16 hrs., that obviously revealed a strong dependence of percent released (selected as a response) on the concentration of polymers. SLS is added to the formula as a wetting agent for the inherent hydrophobicity of APX (Jia et al., 2017). As the added quantity of API is 5 mg, the sink conditions were achieved and the SLS quantity showed no effect on the drug release.
Fig. 5.
In vitro dissolution of APX. formulations; (A) APX 1––4, (B) APX 5––8, (C) APX 9––12, (D) APX. 13–16.
Table 8.
In vitro dissolution response parameters:
| Formula Code | T25% (h) | T50% (h) | T90% (h) | MDT |
|---|---|---|---|---|
| APX 1 | 5.618 | 11.237 | 20.227 | 10.901 |
| APX 2 | 5.6 | 8.94 | 16 | 9.239 |
| APX 3 | 6.785 | 12.293 | 20.498 | 12.047 |
| APX 4 | 4.125 | 8.135 | 14.472 | 8.351 |
| APX 5 | 3.849 | 7.699 | 13.858 | 7.956 |
| APX 6 | 3.345 | 7.153 | 13.594 | 7.378 |
| APX 7 | 3.373 | 8.945 | 18.730 | 9.629 |
| APX 8 | 4.716 | 9.399 | 16.869 | 9.647 |
| APX 9 | 4.201 | 8.851 | 16.557 | 8.765 |
| APX 10 | 5.4 | 13.25 | More than 24 | 10.756 |
| APX 11 | 6.103 | 12.207 | 21.972 | 12.177 |
| APX 12 | 5.951 | 12.519 | 23.521 | 12.177 |
| APX 13 | 3.778 | 7.555 | 13.600 | 6.886 |
| APX 14 | 5.886 | 11.772 | 21.190 | 11.567 |
| APX 15 | 4.680 | 9.361 | 16.894 | 9.599 |
| APX 16 | 4.050 | 8.099 | 14.579 | 8.318 |
Among all the developed formulations, APX 11 which contains HPMC: Methocel E5: HPC in the ratio 46.2: 10.4: 36.3 gave ER for 24 hr. was selected as the optimum formula.
For CLOP 1, complete drug release was achieved within 15 min, indicating that the used excipients did not retard drug release. The CLOP 1 dissolution profile is illustrated in Fig. 6.
Fig. 6.
In vitro dissolution of CLOP.
The dissolution of the final encapsulated FDC after scale up is shown in Fig. 7. The same dissolution results were obtained, as a complete release of the APX 11 was achieved after 24 hrs, and within 15 min of CLOP 1, with slight rapid release in the percent of CDR of APX 10 mg, two tablets of APX 11, compared to 5 mg, one tablet of APX 11.
Fig. 7.
In vitro drug release of the final dosage form of two doses of APX.5 mg and APX. 10 mg.(n = 6).
3.6. Mathematical model analysis
A mathematical model was constructed to quantify the effect of the variables on the response parameters within the experimental design boundaries. Table 9 summarizes the coefficients of model terms. For each response, ANOVA test model results were evaluated and revealed that the sequential p-value was less than 0.05 for each response, and the lack of fit p-values were more than 0.05. The value of R2 was greater than 0.7, the difference between the predicted R2 and R2 was less than 0.2, and the adequate precision values were greater than 8. These values represented a validated design with well fitted responses and insignificant model errors. All responses are shown to be fitted with a linear model.
Table 9.
ANOVA analysis for the selected different responses of APX formulas.
| Response | Model | Sequential p-value | Lack of fit p-value | R2 | Adjusted R2 |
Predicted R2 | Adequate Precision | F- value |
|---|---|---|---|---|---|---|---|---|
| T25% | Linear | <0.0001 | 0.712 | 0.8697 | 0.8372 | 0.7412 | 15.0793 | 26.71 |
| MDT | Linear | 0.0001 | 0.7175 | 0.8165 | 0.7706 | 0.6501 | 10.8904 | 17.79 |
| T90% | Linear | <0.0001 | 0.9236 | 0.8377 | 0.7972 | 0.6737 | 13.1650 | 20.65 |
Evaluating the effect of each component, a significant association was found between the concentration of polymers (HPMC, Methocel E5, and HPC) and the release time of APX. (p < 0.001, p = 0.0003, and p = 0.0006), respectively. An increase in the HPMC concentration was found to retard APX release while increasing Methocel E5 concentration enhances APX release at the beginning, then slowing the drug release. For HPC, increasing its concentration was shown to delay the release of APX at first, then enhancing the release rate later. SLS different quantities showed to have no significant effect on the release process (p = 0.4843). Table 10 and Fig. 8 represent the p-values for the 3 evaluated responses.
Table 10.
Estimated coefficients for responses.
| Component | MDT |
T25% |
T90% |
|||
|---|---|---|---|---|---|---|
| p-value | Co-efficient | p-value | Co-efficient | p-value | Co-efficient | |
| HPMC | <0.0001 | +0.305009 | 0.0001 | +0.147837 | <0.0001 | +0.652761 |
| Methocel E5 | 0.0003 | +0.031030 | <0.0001 | −0.019282 | 0.0018 | +0.08737 |
| HPC | 0.0006 | +0.024017 | 0.0066 | +0.027684 | <0.0001 | −0.058459 |
| SLS | 0.4843 | +1.66680 | 0.0533 | +2.43533 | 0.5933 | +2.59188 |
Fig. 8.
3D response –surface showing the influence of independent variables on responses.
3.7. Drug release kinetic study
The dissolution data of the sixteen formulas were fitted into different kinetic models and the regression coefficient (R2) was close to 1 reflecting the most suitable model for selection (Singhvi and Singh, 2011). Table 11 shows the obtained data. Most of the formulated matrices fitted well into zero order, combined with Korsmeyer-Peppas model. Among formulas having “n” higher than 0.89, the release follows the super case II transport mechanism. While APX formulas 7 and 10 fitted the Hixson-Crowell model, and the “n” value was between 0.45 < n < 0.89, which indicates the non-Fickian diffusion (anomalous diffusion) model.
Table 11.
In vitro release kinetics parameters:
| Formula code | Zero order |
First order |
Higuchi Model |
Hexon Crowell Model |
Korsmeyer-Peppas Model |
||||||
|---|---|---|---|---|---|---|---|---|---|---|---|
| k0 | R2 | K1 | R2 | kH | R2 | kHC | R2 | kKP | R2 | N | |
| APX 1 | 4.450 | 0.9815 | 0.079 | 0.9309 | 17.615‘ | 0.8596 | 0.022 | 0.958 | 4.574 | 0.9792 | 0.991 |
| APX 2 | 5.345 | 0.9778 | 0.094 | 0.9217 | 19.204 | 0.8413 | 0.027 | 0.9502 | 5.059 | 0.9748 | 1.02 |
| APX 3 | 4.315 | 0.9749 | 0.072 | 0.8997 | 16.875 | 0.8185 | 0.021 | 0.931 | 2.997 | 0.9814 | 1.158 |
| APX 4 | 6.202 | 0.9854 | 0.104 | 0.9419 | 19.889 | 0.8512 | 0.030 | 0.9918 | 5.891 | 0.9935 | 1.022 |
| APX 5 | 6.499 | 0.9794 | 0.111 | 0.9173 | 20.743 | 0.8265 | 0.032 | 0.9468 | 5.684 | 0.977 | 1.054 |
| APX 6 | 6.672 | 0.9736 | 0.122 | 0.9457 | 21.632 | 0.8655 | 0.035 | 0.9698 | 8.295 | 0.9725 | 0.915 |
| APX 7 | 4.835 | 0.9701 | 0.099 | 0.9669 | 19.549 | 0.9235 | 0.027 | 0.9868 | 8.779 | 0.9860 | 0.794 |
| APX 8 | 5.337 | 0.9763 | 0.094 | 0.9245 | 19.204 | 0.8438 | 0.027 | 0.9522 | 5.258 | 0.9763 | 1.005 |
| APX 9 | 5.508 | 0.9814 | 0.101 | 0.9356 | 19.940 | 0.8660 | 0.029 | 0.9630 | 6.276 | 0.9796 | 0.952 |
| APX 10 | 3.518 | 0.9864 | 0.056 | 0.9901 | 14.148 | 0.9173 | 0.016 | 0.9963 | 5.511 | 0.9944 | 0.845 |
| APX 11 | 4.098 | 0.986 | 0.067 | 0.9276 | 16.121 | 0.8443 | 0.019 | 0.9591 | 3.241 | 0.9873 | 1.086 |
| APX 12 | 3.892 | 0.9878 | 0.064 | 0.9533 | 15.554 | 0.8933 | 0.018 | 0.9690 | 4.755 | 0.9877 | 0.935 |
| APX 13 | 6.624 | 0.9655 | 0.119 | 0.9313 | 21.372 | 0.8446 | 0.034 | 0.9572 | 7.577 | 0.9609 | 0.947 |
| APX 14 | 4.254 | 0.9886 | 0.073 | 0.9407 | 16.855 | 0.8671 | 0.021 | 0.9635 | 4.353 | 0.9862 | 0.994 |
| APX 15 | 5.341 | 0.9812 | 0.094 | 0.9264 | 19.223 | 0.8482 | 0.027 | 0.9543 | 5.203 | 0.9785 | 1.010 |
| APX 16 | 6.173 | 0.9940 | 0.104 | 0.9436 | 19.841 | 0.8562 | 0.03 | 0.9674 | 5.990 | 0.9930 | 1.014 |
3.8. Swelling and erosion studies
Fig. 9 showed the swelling and erosion plots. The plot revealed that the matrix tablets undergo both swelling and erosion spontaneously. The first two hours showed very rapid water uptake with no erosion and water uptake was the dominant process till twelve hours, followed by matrix erosion as the predominant process. After the first 0.5 hrs., an increase in weight was observed even after drying, which may be due to entrapped water within the matrix that cannot evaporate after drying. These results ascertain that the drug release was ruled out according to zero order, combined with the Korsmeyer-Peppas model.
Fig. 9.
Swelling and erosion study results.
3.9. Stability study
A short-term stability study results for FDC tablets are shown in Table 12. The study was carried out on the optimized formulation, a capsule containing (CLOP 1 and APX 11), for three months at 40 ± 2 °C/ 75 ± 5 % RH. Stability studies have shown no significant changes in the appearance and percent of APX drug content. For CLOP, a slight decrease in the active substance assay was found. So, it was considered that the formulation has good stability.
Table 12.
Stability studies of optimized batch.
| Parameter | Appearance APX./ CLOP. |
Drug content APX./ CLOP. |
|
|---|---|---|---|
| Initial | APX | Good appearance | 95.77 % |
| CLOP | Good appearance | 99.80 % | |
| After 3 months | APX | Good appearance | 95.74 % |
| CLOP | Good appearance | 97.55 % | |
4. Discussion
Drugs excipients compatibility study was conducted to evaluate the compatibility of CLOP with APX and the compatibility of each API with pharmaceutical excipients expected to be used in the formulation process (WHO Expert Committee on Specifications for Pharmaceutical Preparations, 2012, World Health Organization, 2005). The most suitable excipients are chosen based on the literature and RLD product search to be suitable and fulfill the purpose of the study.
Since CLOP is formulated via direct compression, excipients with good flow properties are needed. The study results revealed using mannitol powder enhances the stickiness tendency as Sha et al. study (Sha et al., 2015). Using MCC founded to retard the disintegration and dissolution of the produced tablets due to the higher binding properties and hydrophobicity of that diluent (Chaerunisaa and Sriwidodo, 2019).
Regarding APX, it has low compressibility, low flowability (Gambhire et al., 2017), low dose, and is prepared with hygroscopic polymers; wet granulation technology was chosen. It aids in the preparation of a more stable formula that is less likely to cake or harden, has improved flowability and compression characteristics, decreased weight variation and loss of blended powder quality, enhanced drugs wettability, bioavailability as well as content uniformity (Augsburger and Hoag, 2010, Loyd and Allen, 2018).
Evaluating blend flow revealed accepted values, and that is related to the manufacturing process as the formulation was prepared in the wet granulation method (Augsburger and Hoag, 2010). Add that CLOP 1 formulation showed good flow even the bad flowability of CLOP using spray dried lactose. The spray dried process was known to yield spherical lactose particles with uniform size and flow easily (Vail and Cotts, n.d.), reflecting on the final blend's flowability.
In addition, this study revealed a strong dependence of % released (selected as a response) on the concentration of polymers that increasing the concentration of polymer increases the viscosity of the gel layer and retard the drug release (Salih, 2015, Samie et al., 2018). Increasing HPC level enhances the drug release and indicates the predominance of swelling, which enhances the chance of diffusion over erosion (Dürig et al., 2011). While increasing the quantity of HPMC retarded the release due to the rapid formation of a strong, thick, and turbid gel layer, that resists water diffusion and surface erosion process; and that retard water uptake, drug diffusion, or release (Li et al., 2010, Mohamed et al., 2013). Comparing HPMC and Methocel E5, the polymer with a more hydrophobic methoxy group, HPMC, is less likely to form hydrogen bonding with water, within and between the polymer particles, so less hydration of the core occurs, and slower drug release is achieved compared to Methocel® (Chang et al., 2020, Li et al., 2010) and that explains the slight increase in the amount of methocel increased the percent of drug released.
Moreover, comparing the dissolution of the 2 doses of APX, a slight rapid release in the percent of CDR of APX 10 mg, two tablets of APX 11, compared to 5 mg, one tablet of APX 11. The total amount of SLS in the dissolution media of 10 mg dose tablets is doubled among the same volume of dissolution media compared to 5 mg. SLS is a surfactant that enhances the solubility of APX, which may be the cause behind the higher percentage of cumulative drug release for the dose of 10 mg compared to 5 mg (Sheskey et al., 2017).
Release kinetics results showed that most of the formulated matrices fitted well into zero order, combined with Korsmeyer-Peppas model. As APX has low water solubility and is formulated into a hydrophilic matrix, it is expected to follow zero order kinetics (Singhvi and Singh, 2011). These results indicate that both diffusion of the drug from the swelling gel layer and erosion of this layer are the drug release mechanisms. Reviewing the behavior of hydrophilic polymers such as HPMC and HPC with low water soluble API revealed that the hydrophilic polymers swell upon hydration, and then the drug dissolved and diffused out the system, then the matrix dissolved which explains the erosion process for drug release (Debotton and Dahan, 2017, Siepmann et al., 2002).
Analyzing the erosion study results, the first two hours showed very rapid water uptake with no erosions, which related to water diffusion into the system, relaxation of the polymer chain, and volume expansion upon exposure to biological fluid (Siepmann et al., 2002).
For CLOP, many studies performed to establish the compatibility between CLOP and different lubricants. Mg. stearate was found incompatible, and another alternative was found to have a less degradative effect such as stearic acid and SSF (Amer et al., 2016; Bharate et al., 2010; Sherman, 2005). This study found a slight decrease in the active substance assay in the formula due to SSF. This result was consistent with Sherman’s study results, where around 1 % of CLOP degraded after applying stress conditions. He revealed that SSF is a more efficient lubricant than castor oil and PEG, and has a lower degradative effect than Mg. stearate, calcium stearate, zinc stearate, and stearic acid (Sherman, 2005). So, it was considered that the formulation has good stability, and a long-term stability study needed to be performed.
5. Conclusion
Oral dosage forms are the most popular route of administration. The FDC tablet offers safety and efficacy advantages by improving patient medication adherence, decreasing polypharmacy, and reducing medication costs. In addition, MPS for FDC products is a valuable tool for preparing medications with different dissolution profiles, pharmacokinetics, and pharmacodynamics. APX and CLOP are antithrombotic substances indicated for patients diagnosed with AF who had ACS and undergone percutaneous coronary intervention. The formulation of a novel FDC tablet of CLOP and APX will improve patient compliance and facilitate the dosing regimen.
The results revealed that CLOP 1 prepared using spray dried lactose as diluent gives a formula with good characteristics and a fast disintegrated effect, and sodium stearyl fumarate showed good compatibility with CLOP. For APX ER formula, the type and quantity of used polymers are shown to be important factors that can affect the drug release from the matrix. APX 11 which contains HPMC: Methocel E5: HPC in the ratios 46.2: 10.4: 36.3 gave ER for 24 hrs. was selected as the best formula. The drug release kinetics follows Korsmeyer-Peppas combined with zero order, and the mechanism was found to be super case Ⅱ transport. The stability studies indicate that the selected formula was stable.
Further study may be implemented to evaluate the dissolution kinetics of FDC in simulated gastric and intestinal media mimicking fasting and fed conditions and to perform a bioequivalence study. A full stability study design following ICH guidelines is also recommended.
Funding.
This research was funded by the Scientific Research Committee, Faculty of Graduate Studies, Birzeit University, Palestine.
Ethical Consideration
Ethics approval was not sought for this study; experimental in-vitro research was conducted in the lab without involvement of any research subjects.
CRediT authorship contribution statement
Ni'meh Al-Shami: Writing – original draft, Writing – review & editing. Hani Naseef: Supervision. Feras Kanaze: Project administration, Supervision, Validation, Visualization.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Acknowledgments
This paper is part of master’s thesis in Industrial Pharmaceutical Technology at Birzeit University.
The authors are thankful to Assem Mubarak for analyzing compatibility study samples on FTIR. Thankful to Ramzi Moqadi for performing the HPLC analysis.
The authors are thankful to Pharmacare PLC (Palestine) for providing a gift sample of Apixaban, Clopidogrel Hydrogen Sulfate, and excipients.
Contributor Information
Ni'meh Al-Shami, Email: nalshami@birzeit.edu.
Hani Naseef, Email: hshtaya@birzeit.edu.
Feras Kanaze, Email: fqanaze@birzeit.edu.
References
- Abdulbaqi M.R., Rajab N.A. Preparation, characterization and ex vivo permeability study of transdermal apixaban O/W nanoemulsion based gel. Iraqi. J. Pharm. Sci. 2021;29:214–222. [Google Scholar]
- Ali B., Khan A., Alyami H.S., Ullah M., Wahab A., Badshah M., Naz A. Evaluation of the effect of carrier material on modification of release characteristics of poor water soluble drug from liquisolid compacts. PLoS One. 2021;16:1–18. doi: 10.1371/journal.pone.0249075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al-Shami N., Naseef H., Moqadi R., Kanaze F. HPLC method development and validation for the determination of apixaban and clopidogrel in novel fixed-dose combination tablets. J. Chem. 2024;2024 doi: 10.1155/2024/2675736. [DOI] [Google Scholar]
- Amer M., Saeed Z., Ur S., Nazir R., Nadeem M., Sultana M., Campus I., Campus I., Ur S., Nazir R. Effect of lubricants on the stability of clopidogrel bisulphate tablets. INDO Am. J. Pharm. Sci. 2016;3:1482–1487. [Google Scholar]
- Anderson M.J., Whitcomb P.J., Kraber S.L., Adams W. Stat-ease handbook for experimenters. Stat-Ease, Inc. 2017;11:1–5. [Google Scholar]
- Andreou I., Briasoulis A., Pappas C., Ikonomidis I., Alexopoulos D. Ticagrelor versus clopidogrel as part of dual or triple antithrombotic therapy: a systematic review and meta-analysis. Cardiovasc. Drugs Ther. 2018;32:287–294. doi: 10.1007/s10557-018-6795-9. [DOI] [PubMed] [Google Scholar]
- Augsburger L., Hoag S.W. Pharmaceutical Dosage Forms: Tablets. Informa Healthcare; New York: 2010. pp. 261–301. [Google Scholar]
- Barnes G.D. Combining antiplatelet and anticoagulant therapy in cardiovascular disease. Hematol. (united States) 2020;20:642–648. doi: 10.1182/HEMATOLOGY.2020000151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bell J.S., O’Reilly C.L., Chen T.F. Use of consumer educators in mental health pharmacy education. Am. J. Pharm. Educ. 2009;73:2–4. [PMC free article] [PubMed] [Google Scholar]
- Bell, D.S.H., 2013. of Fixed-Dose Combination Therapy. Diabetes, Obes. Metab. 15, 291–300. [DOI] [PubMed]
- Bergmark B.A., Mathenge N., Merlini P.A., Lawrence-Wright M.B., Giugliano R.P. Acute coronary syndromes. Lancet. 2022;399:1347–1358. doi: 10.1016/S0140-6736(21)02391-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bharate S.S., Bharate S.B., Bajaj A.N. Interactions and incompatibilities of pharmaceutical excipients with active pharmaceutical ingredients: a comprehensive review. J. Excipients Food Chem. 2010;1:3–26. [Google Scholar]
- Brodmann, M., Cahill, T.J., Hil, D.P., Carapetis, J., Catapano, A.L., Chugh, S.S., Cooper, L.T., Coresh, J., 2020. Global Burden of Cardiovascular Diseases 76. 10.1016/j.jacc.2020.11.010.
- Caldwell WB., Kaushal AM. Multiparticulate technologies for fixed-dose combinations. Springer, New York. 2017 doi: 10.1007/978-1-4939-7012-4_4. [DOI] [Google Scholar]
- A. Y. Chaerunisaa, S., Sriwidodo, M., A., 2019. Pharmaceutical Formulation Design, in: U., Ahmad, J., A. (Ed.),. IntechOpen, London. 10.5772/intechopen.88092.
- Chang Y., Hu C., Yang R., He D., Wang X., Ning B., Sun H., Xiong Y., Tu J., Sun C. A Raman imaging-based technique to assess HPMC substituent contents and their effects on the drug release of commercial extended-release tablets. Carbohydr. Polym. 2020;244 doi: 10.1016/j.carbpol.2020.116460. [DOI] [PubMed] [Google Scholar]
- Dave V.S., Haware R.V., Sangave N.A., Sayles M., Popielarczyk M. Drug-excipient compatibility studies in formulation development : current trends and techniques. Am. Assoc. Pharm. Sci. 2015;1–2015:9–15. [Google Scholar]
- Debotton N., Dahan A. Applications of polymers as pharmaceutical excipients in solid oral dosage forms. Med. Res. Rev. 2017;37:52–97. doi: 10.1002/med.21403. [DOI] [PubMed] [Google Scholar]
- Desai N., Purohit R. Development of novel high density gastroretentive multiparticulate pulsatile tablet of clopidogrel bisulfate using quality by design approach. AAPS PharmSciTech. 2017;18:3208–3218. doi: 10.1208/s12249-017-0805-2. [DOI] [PubMed] [Google Scholar]
- Dürig T., Lusvardi K.M., Harcum W.W. Hydroxypropylcellulose in modified release matrix systems : polymer molecular weight controls drug release rate and mechanism. Pharmaceutical Technology Report. 2011;1:1–7. [Google Scholar]
- EMC, 2022. Eliquis 5 mg film-coated tablets [WWW Document]. URL https://www.medicines.org.uk/emc/product/2878/smpc#gref (accessed 12.14.22).
- FDA, U.F. and D.A., 1997. US Food and Drug Administration. Center for Drug Evaluation and Research. Application number 20839. Clinical Pharmacology and Biopharmaceutical Review(s). 4, 88–100.
- FDA, U.F. and D.A., 2012. US Food and Drug Administration. Center for Drug Evaluation and Research. Application number 202155. Clinical Pharmacology and Biopharmaceutical Review(s).
- Frost C., Nepal S., Wang J., Schuster A., Byon W., Boyd R.A., Yu Z., Shenker A., Barrett Y.C., Mosqueda-Garcia R., Lacreta F. Safety, pharmacokinetics and pharmacodynamics of multiple oral doses of apixaban, a factor Xa inhibitor, in healthy subjects. Br. J. Clin. Pharmacol. 2013;76:776–786. doi: 10.1111/bcp.12106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gambhire M., Gujar K., Vaidhya A. Formulation, optimization and evaluation of immediate release tablet of apixaban. Int. J. Chemtech Res. 2017;10:424–435. [Google Scholar]
- Gerogiannis V.S., Rekkas D.M., Dallas P.P., Choulis N.H. Floating and swelling characteristics of various excipients used in controlled release technology. Drug Dev. Ind. Pharm. 1993;19:1061–1081. doi: 10.3109/03639049309063001. [DOI] [Google Scholar]
- Grassi M., Grassi G. Application of mathematical modeling in sustained release delivery systems. Expert Opin. Drug Deliv. 2014;11:1299–1321. doi: 10.1517/17425247.2014.924497. [DOI] [PubMed] [Google Scholar]
- Hao J., Rodriguez-Monguio R., Seoane-Vazquez E. Fixed-dose combination drug approvals, patents, and market exclusivities compared to single active ingredient pharmaceuticals. PLoS One. 2015;10:1–13. doi: 10.1371/journal.pone.0140708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang Z., Lozano R., Francis R., Aubry A.F., Steckbeck A., Sciascia D.O. Development of a single in vitro dissolution method for a combination trilayer tablet formulation of clopidogrel and pravastatin. Dissolution Technol. 2011;18:12–19. doi: 10.14227/DT180111P12. [DOI] [Google Scholar]
- ICH, 2003. International conference on harmonization (ICH). Guidance for industry: Q1A(R2) stability testing of new drug substances and products. Ich Harmon. Tripart. Guidel. 4, 24.
- Jassim Z.E., Hussein A.A. Formulation and evaluation of clopidogrel tablet incorporating drug nanoparticles. Int. J. Pharm. Pharm. Sci. 2014;6:838–851. [Google Scholar]
- Jia, J., Mead, B., Us, N.J., 2017. (12) Patent Application Publication (10) Pub. No.: US 2017/0202825 A1.
- Kavanagh O.N., Albadarin A.B., Croker D.M., Healy A.M., Walker G.M. Maximising success in multidrug formulation development: a review. J. Control. Release. 2018;283:1–19. doi: 10.1016/j.jconrel.2018.05.024. [DOI] [PubMed] [Google Scholar]
- Kavya K., Sobhita Rani P., Pavani A., Raja Reddy R. Formulation and evaluation of immediate release tablets of Atazanavir. Int. J. Pharm. Sci. Rev. Res. 2014;24:119–123. [Google Scholar]
- Kim D.-W., Weon K.Y. Pharmaceutical application and development of fixed-dose combination: dosage form review. J. Pharm. Investig. 2021;51:555–570. doi: 10.1007/s40005-021-00543-x. [DOI] [Google Scholar]
- Kumar K.K., Kumar K.S., Satheesh G. Formulation and evaluation of Clopidogrel bisulfate immediate release tablets USP 75MG. Int. J. Pharm. Technol. 2013;5:5481–5504. [Google Scholar]
- Kumar Shaha, M., Mehdi Hasan, M., 2018. Formulation Design, Development, and Optimization Of Apixaban INN Tablet A New Era On Immediate Release Drug Delivery System Cilndipine View project Screening of In vitro Antioxidant,Cytotoxic and Antimicrobial Activities of Belamcanda chinensis Aerial par.
- Lassen M.R., Davidson B.L., Gallus A., Pineo G., Ansell J., Deitchman D. The efficacy and safety of apixaban, an oral, direct factor Xa inhibitor, as thromboprophylaxis in patients following total knee replacement. J. Thromb. Haemost. 2007;5:2368–2375. doi: 10.1111/j.1538-7836.2007.02764.x. [DOI] [PubMed] [Google Scholar]
- Lexicomp, 2022. Apixaban (Lexi-Drugs Multinational) [WWW Document]. Wolters Kluwer Heal. Inc. Riverwoods,. URL https://ssl.birzeit.edu/proxy/1198f1cf/https/online.lexi.com/lco/action/doc/retrieve/docid/multinat_f/4669597?cesid=1bWSR0G0CFo&searchUrl=https%3A%2F%2Fonline.lexi.com%2Flco%2Faction%2Fsearch%3Fq%3DApixaban%26t%3Dname%26acs%3Dfalse%26acq%3DApixaban#fbnlis (accessed 12.20.22a).
- Lexicomp, 2022. Clopidogrel (Lexi-Drugs Multinational) [WWW Document]. Wolters Kluwer Heal. Inc. Riverwoods,. URL https://ssl.birzeit.edu/proxy/1198f1cf/https/online.lexi.com/lco/action/doc/retrieve/docid/multinat_f/4668943?cesid=6xWTvKO3HJv&searchUrl=https%3A%2F%2Fonline.lexi.com%2Flco%2Faction%2Fsearch%3Fq%3DClopidogrel%26t%3Dname%26acs%3Dfalse%26acq%3DClopidogrel# (accessed 12.20.22b).
- Li C.L., Martini L.G., Ford J.L., Roberts M. The use of hypromellose in oral drug delivery. J. Pharm. Pharmacol. 2010;57:533–546. doi: 10.1211/0022357055957. [DOI] [PubMed] [Google Scholar]
- Lip, G., 2020. Acute coronary syndrome: Oral anticoagulation in medically treated patients [WWW Document]. Uptodate. URL https://www.uptodate.com/contents/acute-coronary-syndrome-oral-anticoagulation-in-medically-treated-patients?search=acute coronary syndrome in atrial fibralation management&source=search_result&selectedTitle=1∼150&usage_type=default&display_rank=1.
- Loyd V., Allen J. 11th ed. Wolters Kluwer; 2018. Ansel’s pharmaceutical dosage forms and drug delivery systems. [Google Scholar]
- Lupercio F., Giancaterino S., Villablanca P., Han F.T., Hoffmayer K.S., Ho G., Raissi F., Krummen D., Birgersdotter-green U., Feld G.K., Reeves R.R., Mahmud E., Hsu J.C., Diego S., Jolla L., Hospital H.F. Health Research Alliance. 2020;106:575–583. doi: 10.1136/heartjnl-2019-315963.P2Y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahrous G.M., Kassem M.G., Ibrahim M.A., Auda S.H. Formulation and evaluation of orally disintegrating clopidogrel tablets. Brazilian J. Pharm. Sci. 2016;52:309–317. doi: 10.1590/S1984-82502016000200009. [DOI] [Google Scholar]
- Merck, n.d. Clopidogrel Hydrogen Sulfate [WWW Document]. URL https://www.sigmaaldrich.com/IL/en/product/sial/y0001333 (accessed 1.14.23).
- Mohamed F.A.A., Roberts M., Seton L., Ford J.L., Levina M., Rajabi-Siahboomi A.R. The influence of HPMC concentration on release of theophylline or hydrocortisone from extended release mini-tablets. Drug Dev. Ind. Pharm. 2013;39:1167–1174. doi: 10.3109/03639045.2012.681053. [DOI] [PubMed] [Google Scholar]
- Mohammed A.M., Mohammed E.J. Preparation and in-vitro evaluation of clopidogrel bisulfate liquisolid compact. Iraqi J. Pharm. Sci. 2018;27:135–149. doi: 10.31351/vol27iss2pp135-149. (P-ISSN 1683 - 3597 , E-ISSN 2521 - 3512) [DOI] [Google Scholar]
- Patel V., Kukadiya H., Mashru R., Surti N., Mandal S. Development of microemulsion for solubility enhancement of clopidogrel. Iran. J. Pharm. Res. 2010;9:327–334. [PMC free article] [PubMed] [Google Scholar]
- Patel D.J., Vyas A.M., Rathi S.G., Shah S.K. Formulation and evaluation of transdermal patch of apixaban. Int. J. Pharm. Sci. Rev. Res. 2021;69:57–63. doi: 10.47583/ijpsrr.2021.v69i02.009. [DOI] [Google Scholar]
- Rao Kr.K., Lakshmi Kr. Design, development and evaluation of clopidogrel bisulfate floating tablets. Int. J. Pharm. Investig. 2014;4:19. doi: 10.4103/2230-973x.127736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ravi P.R., Ganga S., Saha R.N. Design and study of lamivudine oral controlled release tablets. AAPS PharmSciTech. 2007;8 doi: 10.1208/pt0804095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rxlist, 2022. Elquis (Apixaban tablets) drug [WWW Document]. URL https://www.rxlist.com/eliquis-drug.htm (accessed 12.15.23).
- Salih O.S. Study the sustain release effect of different polymers used in the formulation of aspirin-rosuvastatin tablets. Int. J. Pharm. Pharm. Sci. 2015;7:166–172. [Google Scholar]
- Samie M., Bashir S., Abbas J., Khan S., Aman N., Jan H., Muhammad N. Design, formulation and in vitro evaluation of sustained-release tablet formulations of levosulpiride. Turkish J. Pharm. Sci. 2018;15:309–318. doi: 10.4274/tjps.29200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saritha A. Design and evaluation of Clopidogrel bisulphate fast disintegrating films. World J. Pharm. Med. Res. 2018;4:201–204. [Google Scholar]
- Seo K.S., Han H.K. Multilayer-coated tablet of clopidogrel and rosuvastatin: preparation and in vitro/in vivo characterization. Pharmaceutics. 2019;11:1–13. doi: 10.3390/pharmaceutics11070313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sha, P., Jin, R., Shi, Y., Shen, X., Sha, P., Jin, R., Shi, Y., Shen, X., 2015. (12) United States Patent 2.
- Shah R.P., Patel A.K., Patel V.M. Characterization and optimization of mouth dissolving film of an anticoagulant drug. Apixaban. 2022;7:190–210. doi: 10.35629/7781-0703190210. [DOI] [Google Scholar]
- Shah H.P., Prajapati S.T. Quality by design based development and optimization of novel gastroretentive floating osmotic capsules of clopidogrel bisulfate. J. Pharm. Investig. 2019;49:295–311. doi: 10.1007/s40005-018-0405-5. [DOI] [Google Scholar]
- Sherman, B., 2005. (12) United States Patent.
- Sheskey P., Cook W., Cable C., editors. Handbook of Pharmaceutical Excipients. 8th ed. Pharmaceutical Press; London: 2017. [Google Scholar]
- Siepmann J., Streubel A., Peppas N.A. Understanding and predicting drug delivery from hydrophilic matrix tablets using the “sequential layer” model. Pharm. Res. 2002;19:306–314. doi: 10.1023/A:1014447102710. [DOI] [PubMed] [Google Scholar]
- Singhvi G., Singh M. Review: in vitro drug release characterization models. Int. J. Pharm. Stud. Res. 2011;2:77–84. [Google Scholar]
- Song Y., Wang X., Perlstein I., Wang J., Badawy S., Frost C., Lacreta F. Relative bioavailability of apixaban solution or crushed tablet formulations administered by mouth or nasogastric tube in healthy subjects. Clin. Ther. 2015;37:1703–1712. doi: 10.1016/j.clinthera.2015.05.497. [DOI] [PubMed] [Google Scholar]
- United States Pharmacopeial Convention, 2011. U.S. Pharmacopeial guidelines Dissolution. <711> Dissolution 1, 1–8.
- United States Pharmacopeial, 2011. USP Powder Flow. “Physical Charact. Food Powders,” Phys. Prop. Foods (M. Peleg E. B. Bagley, eds.), Westport, CT AVI, Inc., 30(60), 293–323.
- USPC, U.S.P.C., 2014. Method II-Measurement in a Volumeter. United States Pharmacopeial Conv. 06, 2014–2016.
- Vadaliwala M.A., Patel A.K., Patel V.M. Development and Evaluation of Orodispersible Tablet of Apixaban. 2019;8:955–974. doi: 10.20959/wjpps20195-13702. [DOI] [Google Scholar]
- Vail, J., Cotts, S., n.d. Powder Rheology of Lactose : Impacts of powder morphology on performance of pharmaceutical excipients 2–5. chrome-extension://efaidnbmnnnibpcajpcglclefindmkaj/https://www.tainstruments.com/pdf/literature/RH123.pdf.
- Virginia W., Osman M., Virginia W., Khan M.U., Virginia W., Khan M.S. HHS Public Access. 2021;172:474–483. doi: 10.7326/M19-3763.Dual. [DOI] [Google Scholar]
- WHO Expert Committee on Specifications for Pharmaceutical Preparations, 2012. Annex 3 Pharmaceutical development of multisource (generic) finished pharmaceutical products – points to consider. Tech. Rep. Ser. 970 Forthy-six, 91–120.
- World Health Organization, 2005. Annex 5 Guidelines for registration of fixed-dose. WHO Tech. Rep. Ser. 94–142.
- 马玉国, 2016. Apixaban sustained-release tablet and preparation method thereof.









