ArticlePDF Available

Improvement of Physicochemical and Solubility of Dipyridamole by Cocrystallization Technology

Authors:
  • Dr. Vishwanath Karad MIT World Peace University, Pune
  • School of Pharmacy Dr. Vishwanath Karad MIT World Peace University Pune.
  • Dr. Vishwanath Karad MIT World Peace University School of Pharmacy

Abstract and Figures

The aim of this study was to develop a pH-independent release formulation of dipyridamole (DP) by the combined use of pH-modifier technology and cocrystal technology tartaric acid (TA) was selected as an appropriate pH-modifier in terms of improving physicochemical properties and dissolution behavior of DP under neutral conditions. Molecular docking method was used to identify the suitable conformer. Upon optimization of the ratio of TA to DP (molar ratio of 1:1, 1:2 and 1:3) was prepared by a solvent assisted griding method. Scanning electron microscopy images revealed that formation of DP-TA co crystals supported by supported by powder X-ray diffraction and differential scanning calorimetry analyses. Spectroscopic analysis suggested that there might be inter-molecular interaction among DP and TA resulting in pH independent dissolution behavior of drug substance. The study confirmed the selection of proper coformer and exhibited enhanced physicochemical, solubility and stability of the Dipyridamole cocrystals. Hence, based upon results it revealed that cocrystallization helps in improving the physicochemical properties of the API. Keywords: Dipyridamole, Coformer, Molecular docking, Radar chart, solvent assisted griding, Cocrystals
Content may be subject to copyright.
Gawade et al Journal of Drug Delivery & Therapeutics. 2021; 11(1-s):43-48
ISSN: 2250-1177 [43] CODEN (USA): JDDTAO
Available online on 15.02.2021 at http://jddtonline.info
Journal of Drug Delivery and Therapeutics
Open Access to Pharmaceutical and Medical Research
© 2011-21, publisher and licensee JDDT, This is an Open Access article which permits
unrestricted non-commercial use(CC By-NC), provided the original work is properly cited
Open Access Full Text Article Research Article
Improvement of Physicochemical and Solubility of Dipyridamole by
Cocrystallization Technology
Gawade Ashwini1*, Kuchekar Ashwin1, Boldhane Sanjay2, Baheti Akshay1
1 School of Pharmacy, Dr. Vishwanath Karad, MIT WPU, Paud Road, Kothrud, Pune 411 038
2 Sr. General Manager -Formulation Development at Micro Labs Ltd., Bangalore 560 001
Article Info:
_________________________________________
Article History:
Received 08 Dec 2020;
Review Completed 21 Jan 2021
Accepted 30 Jan 2021;
Available online 15 Feb 2021
_________________________________________
Cite this article as:
Gawade A, Kuchekar A, Boldhane S, Baheti A,
Improvement of Physicochemical and Solubility of
Dipyridamole by Cocrystallization Technology,
Journal of Drug Delivery and Therapeutics. 2021;
11(1-s):43-48
DOI:http://dx.doi.org/10.22270/jddt.v11i1-s.4696
Abstract
______________________________________________________________________________________________________
The aim of this study was to develop a pH-independent release formulation of dipyridamole
(DP) by the combined use of pH-modifier technology and cocrystal technology tartaric acid
(TA) was selected as an appropriate pH-modifier in terms of improving physicochemical
properties and dissolution behavior of DP under neutral conditions. Molecular docking
method was used to identify the suitable conformer. Upon optimization of the ratio of TA to
DP (molar ratio of 1:1, 1:2 and 1:3) was prepared by a solvent assisted griding method.
Scanning electron microscopy images revealed that formation of DP-TA co crystals supported
by supported by powder X-ray diffraction and differential scanning calorimetry analyses.
Spectroscopic analysis suggested that there might be inter-molecular interaction among DP
and TA resulting in pH independent dissolution behavior of drug substance. The study
confirmed the selection of proper coformer and exhibited enhanced physicochemical,
solubility and stability of the Dipyridamole cocrystals. Hence, based upon results it revealed
that cocrystallization helps in improving the physicochemical properties of the API.
Keywords: Dipyridamole, Coformer, Molecular docking, Radar chart, solvent assisted
griding, Cocrystals
*Address for Correspondence: Dr. Ashwini Gawade, School of Pharmacy, Dr. Vishwanath Karad, MIT WPU, Paud Road, Kothrud, Pune 411 038
INTRODUCTION
Dipyridamole USP is a platelet inhibitor chemically
described as 2,2',2",2"'-[(4,8- Dipiperidinopyrimido[5,4-
d]pyrimidine-2,6-diyl)dinitrilo]-tetraethanol.
Dipyridamole is an odorless yellow crystalline powder,
having a bitter taste. It is soluble in dilute acids, methanol
and chloroform, and practically insoluble in water.
Dipyridamole is BCS class II drug having low solubility and
high permeability. It is soluble at low pH but insoluble in
high pH (i.e., alkaline ph of small intestine ) has a narrow
absorption window and is primarily absorbed in the
stomach, its oral bioavailability is 37 - 66% & biological
half life is also short (40 min). Dipyridamole is highly
bound to plasma proteins. It is metabolized in the liver
where it is conjugated as a glucuronide and excreted with
the bile 1-4.
The low water solubility of the active substance is
responsible for the risks of low oral bioavailability. In
order to enhance the therapeutic efficacy, Dipyridamole
needs an alternative drug delivery system. Cocrystals have
recently drawn significant attention in the delivery of
drugs by enhancing the drug physicochemical properties
such as melting point, solubility, dissolution rate, stability
and bioavailability without changing their chemical
structure 5,6. Cocrystals are compounds with a
stoichiometric ratio of drug substance and cocrystal
coformers (CCFs) (1:1, 1:2, 1:3 or vice versa) 7. These
cocrystals are combined by non-covalent interfaces like
hydrogen bonds, Van Der Waals forces and π-π packaging
which are robust at room temperature 8 , 9, 10, 11. Cocrystal
former is a ballast molecule. Identifying and selecting the
appropriate conformer continues the most important
factor in the effective co-crystal growth 7. This technology
is explored effectively for the delivery of various drugs
such as acyclovir 12, gliclazide 13, piracetam 14, fexofenadine
15, furosemide 16, quercetin 17, baicalein 18, myricetin 19 etc
to improve the therapeutic efficacy.
Figure 1: Structure of Dipyridamole
Gawade et al Journal of Drug Delivery & Therapeutics. 2021; 11(1-s):43-48
ISSN: 2250-1177 [44] CODEN (USA): JDDTAO
The objective of present paper is to enrich the rate of
dissolution, efficacy of Dipyridamole absorption using
solvent assisted griding method. Literature study shows that
there have been few Dipyridamole formulations reported to
date. Dipyridamole was therefore chosen as the poorly
soluble model drug in this work; the solvent assisted
grinding method developed Dipyridamole tartaric acid pH-
independent cocrystals. Fourier transform-infrared
spectroscopy, Differential scanning calorimetry, scanning
electron microscopy and powder X-ray diffraction defined
the cocrystals produced and solubility and dissolution
studies defined the enhancement of solubility and % drug
release.
MATERIALS AND METHODS
Materials:
Dipyridamole was obtained from Micro Advanced Research
Center (Bangalore, India) as a gift sample. Tartaric acid was
collected from Poona Chemical Laboratory. Other reagents
have been bought from S. D. Fine Limited Chemicals
(Mumbai, India).
Methods:
Molecular docking and Selection of coformers
Molecular docking is effective approach for computer aided
structure-based drug discovery. This strategy predicts the
probability of binding and orienting one molecule (API) is
connected to a second molecule (coformer) to form a new
complex 18. The strength of the binding affinity between two
molecules by means of scoring features can be determined 19,
20. Based on the literature, seven coformers were selected for
the preparation of the Dipyridamole cocrystals. The research
of molecular docking was carried out on the seven coformers
chosen. Furthermore, the radar chart was used to visually
compare the quality docking score of coformer information
with an advantage of showing multidimensional information
without the use of statistical methods. Tartaric acid was
verified among the seven coformers based on the potential
for interaction type, compatibility, and docking score for
cocrystals confirmation with Dipyridamole.
Solvent assisted griding method of cocrystallization for
Dipyridamole
Liquid assisted grinding involves the addition of a solvent,
typically in a very small amount, to the dry solids prior to the
initiation of milling. The solvent has a catalytic role in
assisting cocrystal formation and should persist for the
duration of the grinding process. More efficient cocrystal
formation is suggested for liquid assisted methods than with
neat methods.
Dipyridamole cocrystals synthesis was performed using
Liquid assisted grinding technique. Screening of formation of
DYP cocrystals was performed by various coformers in an
optimal molar ratio (1:1, 1:2 and 1:3). A mixture of 1:1
Dipyridamole and tartaric acid (TA) griding was carried out
in mortar and pestle for 30 minutes with the addition of 10
mL ethanol drop wise. And wet crystal was dried in oven and
store in desiccator. Crystals were triturated in mortal and
pestle and stored at room temperature 21, 22.
Figure 2: Preparation of DYP TA Cocrystals by Solvent assisted griding method
Characterization of DEM cocrystals
Differential scanning calorimetry (DSC)
A differential calorimeter scanning (DSC7020 thermal
analysis system HITACHI) was used for thermal analysis of
DYP, DYP cocrystals samples. Powder samples of
approximately 2.0 mg were placed in aluminum open
crucibles and heated at a rate of 10°C/ min up to 400°C.
Fourier transform-infrared spectroscopy (FT -IR)
FT-IR spectra were registered on a Nicolet iS10
spectrophotometer from 4,000 cm-1 to 500 cm-1 (Thermo
Fisher Scientific, Madison, USA). With 40 scans per spectrum
at a resolution of 0.4 cm-1, DYP cocrystals were obtained and
analyzed using the DTGS KBr detector.
Powder X-ray diffraction (PXRD)
DYP, DYP cocrystals XRD patterns were achieved using
Shimadzu XRD-6000X system at ambient temperature
(Shimadzu, Japan). Samples with Ni-filtered Cu-K (α)
radiations were irradiated at a voltage of 40.0 kV and a
current of 40.0 mA. The scanning rate ranged from 3º to 50º
over a diffraction angle of 2º/min.
Invitro dissolution study
The dissolution studies were performed in a dissolution
apparatus Electrolab, Navi Mumbai using the paddle method
in 900 mL of pH 1.2 and 6.8 phosphate buffer at 75 rpm
maintained at 37±0.5°C. The dissolution medium was added
an amount equal to 75 mg of cocrystals and the samples
were withdrawn at appropriate intervals. The samples were
filtered through Whitman filter paper No. 41, diluted, and
spectrophotometrically analyzed at 282 nm.
RESULTS AND DISCUSSION
Dipyridamole structure consists of two aromatic rings
(pyrimido and pyridine), four hydroxyl groups, eight
aromatic nitrogen atoms . Dipyridamole molecule has four
hydrogen bond donors as well as twelve hydrogen bond
acceptors due to aromatic nitrogen (N) in pyrimidine ring,
pyrimidine ring and hydroxy groups and significant
conformational flexibility; hence it is possible to form co-
Gawade et al Journal of Drug Delivery & Therapeutics. 2021; 11(1-s):43-48
ISSN: 2250-1177 [45] CODEN (USA): JDDTAO
crystals with certain co-formers 23. Depending on the ability
of interaction, hydrogen bond, docking score and
compatibility, tartaric acid was selected as coformer for
preparation of Dipyridamole cocrystals (Figure 2). The
details are summarized in the below Table 1. Figure 3
represents visual comparison of docking score in form of
Radar chart. There are several axes in a radar chart where
the information can be plotted. Every axis is one category.
The information is displayed on the axis as points. It is
possible to join the points belonging to a one data series. A
point near the center of an axis shows a reduced value and
vice versa. Through the visual comparison from Radar charts
and docking score from molecular modeling, tartaric acid
showed the lowest score. Van Der Waals and the
electrostatic energy define the interaction between the API
and coformer. Higher docking results in the potential for
repulsion. In contrast, reduced docking score relates to
reduced potential.
Figure 2: Hydrogen bonding of Dipyridamole- tartaric
acid cocrystals
Figure 3: Application of radar chart to evaluate the coformers
Table 1: Molecular modeling of Coformers
Sr No.
Name
Structure
Hydrogen bonding
01
Adipic acid
HO
OOH
O
Hydrogen bonding
possible
02
Malic acid
OOHHO O
Hydrogen bonding
possible
03
Succinic acid
HO
O
O
OH
No bonding
04
Nicotinamide
N
NH2
O
No bonding
05
Sodium acetate
No hydrogen bond.
06
Benzoic acid
No hydrogen bond.
07
Tartaric acid
Hydrogen bonding
possible
Solvent assisted gridding using ethanol as a solvent and
tartaric acid as a coformer resulted in adequate
Dipyridamole cocrystal formation. Preliminary cocrystal
formation evaluation was performed by comparing pure
drug and cocrystals.
Gawade et al Journal of Drug Delivery & Therapeutics. 2021; 11(1-s):43-48
ISSN: 2250-1177 [46] CODEN (USA): JDDTAO
Differential scanning calorimetry (DSC)
An endothermic peak at 156°C represented the melting point
of pure drug. Significant difference in the melting point (156
°C) of cocrystals was observed compared to pure drug
(163°C), sharp endothermic peaks appear at lower
temperatures in comparison to the sharp endothermic peaks
at higher temperatures of the individual components in
Figure 4. This could suggest interaction between the
components, and formation of co-crystallization.
Temp Cel 350.0300.0250.0200.0150.0100.0
DSC mW
0.200
0.000
-0.200
-0.400
-0.600
-0.800
-1.000
DDSC mW/min
Temp Cel 300.0250.0200.0150.0100.0
DSC uW
-50.0
-100.0
-150.0
-200.0
-250.0
-300.0
-350.0
-400.0
-450.0
-500.0
-550.0
DDSC uW/min
57.4Cel
124.3Cel
Figure 4: Differential scanning Calorimetry of a) Dipyridamole and b) Dipyridamole cocrystals
Fourier transform-infrared spectroscopy (FT -IR)
The FTIR showed shifts and changes in the intensity of the
peaks in the DYP and DYP co-crystals, as shown in Figure 9.4.
Hydrogen bonding in the co-crystals was identified by
decreasing the intensity of the O-H peak. A decrease in the N-
H stretching and bending frequency indicates that hydrogen
is involved in hydrogen bonding. The degree of decrease in
frequency and the relative broadening of the band can
determine the magnitude of hydrogen bonding. The role of
the degree and strength of the hydrogen bond is to reduce
the frequency. Significant modifications in the area of
covalent bond between C-C and amine (N-H) stretch have
shown the formation of new hydrogen 24, 25.
Figure 5: FTIR graph of a) Dipyridamole and b) Dipyridamole cocrystals
Powder X-ray diffraction (PXRD)
X-ray diffractograms of DYP showed an intense peak at 2 to
20º signifying the crystalline nature of the drug. DYP
cocrystals showed no intense drug peaks were observed at
of 20º indicating existence of the amorphous phase
(Figure 6). In the case of Dipyridamole cocrystals, however,
there were no intensive drug peaks at 2θ of 20º showing the
presence of the amorphous stage. Decreased intensities and
fewer peaks may be due to changes in crystal habit or
amorphous form. Reduced crystalline characteristics may
result in enhanced dissolution of Dipyridamole compared to
pure drug 26.
Figure 6: XRD spectra of a) Dipyridamole and b) Dipyridamole cocrystals
Gawade et al Journal of Drug Delivery & Therapeutics. 2021; 11(1-s):43-48
ISSN: 2250-1177 [47] CODEN (USA): JDDTAO
Invitro drug release
As reported in literature, Dipyridamole solubility depends
on pH. It is highly soluble in acidic media and poorly soluble
in alkaline media. Dipyridamole is also susceptible to acid
and undergoes degradation. Hence, Dipyridamole cocrystals
were developed by improving the physicochemical
properties. The dissolution experiment was performed on
Dipyridamole and Dipyridamole cocrystals at pH 1.2 and pH
6.8 to verify the pH independent solubility and release of
drugs. The Dipyridamole and Dipyridamole cocrystals
dissolution profile is depicted in Figure 8 and 9. The
Dipyridamole dissolution profile shows that API has a good
rate of dissolution in acidic pH (29.03%) as compared to
alkaline pH 48.1%) % at 30 minutes respectively. The
complete quantity of Dipyridamole dissolved in 45 min was
62% in acid pH and 32 % in alkaline pH. However,
Dipyridamole cocrystals dissolution rate led in a substantial
rise as function of pH independent drug release. The
quantity of Dipyridamole released from cocrystals in the
first 30 minutes was 58.58% and the dissolved quantity was
67.5% at pH 1.2. In the first 30 minutes, the amount of
substance dissolved 82.8% drug release in 30 minutes and
96.48 percent drug release at 45 minutes in pH 6.8 (Figure 8
& Figure 9). In addition, higher dissolution of Dipyridamole
cocrystals can be ascribed to changes in the crystalline
pattern, size and shape of cocrystals that escort to increased
cocrystal solubility in dissolution media 16,24, 27, 32- 37.
Figure 8: Invitro dissolution profile for Dipyridamole and DP-TA cocrystals at pH 1.2
Figure 9: Invitro dissolution profile for Dipyridamole and DP-TA cocrystals at pH 6.8
CONCLUSION:
In the present work prepared Dipyridamole cocrystals
exhibited excellent physicochemical properties (solubility
and dissolution) properties when compared to pure drugs.
From the conducted study, we can conclude that cocrystals
with tartaric acid prepared by technique of solvent assisted
gridding techniques showed an improvement in the
solubility and pH independent drug release, rate of
dissolution and stability compared to pure drug.
ACKNOWLEDGEMENT:
The authors are grateful to Dr. B. S. Kuchekar Dean,
Pharmacy School, Dr. Vishwanath Karad MIT World Peace
University, Pune for offering the required equipment for
conducting the experiment.
Conflict of interest: The authors declare no conflict of
interest.
Gawade et al Journal of Drug Delivery & Therapeutics. 2021; 11(1-s):43-48
ISSN: 2250-1177 [48] CODEN (USA): JDDTAO
REFERENCES:
1. EP1894561A1 Al, Dipyridamole pharmaceutical compositions
2. Shankar P, Rajesh N, Prashant P, Hrutuja C, Harsha P, Scientific
coformer screening, preparation and evaluation of Dabigatran
Etexilate Mesylate Cocrystal, Asian Journal of Pharmacy and
Pharmacology 2018; 4(6):810-820
3. Nate S and Ann N., Pharmaceutical Cocrystals and Their
Physicochemical Properties, Crystal Growth & Design, 2009; 9:6
4. Elena IK, Kratochvíl B., Pharmaceutical Cocrystals, Procedia
Chemistry 2014; 10:473 476
5. Emily S and John CW, Non-Covalent Derivatives: Cocrystals and
Eutectics , Molecules 2015; 20:14833-14848
6. Xia LD, Jia MC and Tong BL, Pharmaceutical cocrystallization: an
effective approach to modulate the physicochemical properties
of solid-state drugs, Cryst Eng Comm, 2013; 1-24
7. Manjunath GJ, Pradip KM and Deepak Chopra, Cocrystals: A
Review of Recent Trends in Pharmaceutical and Material Science
Applications, Material Science Research India, 2017; 14(1):09-
18
8. Anna K, Maria M and Kyriakos K, Pharmaceutical Cocrystals:
New Solid Phase Modification Approaches for the Formulation of
APIs, Pharmaceutics 2018; 10( 18).
9. Jignasa KS, Chirag P, Improvement of physicochemical
parameters of acyclovir using cocrystallization approach,
Brazilian Journal of Pharmaceutical Sciences, 2016; 52:4.
10. Reddy MN and Anusha P, Development and characterisation of
gliclazide cocrystals, International Journal of Pharma and Bio
Sciences, 2018; 8(1):22-32.
11. Sonke R et al., Investigation of the Formation Process of Two
Piracetam Cocrystals during Grinding, Pharmaceutics 2011;
3:706-722
12. Mounika P. et al., Preparation and Characterization of Novel Co-
Crystal Forms of Fexofenadine, International Journal of
Innovative Pharmaceutical Re search. 2015; 6(1):458-463
13. Goud NR et al., Novel Furosemide Cocrystals and Selection of
High Solubility Drug Forms, Jour nal of Pharmaceutical Sciences,
2012; 101: 2.
14. Umi A et al., Crystal Engineering Of Quercetin By Liquid assisted
Grinding Method, Jurnal Teknologi (Sciences & Engineering)
2019; 81(1):3945
15. Jiaxin P, A nano cocrystal strategy to improve the dissolution
rate and oral bioavailability of baicalein, Asian Journal of
Pharmaceutical Sciences; 2019; 14:154164.
16. Mingyu L et al., Development of a pharmaceutical cocrystal with
solution crystallization technology: preparation,
characterization, and evaluation of myricetin-proline cocrystals,
European Journal of Pharmaceutics and Biopharmaceutics,
http://dx.doi.org/10.1016/j.ejpb.2016.07.008 (Accepted
manuscript)
17. Lengauer T, Rarey M. Computational methods for biomolecular
docking. Current Opinion in Structural Biology, 1996; 6(3):402-
6.
18. Gaba M, Gaba P, Singh S, Gupta GD. An overview on molecular
docking. International Journal of Drug Development & Research,
2010; 2(2):219-31.
19. Meng XY et al.. Molecular Docking: A powerful approach for
structure-based drug discovery. Current Computer-Aided Drug
Design, 2011; 7(2):14657
20. Maryam KJ et al., Creating Cocrystals: A Review of
Pharmaceutical Cocrystal Preparation Routes and Applications
Cryst. Growth Des .2018; 18:6370−6387.
21. Hilya NI et al., Improvement of Dissolution Properties Through
Acyclovir - Succinic Acid Cocrystal Using Solvent Evaporation
Technique, International Journal of Drug Delivery Technology
2017; 7(4):304-309.
22. Cavan K et al., Application of radar chart array analysis to
visualize effects of formulation variables on IgG1 particle
formation as measured by multiple analytical techniques, J
Pharm Sci. 2013; 102(12).
23. Douglas BK et al., Docking and scoring in virtual screening for
drug discovery: methods and applications, Nature Reviews, Drug
Discovery, 2004; 3:935- 949.
24. Patrycja S, Marek W, DSC as a screening tool for rapid co-crystal
detection in binary mixtures of benzodiazepines with co-
formers, Journal of Thermal Analysis and Calorimetry; 2018;
133:785795
25. Samsodien H et al., FTIR, Dissolution and Anti-viral Activity of
Nevirapine Co-crystals, Pharm Anal Acta 2017; 8:9
26. Dipak DG et al., Cocrystallization of Etodolac: Prediction of
Cocrystallization, Synthesis, Solid State Characterization and In
Vitro Drug Release, Marmara Pharmaceutical Journal 2017;
21:78-88.
27. Setyawan D et al., Preparation and ch aracterization of
Artesunate - Nicotinamide cocrystal by solvent evaporation and
slurry method, Asian J Pharm Clin Res, 2014; 7 (1):62-65.
28. Pinki R et al., Co-Crystals Formation of Clarithromycin with
Urea: An Efficient Approach to Enhance the Solubility and
Dissolution Rate; AJADD . 2016; 4(02):012-020.
29. Krishna et al., 'Preformulation, Formulation Development And
Drug Release Studies Of Dipyridamole Floating Microballoons'
IJPSR, 2020; 11(9):4637-4647.
30. Syed Reshma Sulthana et al., 'Enhancement of solubility and
dissolution rate of poorly water soluble dipyridamole using fr uit
powder' Asian Journal of Pharmacy and Pharmacology 2019;
5(2):272-280
31. Girdhar A. et al., 'Per meability Behavior of Nanocrystalline Solid
Dispersion of Dipyridamole Generated Using NanoCrySP
Technology' Pharmaceutics 2018; 10(160):1-18.
32. Mizoguchi M. et al, 'Development of pH-Independent Drug
Release Formulation Using Lipocalin-Type Prostaglandin D
Synthase' , Journal of Pharmaceutical Sciences, 2016; 105(
9):2735-2742
33. Anilkumar A, Murthy TEGK, Rani AP. Development of ph
Independent Drug Release System for Dipyridamole. Indian J of
Pharmaceutical Education and Research. 2018; 52(3):374-80.
34. Kojo Y.et al 'Improved Dissolution of Dipyridamole with the
Combination of pH-Modifier and Solid Dispersion Technology',
Chem. Pharm. Bull. 2017; 65:426431
35. Gawade AR and Boldhane: Solid dosage form development of
Dabigatran Etexilate Mesylate with increased solubility and
dissolution using co-crystallization. Int J Pharm Sci & Res 2020;
11(5):2899-05.
36. Kuchekar A, B, Pawar A. P: Screening of factors using Plackett
Burman design in the preparation of Capecitabine-loaded nano
polymeric micelles. Int J Pharm Pharm Sci. 2015; 6(5); 489-96.
37. Pawar AP, Munde PL, Bothiraja C, Kuchekar AB, Development of
ranolazine loaded floating biomaterial gellan beads using Box-
Behnken factorial design. Materials Technology, 2012;
201(30):33-42.
... Various co-formers in an ideal molar ratio screened the development of CAM 2HCl-FA co-crystals (1:1, 1:2, and 1:3). A blend of the 1:3 ratio of CAM 2HCl and FA was kneaded in a mortar and pestle for five minutes at high speed as shown in (Figure 2) [15]. Formed co-crystals were stored at accelerated ambient, intermediate, and accelerated stability conditions as per ICH recommendations [16]. ...
... The paddles were initiated at the present rate as soon as the tablets were dropped into the medium (50 rpm). 5 ml of the sample is withheld over the indicated time (5,15,30,45, and 60 min). At 259 nm, those samples are examined utilizing a UV spectrophotometer. ...
... The application of a Radar chart to evaluate the docking score of selected co-formers is represented in (Figure 5). FA showed the maximum amount of hydrogen bonds formed with CAM 2HCl and the lowest Glide score of -5.21 kcal/mol [15]. FA had the lowest docking score when compared visually using Radar charts and molecular modeling. ...
Article
In a recent study, the objective was to select an appropriate co-former and investigate its impact on the formation of co-crystals involving Camylofin dihydrochloride and Fumaric acid. To determine a suitable co-former, a molecular docking study was conducted, and among the compounds evaluated, fumaric acid exhibited the highest number of hydrogen bonds formed with Camylofin dihydrochloride and demonstrated a favorable Glide score of -5.21 kcal/mol. The kneading method was employed after optimizing the molar ratio of Camylofin dihydrochloride to Fumaric acid, which was found to be 1:1, 1:2, and 1:3. The resulting Camylofin dihydrochloride co-crystals underwent various analytical techniques, including Fourier Transform Infrared Spectroscopy, Scanning Electron Microscopy, Powder X-ray Diffraction, and Differential Scanning Calorimetry. The Camylofin dihydrochloride - Fumaric acid co-crystals, immediate-release tablets were formulated. Stability analysis demonstrated that the final batch F5 remained stable under accelerated ambient stability conditions (40°C±2°C, 75% RH±5%RH) and accelerated stability conditions (25°C±2°C). In conclusion, the co-crystal technology utilized in this study successfully improved the stability of Camylofin dihydrochloride without altering its chemical composition. This approach holds significant potential for enhancing the performance and characteristics of pharmaceutical formulations, thereby contributing to the development of more effective and stable medications.
Article
Full-text available
Dipyridamole (DYP) is potent drug that prevents the thromboembolic risk. It has been clinically used for chronic treatment of angina pectoris treatment and during the valve replacement. heart valve replacement and long-term angina pectoris treatment and is well absorbed in the stomach with BCS class II drug and low oral bioavailability. The present research investigation was focused on the formulation of matrix tablets of Dipyridamole cocrystals and the evaluation of In vivo anticoagulant activity. The results of the study showed that the formulated matrix tablets of dipyridamole cocrystals showed improved efficacy in comparison with the plain drug by enhancing the pre-compression parameters such as bulk density, tap density, Carr's index, angle of repose and Hausner's ratio and post-compression parameters like thickness and weight variation, hardness and friability, In vitro dissolution parameters. The improved efficacy was confirmed by improvement in the pharmacodynamic parameters such as cutaneous bleeding time and clotting time indicative of enhanced bioavailability of dipyridamole. Thus, it can be concluded that the dipyridamole matrix tablets prove to be more effective in producing the anticoagulant effect in clinical practice as compared to the plain drug resulting in more patient compliance.
Article
Full-text available
Viruses are the current big enemy of the world’s healthcare systems. As the small infector causes various deadly diseases, from influenza and HIV to COVID-19, the virus continues to evolve from one type to its mutants. Therefore, the development of antivirals demands tremendous attention and resources for drug researchers around the world. Active pharmaceutical ingredients (API) development includes discovering new drug compounds and developing existing ones. However, to innovate a new antiviral takes a very long time to test its safety and effectiveness, from structure modeling to synthesis, and then requires various stages of clinical trials. Meanwhile, developing the existing API can be more efficient because it reduces many development stages. One approach in this effort is to modify the solid structures to improve their physicochemical properties and enhance their activity. This review discusses antiviral multicomponent systems under the research phase and has been marketed. The discussion includes the types of antivirals, their counterpart compound, screening, manufacturing methods, multicomponent systems yielded, characterization methods, physicochemical properties, and their effects on their pharmacological activities. It is hoped that the opportunities and challenges of solid antiviral drug modifications can be drawn in this review as important information for further antiviral development.
Article
The clinical utility of doxorubicin is compromised due to dose related toxic side effects and limited oral bioavailability with no oral formulation being marketed. Enhancement of intestinal absorption and magnification of cytotoxicity can overcome these limitations. Accordingly, the objective was to probe penetration enhancers, bilosomes and their combinations for enhanced intestinal absorption and improved cytotoxicity of doxorubicin. Piperine and dipyridamole were tested as enhancers alone or encapsulated in bilosomes comprising Span60, cholesterol and bile salts. Bilosomes were nanosized spherical vesicles with negative zeta potential and were able to entrap doxorubicin with efficiency ranging from 45.3% to 53%. Intestinal absorption studies utilized in-situ rabbit intestinal perfusion which revealed site dependent doxorubicin absorption correlating with regional distribution of efflux transporters. Co-perfusion with the enhancer increased intestinal absorption with further augmentation after bilosomal encapsulation. The latter increased the % fraction absorbed by 4.5-6 and 1.8-2.5-fold from jejuno-ileum and colon, respectively, depending on bilosomes composition. Additionally, doxorubicin cytotoxicity against breast cancer cells (MCF-7) was significantly improved after bilosomal encapsulation and the recorded doxorubicin IC50 value was reduced from 13.3 μM to 0.1 μM for the best formulation. The study introduced bilosomes encapsulating absorption enhancers as promising carriers for enhanced cytotoxicity and oral absorption of doxorubicin.
Article
Full-text available
Nanocrystals research has been an area of significant interest lately, providing oral bioavailability benefits to solubility- and/or dissolution rate-limited drugs. Drug nanocrystals are generated using top-down or bottom-up technologies. Combination technologies (Nanoedge, Nanopure XP and SmartCrystal) have been recently developed to generate nanocrystals of improved properties. Our lab has also contributed in this field by providing a ‘novel’ platform technology, NanoCrySP, for the generation of nanocrystals. NanoCrySP-generated nanocrystals have improved the oral bioavailability of various molecules. In this study, we aim to assess the permeability behavior of nanocrystals generated by NanoCrySP. Three samples of Dipyridamole (DPM) drug were used in this study: (1) DPM (micron-sized powder), (2) nanocrystals of DPM (NS), generated by media milling (as control) and, (3) nanocrystalline solid dispersion containing DPM (NSD) in the matrix of mannitol (MAN), generated using NanoCrySP technology. In vitro (Caco-2 cell lines) and ex vivo (everted gut sac) studies were conducted in this work. Cellular permeability (Papp) from apical-to-basolateral side in Caco-2 cell monolayer was found to be in the order NS > NSD > DPM, which was the same as their apparent solubility values. Higher Papp from a basolateral-to-apical side suggested a significant contribution of the P-gp efflux transport for DPM, while NS exhibited much higher inhibition of the efflux mechanism than NSD. Both NS and NSD showed higher permeation from the jejunum region in the ex vivo everted gut sac study. Interestingly, Papp of NSD was similar to NS in ex vivo everted gut sac model, however, NSD showed higher mucoadhesion than NS and DPM in this study.
Article
Full-text available
Originally discovered almost a century ago, cocrystals continue to gain interest in the modern day due to their ability to modify the physical properties of solid-state materials, particularly pharmaceuticals. Intensification of cocrystal research efforts has been accompanied by an expansion of the potential applications where cocrystals can offer a benefit. Where once solubility manipulation was seen as the primary driver for cocrystal formation, cocrystals have recently been shown to provide attractive options for taste masking, mechanical property improvement and intellectual property generation and extension. Cocrystals are becoming a commercial reality with a number of cocrystal products currently on the market, and more following in registration and clinical trial phases. Increased commercialisation of cocrystals has in turn necessitated additional research on methods to make cocrystals, with particular emphasis placed on emerging technologies that can offer environmentally attractive and efficient options. Methods of producing cocrystals and of harnessing the bespoke physical property adjustment provided by cocrystals are reviewed in this article, with a particular focus on emerging trends in these areas.
Article
Full-text available
Introduction: Dipyridamole is an Anti-platelet agent exhibits release problems at higher pH of small intestine due to its pH dependent solubility and precipitation followed by interruption of drug release from dosage form. To overcome this extended release formulation was developed by using pH modulating agent (tartaric acid). Objective: Present study was undertaken with a view of the formulations evaluated by performing dissolution testing on developed extended released tablets. Method: development of dissolution method at different time points and USP Apparatus 1 (basket) and 2 (paddle) at rotating speeds of 50 or 100 rpm used to evaluate the release characteristics of the formulations. Furthermore, solubility and in vitro dissolution studies of formulated tablets were performed at pH values of 1.2 and 5.5. Results: In this study we found increasing volume of dissolution medium pH 5.5 phosphate buffers drug precipitation is increased. The developed dissolution method was validated according to ICH guidelines for various parameters such as specificity, accuracy, precision, and stability. The dissolution method was confirmed by determining the dissolution rate of extended released Dipyridamole tablets containing pH modulating agent. The best in vitro dissolution profile was obtained using pH 5.5 phosphate buffer as the dissolution medium (500 ml) stirred at 100 rpm. A comparison of the dissolution profiles in official and developed media showed significant differences based on f1 and f2 values. Conclusion: The developed dissolution test exhibited a higher capacity than the compendia methods in differentiating the release profiles of pH independent extended release tablets. It can be applied during formulation development and quality control analysis of pH independent extended release tablets for evaluation of the effects of pH modifier in dissolution medium and processing parameters. © 2018, Association of Pharmaceutical Teachers of India. All rights reserved.
Article
Full-text available
Baicalein (BE) is one of the main active flavonoids representing the variety of pharmacological effects including anticancer, anti-inflammatory and cardiovascular protective activities, but it's very low solubility, dissolution rate and poor oral absorption limit the therapeutic applications. In this work, a nano-cocrystal strategy was successfully applied to improve the dissolution rate and bioavailability of BE. Baicalein-nicotinamide (BE-NCT) nano-cocrystals were prepared by high pressure homogenization and evaluated both in vitro and in vivo. Physical characterization results including scanning electron microscopy, dynamic light scattering, powder X-ray diffraction and differential scanning calorimetry demonstrated that BE-NCT nano-cocrystals were changed into amorphous state with mean particle size of 251.53 nm. In the dissolution test, the BE-NCT nano-cocrystals performed 2.17-fold and 2.54-fold enhancement than BE coarse powder in FaSSIF-V2 and FaSSGF. Upon oral administration, the integrated AUC0 - t of BE-NCT nano-cocrystals (6.02-fold) was significantly higher than BE coarse powder (1-fold), BE-NCT cocrystals (2.87-fold) and BE nanocrystals (3.32-fold). Compared with BE coarse powder, BE-NCT cocrystals and BE nanocrystals, BE-NCT nano-cocrystals possessed excellent performance both in vitro and in vivo evaluations. Thus, it can be seen that nano-cocrystal is an appropriate novel strategy for improving dissolution rate and bioavailability of poor soluble natural products such as BE.
Article
Full-text available
Cocrystals can be used as an alternative approach based on crystal engineering to enhance specific physicochemical and biopharmaceutical properties of active pharmaceutical ingredients (APIs) when the approaches to salt or polymorph formation do not meet the expected targets. In this article, an overview of pharmaceutical cocrystals will be presented, with an emphasis on the intermolecular interactions in cocrystals and the methods for their preparation. Furthermore, cocrystals of direct pharmaceutical interest, along with their in vitro properties and available in vivo data and characterization techniques are discussed, highlighting the potential of cocrystals as an attractive route for drug development.
Article
Full-text available
The objective of this research was to prepared acyclovir cocrystals with succinic acid as coformer using three different solvents (ethanol, acetic acid glacial, and 0.1N HCl) to influence the characters and improve the dissolution rate of acyclovir. Cocrystallization of acyclovir with succinic acid as coformer was successfully prepared by solvent evaporation technique using three different solvents (ethanol, acetic acid glacial, and 0.1N HCl). The screening indicated that acyclovir formed novel cocrystals with succinic acid in the three different solvents. PXRD profile show that there is three peaks in ethanol cocrystal in angle 2θ 5.9134 o ; 9.1645 o and 13.4044 o. For acetic acid glacial and 0.1N HCl cocrystal there is one peak in angle 2θ 5.9263 o and 2θ 9.6011 o. In analysis diffractogram DSC formed ethanol cocrystal with melting point 175.84 o C. The melting point of acetic acid glacial cocrystal is 178.41 o C and 0.1N HCl cocrystal is 156.75 o C. The dissolution rate of the cocrystals measured by the effisiency disolution (ED 45) that considerable faster than that pure acyclovir and the physical mixtures. On the result of FTIR analysis there is changes of the wavenumber that indicated that there is cocrystal formed. And for SEM analysis, morphology of cocrystals was different than the original materials. In dissolution test, ethanol and acetic acid glacial cocrystals have better efficiency dissolution (ED 45) (92.96 %) than the acyclovir ED 45 (84.48 %). But 0.1N HCl cocrystal has lower ED 45 than acyclovir that is 48.19 %. The results obtained in this research indicated the acyclovir cocrystals have formed with succinic acid as coformer using three different solvents. The physical properties was different from the three cocrystals. Dissolution rate of acyclovir cocrystals using ethanol and acetic acid glacial solvents was increase, whereas using 0.1N HCl was decrease rather than pure acyclovir.
Article
Full-text available
In recent years, co-crystals have been the subject of increasing interest within the pharmaceutical industry because these new solid forms of active pharmaceutical ingredients have the ability to enhance the bioavailability of poorly water-soluble drugs with a low dissolution rate. For this reason, it is crucial to prepare co-crystals of benzodiazepines, i.e. psychoactive drugs with a wide range of medical applications but classified as very slightly or practically insoluble in water. Thus, the objective of this research was to show to what extent the DSC method can be used as a screening tool for detection of co-crystal formation in binary physical mixtures of drugs and co-formers. To obtain potential co-crystals, eight benzodiazepines (diazepam, temazepam, oxazepam, lormetazepam, lorazepam, clonazepam, estazolam and chlordiazepoxide) were gently mixed in an agate mortar at 1:1 molar ratios with nine co-formers—succinic, glutaric, fumaric, citric and p-aminobenzoic acids, nicotinamide, saccharin, urea and caffeine, and heated under DSC conditions. A detailed comparison of the DSC scans of mixtures against scans of both ingredients in isolation indicates the occurrence of subtle physical changes in the samples. Thus, additional endothermic or exothermic peaks due to melting or crystallisation suggested the formation of potential co-crystals. To conclude, this study confirms that the DSC method can be used as a rapid screening tool for co-crystal detection. In this case, based on the DSC scans, 15 physical mixtures of benzodiazepines (clonazepam, diazepam, lorazepam, oxazepam and temazepam) with co-formers (citric, fumaric, glutaric, p-aminobenzoic and succinic acids, nicotinamide, saccharin and urea) have been selected as potential co-crystals for further detailed study.
Article
Quercetin has been proposed to exhibit numerous pharmacological benefits yet suffer low bioavailability due to the extremely low solubility. A research to study the impact of cocrystallization of quercetin with succinic acid on the solubility and dissolution profile has been performed. Cocrystallization in molar stoichiometry of 1:1 was carried out via liquid assisted grinding with methanol in ball milling apparatus. Cocrystal formation was identified by hot stage microscopy (HSM) at first, then cocrystal phase was characterized using differential thermal analysis (DTA), powder X-ray diffractometry (PXRD), scanning electron microscopy (SEM), and fourier-transform infrared (FT-IR) spectroscopy. Solubility and dissolution test were conducted as well. DSC thermogram exhibits new endothermic peak at 280.32°C representing the melting point of cocrystal phase alongside with endothermic point of pure compounds. Powder X-ray diffractograms show new diffraction peaks on behalf of cocrystal formation at 2θ=8.92, 9.88, 13.04, 29.78, 35.35°. FT-IR spectroscopy reveals band shifting in –OH group region. On SEM photographs, one can observe crystal habit of succinic acid being covered by crystal with different habit. This indicates that quercetin interacts with succinic acid only on the surfaces and causes imperfect formation of cocrystal phase. Cocrystallization quercetin improves solubility by 1.62 times higher and dissolution rate by 1.25 higher than pure quercetin (one-way ANOVA, p < 0.05).