You are on page 1of 7

International Journal of Biological Macromolecules 131 (2019) 879–885

Contents lists available at ScienceDirect

International Journal of Biological Macromolecules

journal homepage: http://www.elsevier.com/locate/ijbiomac

Chitosan-telmisartan polymeric cocrystals for improving oral absorption:


In vitro and in vivo evaluation
Mani Ganesh a,⁎, Udhumansha Ubaidulla b, Grace Rathnam b, Hyun Tae Jang a,⁎
a
Department of Chemical Engineering, Hanseo University, 360 Daegok-ri, Seosan-si 356 706, Chungcheongnam-do, South Korea
b
Department of Pharmaceutics, C.L. Baid Metha College of Pharmacy, Chennai, India

a r t i c l e i n f o a b s t r a c t

Article history: This study describes the development of polymeric cocrystals of chitosan-telmisartan (TEL) to improve the oral
Received 27 December 2018 bioavailability of TEL, which has poor oral solubility and bioavailability. The polymeric cocrystal was prepared
Received in revised form 12 March 2019 using chitosan a biopolymer with the aid of sodium citrate as a salting-out agent. The cocrystals were character-
Accepted 21 March 2019
ized by FT-IR spectroscopy, scanning electron microscopy, differential scanning calorimeteri (DSC), thermogravi-
Available online 22 March 2019
metric analysis (TGA), and powder X-ray diffraction (PXRD). The improved solubility of TEL was observed with
Keywords:
cocrystals as compared to that of pure drug in solubility studies with phosphate buffer (pH 7.4). The in vivo
Telmisartan pharmacokinetics properties of cocrystal were studied by an animal model using rats after a single dose oral ad-
Chitosan ministration. The results showed an increased plasma drug concentration (Cmax) of 1.47, μg/ml for cocrystals
Polymeric cocrystals when compared to pure TEL with 0.96 μg/ml with one-fold increased bioavailability (F%) that is, the cocrystals
Oral absortion increases the solubility of the drug and the paracellular drug absorption by tight junction modulation. Further
Bioavailability the elimination constant Kel resulted with higher value of about 0.0085 h−1 when compared to pure drug
with0.0048 h−1 along with improved AUC (14.62 μg/ml).
© 2019 Elsevier B.V. All rights reserved.

1. Introduction in vivo pharmacokinetics [16]. Mutalik et al. reported that aceclofenac-


chitosan crystals could improve in vitro dissolution and in vivo pharma-
Dissolution of a drug is considered to be the rate-determining step cokinetic properties of drugs. The coformer chitosan (CS) is “generally
for oral absorption of the poorly water-soluble drugs. Selection of a recognized as safe” (GRAS) by the US FDA and is widely regarded as a
method to increase solubility is the key to achieve good oral bioavail- biocompatible, nontoxic, and non-allergenic material that is, therefore,
ability, reduce frequency of dosing, have better patient compliance, highly suitable for use in medical and pharmaceutical applications
and lower the cost of production. Many techniques have been reported [17–19].
to improve drug dissolution, such as particle-size reduction, salt forma- Telmisartan (4′-{[4-Methyl-6-(1-methyl-2-benzimidazolyl)-2-
tion, solid dispersion, use of a surfactant, and complexation [1–5]. Re- propyl-1-benzimidazolyl] methyl}-2-biphenylcarboxylic acid) has
cently, pharmaceutical cocrystals have received much attention in the been used for treatment of hypertension that belongs to the BCS class
pharmaceutical industry for improving the bioavailability of poorly II drugs [20,21]. A major problem with this drug is its low solubility
water-soluble drugs, especially for these neutral compounds or those in biological fluids, which results in poor bioavailability after oral ad-
having weakly ionizable groups. Moreover, many reports revealed that ministration. The solubility of TEL in aqueous medium is very low:
cocrystals could improve the physicochemical properties of the drug, 0.078 mg/ml in water. Absolute bioavailability of the TEL was 42–58%.
such as melting point, solubility, dissolution, and stability [6–15]. Poor solubility of TEL leads to poor dissolution and hence it shows
Pharmaceutical cocrystal is prepared based on the supramolecular variations in bioavailability [22]. For improving the solubility and
chemistry; i.e., drug and crystal former are combined to form cocrystals bioavailability of TEL, various formulations have been reported, such
by means of intermolecular interactions, mainly hydrogen bonds and as immediate release tablets [23], nanoparticles [24,25], nano self-
van der Waals forces. Many API–coformer combinations in orally ad- emulsifying drug-delivery systems [26], amorphous formulations [27],
ministrated pharmaceutical cocrystals are considered to dissociate solid dispersions [28,29], and alkalizers [30]. Lin et al. revealed that
upon dissolution and dilution in the gastrointestinal tract, as suggested TEL–chitosan solid dispersions are formed by electrostatic interactions
by studies on the relationship between cocrystal in vitro dissolution and between TEL and CS caused by the anionic nature of the drug and the
strong positive charge of the polymer [29]. Cocrystals using a crystal-
⁎ Corresponding authors. engineering approach is one technique used to improve the dissolution
E-mail addresses: chemgans@gmail.com (M. Ganesh), htjang@hanseo.ac.kr (H.T. Jang). and bioavailability of poorly soluble drugs. The ionizable functionalities

https://doi.org/10.1016/j.ijbiomac.2019.03.141
0141-8130/© 2019 Elsevier B.V. All rights reserved.
880 M. Ganesh et al. / International Journal of Biological Macromolecules 131 (2019) 879–885

of TEL present possibilities for co-crystallization by hydrogen bonding. 5–45° with a 0.02° step size and a 1 s step time was fixed as the param-
There have been very few reports of studies that used this approach, eter for retrieving the XRD.
using various [31–34] acids like citric, gallic, or saccharin as co-
formers. Using weak acids as co-formers will also cause some problem, 2.3.2. Scanning electron microscopy (SEM)
such as poor dissociation, gastric irritation, and alkalizing the microen- To study the morphology of the pure drug and cocrystal formulation,
vironment of the intestinal lumen. Chitosan is the best biodegradable SEM analysis was performed. Samples were dusted over the double-
co-former that can increases the solubility of poorly soluble drugs, sided carbon adhesive tape, placed over the alluminium stub, and
as stated earlier [17–19,34,35]. This has prompted us to explore the coated with gold by a plasma sputter coater (Cressington, Sputter
possibility of developing TEL-CS cocrystals by a one-step process using Coater-108 auto, Korea). The images were recorded using a scanning
a citrate ion without any toxic cross linking agents, and to study their electron microscope (Jeol-JSM 5600, Japan).
potential for improving dissolution rate and physicochemical proper-
ties. Our objective is to develop a TEL cocrystal using chitosan to 2.3.3. FT-IR
increase the solubility and dissolution rate of TEL by preparing its FT-IR spectral analysis of TEL and various drug-chitosan cocrystal
cocrystals using citrate-ion cross linking and to carry out physicochem- formulations were recorded using a Thermoscientific Nicolet IR 200
ical characterization of the optimal formulation by using infrared spectrometer by KBR pressed-pellet technique. The KBr pellets were
spectroscopy, XRD, DSC, and surface morphology, as well as an in vitro made by mixing required quantities of pure drug (TEL), chitosan, and
drug release and in vivo oral absorption study. various formulations (TCC-1 to TCC-8) with 100 mg of dried KBr and
pressed into pellets using a KBr pellet press. Then each pellet was
2. Materials and methods scanned 40 times at 4 cm−1 resolution over the wavenumber range of
4000–400 cm−1.
2.1. Manterials
2.3.4. DSC
Telmisartan (TEL) and chitosan (low molecular weight, N 85% To study the changes in the crystal structure during the cocrystal
deacetylated, poly(d-glucosamine)) were procured from TCI Chemical, formulation, DSC analysis was done. A Scinco DSC N 650 differential
Japan, and glacial acetic acid was purchased from Dae Jung Chemicals scanning calorimeter was used to record the DSC traces of the TEL
and Metals, Korea. All other chemical used herein were of commercial pure and cocrystals. The samples were loaded into the aluminum pan
grade unless otherwise specified in the respective areas. of the DSC instrument and scanned at a heating rate of 10 °C/min
under a helium environment by purging helium at 40 ml/min.

2.2. Preparation of cocrystals 2.3.5. TGA


Pure drug and various cocrystal samples were loaded (ca. 10 mg)
Drug and chitosan macromolecular cocrystals were prepared as re- into the platinum sample pan of the Scinco N-1000 thermogravimeter
ported in our earlier research on aceclofenac chitosan cocrystals unit, and the temperature was programmed to reach 600 °C at a heating
[18,19], but with a little modification. In the typical protocol the drug rate of 10 °C/min in a nitrogen atmosphere.
was separately dissolved in methanol and the chitosan was dissolved
in 1% acetic acid (10 ml). Then the solutions of drug and polymer 2.4. In vitro drug release
were simultaneously added drop wise to 1% sodium citrate solution
(citrate common ion) that acts as a salting-out agent with stirring The in vitro release of TEL and cocrystals was tested in a USP XXII dis-
using high-speed homogenizer (Daihan Scientific,Korea) at a rate of solution apparatus. An equivalent weight of cocrystals containing 40 mg
1000 rpm. The cocrystals were formed while the addition began. After of TEL was placed separately in 900 ml of intestine fluid (pH 7.4) main-
the complete addition of the two solutions, the stirring was continued tained at 37 ± 0.5 °C and stirred at 50 rpm. About 5 ml aliquots of was
for another 30 min to evaporate the methanol; the resultant was then collected at regular time intervals, and the same amount of fresh
filtered through Whatman filter paper (No. 1) and dried under vacuum dissolution medium was replaced into a dissolution vessel to maintain
at 45 °C overnight. The resulted powder was further subjected to sifting the sink condition throughout the experiment. The aliquots were fil-
using a sieve with mesh no #40. Different formulations were prepared tered, further diluted suitably, and estimated spectrophotometrically
using a fixed drug concentration with different concentrations of at 296 nm.
polymer, as listed in Table.1. The formulations obtained were labeled
TCC-1 (Temisartan chitosan cocrystals), and TCC-2 to TCC-8 in terms 2.5. In vivo evaluation
of their drug-polymer weight ratios.
The study protocol was approved by the Research and Ethical Com-
2.3. Characterizations mittee of C.L. Baid Metha College of Pharmacy, India (Ethical clearance
no: 17/321/PO/Re/S/01/CPCSEA). For in vivo studies, healthy albino
2.3.1. XRD rats of either sex, weighing 200 ± 20 g, were used. The rats were
Changes in the crystalline structure of TEL before and after formula- divided into three groups, each consisting of six rats. Group 1 animals
tion were studied using Powder X-ray diffraction. The required quantity were administered the pure drug, Group 2 animals received an
of drug and formulations were placed in the XRD sample holder then optimized formulation (TCC-6), and Group 3 animals were treated as
scanned using Rigaku Miniflex X-ray diffractometer with Cu Kα radia- the control. The rats were kept in individual cage for seven days prior
tion and operated at 40 kV and 30 mA (l = 1.54 (Å); a 2θ range of to the study in the departmental animal house for acclimation. The

Table 1
Composition of various co-crystals formulation.

Component TCC-1 TCC-2 TCC-3 TCC-4 TCC-5 TCC-6 TCC-7 TCC-8

1. TEL(mg) in 10 ml water 40 40 40 40 40 40 40 40
2. Chitosan (mg) in 10 Acetic acid (1%) 40 80 120 160 200 240 280 320
3. Sodium citrate (1%, ml) 100 100 100 100 100 100 100 100
M. Ganesh et al. / International Journal of Biological Macromolecules 131 (2019) 879–885 881

cocrystals were administered to the rats through the feeding tube were observed at 2θ of 6.9°,10.5°, 11.84°, 12.0°, 12.4°, 14.16°, 15°,
(oral gavage) (1 mg/kg., p.o). The blood samples were collected from 18.6°, 23.78°, 25°, 2.9° and 31.0°, 33.74°, 34.14°, and 40.43° [31,32].
the tail vein of rats in centrifuge tubes (containing sodium heparin as The major diffraction peaks from 5 to 40 (2θ), such as 6.9°, 10.5°,
an anti-coagulant). The blood samples were collected at 0.5, 1, 1.5, 2, 11.84°, 12.0, 12.4°, 14.16°, 15°, 18.6°, and 23.7° were also observed
3, 4, and 6 h for the pure drug and cocrystals. The collected blood with formulations like TCC-1 to TCC-4, but with decreased intensity.
samples were centrifuged at 3500 rpm for 15 min to separate the These results proved that the drug-polymer ratio of until 1:4 (wt) has
plasma for drug-content analysis. The plasma was separated carefully less effect on the crystalline structure of TEL, but on further increase in
and stored at 2–10 °C. The plasma samples were analyzed, using a polymer weight, the diffraction intensities were reduced gradually
reversed-phase high-performance liquid chromatography (HPLC) with and the amorphous phase slowly appeared because of the increased
reference to procedure portrayed by Shen et al. [36] as follows about polymer concentration, the polymer deposits masks the diffraction or
0.5 ml of plasma sample was made acidic with 1 M HCl. The solutions reaching of X-rays to the planes of TEL because of the formation of
were violently vortexed with 4 ml of ethyl acetate for 5 min. Then thick gel like layer over the crystals. In case of unit cell structure
they were centrifuged at 4000 g for 10 min and the organic phase was the PXRD patterns of the pure TEL crystals gives a triclinic symmetry
transferred to a clean tube and evaporated to dryness at 50 °C with (a = 14.679 Å,b = 9.689 Å, c = 6.695, α = 95.793, β = 93.125 and
the aid of a gentle stream of N2 and analyzed by HPLC. γ = 94.855) with P1 space group (using EXPO2014 crystal structure so-
lution,freeware from http://www.ba.ic.cnr.it/softwareic/expo/). But in
2.6. In vivo pharmacokinetic analysis case of polymer drug cocrystals the polymer disrupted the crystalline
structure of TEL and forms new amorphous structure by hydrogen
The most common pharmacokinetic parameters, such as peak plasma bonding between carboxyl –C=O TEL with –NH (free amino or
concentration (Cmax), time to reach maximum plasma concentration amide) of the chitosan and forming newer synthons as explained earlier
(Tmax), and total area under the plasma concentration-time curve studies [34]. This further supported by the following FTIR results and
(AUC0–∞), were measured from the plasma-concentration time profile DSC. Crystal structure elucidation for the amorphous co-crystal with
of the cocrystal and pure drug. The peak plasma concentration (Cmax) available PXRD data is impracticable and hence, the results from FT-IR
and time of its occurrence (Tmax) were calculated directly from each study were only used to determine the possible interaction between
plasma concentration. The AUC of cocrystal and pure drug were calcu- the drug and polymer. The similar interaction was also reported with
lated by trapezoidal method. The elimination rate constant (Kel) of TEL cocrystals with atenolol and irbesartan [34].
cocrystals and pure drug was calculated using Eq. (1). Relative bioavail-
ability (%) of TEL cocrystal was calculated using equation (Eq. (2)). 3.2. FT-IR

Kel ¼ Slope=0:203 ð1Þ Fig. 2 shows the FT-IR spectrum of pure TEL and it cocrystal formula-
tions. The FT-IR spectrum of the pure drug is shown in Fig. 1 (TEL), in
AUCtest Doseref which the vibrational band at around 3450 cm−1 was attributed to
%Relative bio−availability ¼   10 ð2Þ
AUCref Dosetest O\\H stretching of \\COOH; the aromatic C\\H stretching vibrations
were observed near 2900 cm−1, \\OH bending and \\C_O stretching
2.7. Statistical analysis of\\COOH acid at 1385 cm- 1, and C_C ring vibrations of the aromatic
nucleus near 1600 cm−1. Further, the bands near 1350–1210 cm−1
All measured data were expressed as mean ± standard deviation were assigned to the C\\N stretching of the imidazole ring of TEL. The
(S.D.), and were analyzed using BioStat version 2009 for Windows pure chitosan (Supplementary Fig. S1) had predominant peaks at
software. 3750 and 3000 cm−1 for the stretching vibrations of OH that was
merged with N\\H stretching of the glucosamine moiety. The vibra-
3. Results and discussions tional band near 2850 cm−1 was attributed to the CH bond of CH2 of
the sugar moiety. In addition, the methylene bending was observed
3.1. XRD near 1375 cm−1 [18,19].
For the formulation TCC-1, the major peaks of TEL in the fingerprint
The XRD results of pure drug TEL and its cocrystal formulation are region were clearly visible. The intensity of the transmittance band
shown in Fig. 1. The major characteristic diffraction peaks for pure TEL

Fig. 1. PXRD properties of Pure and chitosan cocrystals of telmisartan. Fig. 2. FTIR spectrum of Pure drug and chitosan- telmisartan cocrystals.
882 M. Ganesh et al. / International Journal of Biological Macromolecules 131 (2019) 879–885

caused by the aromatic –CH stretching vibrations near 2900 cm−1 were structure under low magnification (figure not shown). At high magnifi-
reduced, but there was a broad band caused by –OH vibration by water cation, the (X5000) crystals of TEL with deposits of polymer were seen
molecules of the solvent, and –OH and –NH stretching of TEL were clearly in TCC-1 and TCC-2, but the number and size of crystals were re-
observed from 2750 to 3600 cm−1 with little shifting from its original duced in TCC-3 and TCC-4. Even though the number of crystals was re-
position and broaden when compared to pure TEL. Further the – C_O duced, their morphology was impervious. The pores over the surface of
stretching of – COOH acid at 1385 cm- 1 and the vibrational bands the cocrystals may support the imbibitions of the solvent and biological
near 1350–1210 cm−1 for C\\N stretching of the imidazole ring of TEL fluids and thereby increase the solubility and bioavailability of TEL as
also shifted/disappeared that proves the participation hydrogen bond- expected. This crystalline character was supported by the XRD data, as
ing during the co-crystallization of newer synthons with (O\\H⋯O, discussed earlier. For the TCC-5 to TCC-8 formulations, the TEL crystals
O\\H⋯N, N\\H⋯O, and N\\H⋯N) interaction of TEL and chitosan to were not visible; only the polymer surface was observed with some
form multi component cocrystals through (1) Non-covalent forces like small protrusions caused by crystals inside the surface.
acid–amide, alcohol-amide, acid–acid, and amide–amide interactions
that are usually of hydrogen bonding nature that holds the drug and 3.4. Differential scanning calorimetric (DSC)
conformer together in the cocrystal. Likewise, the percentage transmit-
tance of the aromatic-ring vibration caused by C_C of the aromatic Fig. 4 is the DSC thermogram of TEL and cocrystal formulations. The
functionality at around 1600–1800 cm−1 was also reduced that was in DSC of pure TEL shows a sharp melting endotherm peak that stretches
observed in all formulations, but the intensity was reduced by increas- from 261 °C to 276 °C, with a peak maximum at 270.5 °C [37], equivalent
ing the ratio of polymer. For the formulations TCC-5-TCC-7, the band to the melting endothermic range of pure TEL. For the cocrystal formu-
at 2900 cm−1 of the aromatic \\CH vibration is not visible, because lations TCC-1 andTCC-2, the endothermic peak for TEL shifted slightly
of over depositing of chitosan over the drug which makes the TEL toward a lower value (266 °C), due to the changes in the crystalline
complete amorphous crystals instead of ionic crystals. Similar type of geometry of TEL caused by the deposition of the chitosan over the TEL
interaction was also reported earlier with cocrystals of TEL with other by the hydrogen-bonded interaction of the –COOH and –NH(imidazole)
co-formers [34,35]. group of TEL with the free amino or amide group of chitosan. However,
the peak appearance clearly shows that there was no ionic interaction
3.3. Scanning electron microscopy that formed with other TEL cocrystals in earlier studies [33,34].
In the formulations from TCC-3 to TCC-8, the endothermic peak
The scanning electron microscopic images of the pure drug TEM and intensity was gradually get reduced and unidentifiable from TCC-6
the formulations are depicted in Fig. 3. Pure TEL shows small, needle- to TCC-8, which shows the changing of the crystalline structure to
like, pure pristine crystals, whereas the TCC-1 to TCC-4 cocrystals amorphosization of TEL crystals caused by the over depositing of
was soft with rough morphological surfaces consisting pores in their chitosan.

Fig. 3. SEM morphological characters of pure drug and chitosan- telmisartan cocrystals.
M. Ganesh et al. / International Journal of Biological Macromolecules 131 (2019) 879–885 883

Fig. 4. DSC traces of pure drug and chitosan- telmisartan cocrystals.

3.5. TGA Fig. 6. In-vitro drug release profile of pure drug and cocrystal formulations.

The results of the thermogravimetric analysis are depicted in Fig. 5.


in which the TGA of pure TEL shows a two-step degradation profile.
The first degradation of TEL starts near 265 °C, as is supported by the the surface morphology of the drug, and reducing particle size. The de-
DSC results as earlier discussed; that is, it starts to melt at 270 °C. After positing of the chitosan polymer over the drug particles was higher and
265 °C to 410 °C, there is a huge weight loss of around 50%; after that, stronger when associated with sodium citrate. The reaction of chitosan
the second weight loss begins and end near 650 °C, with around a 43% with multivalent anion sodium citrate leads to the formation of bridges
weight loss, which is similar with earlier reports [37]. However, for between the polymeric chains, which results in efficient deposition of
TCC-1 and TCC-2, the first weight loss started a little earlier than that chitosan by forming a three dimensional network on the drug crystals.
of pure TEL, at 195 °C, and ends before 350 °C; the second weight loss This might have eventually resulted in efficient adsorption of chitosan
was similar with TEL but the thermogram ended before 600 °C. These on the drug crystals. Hence, these cocrystals can improve the solubility
results prove that the chitosan deposit over the crystals affect the crystal of the drug [18,33]. If the e concentration of the polymer further
structure of the TEL and convert the crystals into amorphous powder increased the dissolution decreases (TCC-7 and TCC-8) due to the
gradually from TCC-1–TCC-8. formation thick gel layer of the chitosan on the surface of the drug,
these gel structure will increase the distance between the core TEL
3.6. In vitro drug-release study crystals from the outside solvent imbibing. These results are well
corroborated by a previous study that described acyclovir cocrystals
All the formulations (TCC-1 to TCC-8) of the prepared cocrystals of prepared by chitosan [38].
TEL were subjected to in vitro release studies, which were carried out
using a USP dissolution apparatus type II using pH 7.4 buffer for 2 h. 3.7. In vivo pharmacokinetics evaluation study
The drug release increased when the concentration of chitosan in-
creased (Fig. 6). Chitosan is a well-known biocompatible co-former The ultimate goal of TEL cocrystals prepared using a biodegradable
used to prepare cocrystals that increases the dissolution rate of TEL by polymer is to improve the drug's oral bioavailability. The evaluation of
increasing its wettability by its hydrophilic nature there by altering the bioavailability of the drug in animals is an important parameter to
consider when preparing new dosage forms. The plasma drug levels
after a 1 mg/kg single dose of pure drug and of TEL cocrystals are
shown in Table 2 and Fig. 7. A significant increase in the peak plasma
concentration (Cmax) was observed for TEL cocrystals (1.47 μg/ml)
over that of pure TEL (0.96 μg/ml). The pharmaceutical practical
implication of these cocrystals is significant, since the ability to form a
supersaturated solution, even transiently that can dramatically affect
the absorption and bioavailability. The level of plasma drug concentra-
tions was increased in the cocrystals. The elimination rate constant (Kel)
of TEL cocrystals (0.0085 h−1) was higher than that of the pure drug
(0.0048 h−1). The improved area under the curve (AUC) (14.62 μg/ml)

Table 2
Pharmacokinetic parameters.

S. No Pharmacokinetics parameters TEL pure drug TEL cocrystals

1. Cmax (ng/ml) 0.96 1.47


2. AUC (ng·ml−1·hr) 10.44 14.62
3. Kel (hr−1) 0.0048 0.0085
4. Relative bioavailability (%) – 140.03
Fig. 5. TGA traces of pure drug and chitosan- telmisartan cocrystals.
884 M. Ganesh et al. / International Journal of Biological Macromolecules 131 (2019) 879–885

are one of best choice improve the solubility and bioavailability of not
only the telmisartan but also have the ability to improve the for other
solubility and bioavailability poor soluble drug upon evaluation.
Supplementary data to this article can be found online at https://doi.
org/10.1016/j.ijbiomac.2019.03.141.

Acknowledgement

This work was financially supported by the Hanseo University Intra-


mural Research Grant 2018.

Declaration of conflicts of interest

None.

References
[1] T. Vasconcelos, B. Sarmento, P. Costa, Solid dispersions as strategy to improve oral bio-
availability of poorly water soluble drugs, Drug Discov. Today 12 (2007) 1068–1074.
[2] D.N. Bikiaris, Solid dispersions, part I: recent evolutions and future opportunities in
manufacturing methods for dissolution rate enhancement of poorly water-soluble
Fig. 7. In-vivo pharmacokinetics of selected formulation. drugs, Expert Opin. Drug Deliv. 8 (2011) (15 01-1 509).
[3] G. Corti, G. Capasso, F. Maestrelli, M. Cirri, P. Mura, Physical– chemical characteriza-
tion of binary systems of metformin hydrochloride with triacetyl-β-cyclodextrin, J.
Pharm. Biomed. Anal. 45 (2007) 480–486.
was found in TEL cocrystals. The increase in Cmax can be attributed to the [4] P. Vishweshwar, J.A. McMahon, J.A. Bis, M.J. Zaworotko, Pharmaceutical co- crystals,
increase in the dissolution and absorption rates of the drug. The improved J. Pharm. Sci. 95 (2006) 499–516.
[5] D.Q.M. Craig, The mechanisms of drug release from solid dispersions in water- sol-
gastrointestinal absorption may result because the positive charge of the
uble polymers, Int. J. Pharm. 231 (2001) 131–144.
chitosan is the factor responsible for interaction with the negative site of [6] A.S. Shete, A.V. Yadav, M.S. Murthy, Enhancement of dissolution rate of Irbesartan by
the tight junction and the subsequent widening of the paracellular chitosan based crystal engineering technique, Ind, J. Pharm. Edu. Res. 46 (2012)
transport route [39] which is an advantage of chitosan cocrystals over 323–329.
[7] G. Corti, F. Maestrelli, M. Cirri, P. Mura, N. Zerrouk, Dissolution and permeation prop-
the conventional acid (or) salt based cocrystals reported in the previous erties of naproxen from solid-state systems with chitosan, Drug Deliv. 15 (2008)
studies [33,34]. Unlike other salt based cocrystals chitosan like polymers 303–312.
will support for the mucoadhesion that may also tends raise the contact [8] F. Maestrelli, M. Cirri, N. Mennini, N. Zerrouk, P. Mura, Improvement of oxaprozin
solubility and permeability by the combined use of cyclodextrin, chitosan, and bile
between the drug and biological membrane and increasing the bioavail- components, Eur. J. Pharm. Biopharm. 78 (2011) 385–393.
ability. Further the chitosan cocrystals are more advantageous when [9] C. Grossjohann, D.R. Serrano, K.J. Paluch, P. O'Connell, L. Vella-Zarb, P. Manesiotis, T.
compared to the chitosan TEL solid dispersion reported earlier inters McCabe, L. Tajber, O.I. Corrigan, A.M. Healy, Polymorphism in sulfadimidine/4-
aminosalicylic acid cocrystals: solid-state characterization and physicochemical
uniform formation because the both drug and coformer was added properties, J. Pharm. Sci. 104 (2015) 1385–1398.
in solution form into the salting out agent, but in the reported solid [10] A.M. Healy, Z.A. Worku, D. Kumar, A.M. Madi, Pharmaceutical solvates, hydrates and
dispersion technique drug only dissolved in the solvent of choice they amorphous forms: a special emphasis on cocrystals, Adv. Drug Deliv. Rev. 117
(2017) 25–46.
selected but not polymer hence improper deposition of drug over the
[11] D.R. Serrano, T. Persoons, D.M. D'Arcy, C. Galiana, M.A. Dea-Ayuela, A.M. Healy,
polymer may possible during the co-evaporation stage [29] that will Modelling and shadowgraph imaging of cocrystal dissolution and assessment of
leads to variations drug distribution that results alterations in bioavail- in vitro antimicrobial activity for sulfadimidine/4-aminosalicylic acid cocrystals,
Eur. J. Pharm. Sci. 89 (2016) 125–136.
ability of final formulations.
[12] C. Almansa, R. Mercè, N. Tesson, J. Farran, J. Tomàs, C.R. Plata-Salamán, Cocrystal of
tramadol hydrochloride–celecoxib (ctc): a novel API–API Co-crystal for the treat-
4. Conclusions ment of pain, Cryst. Growth Des. 17 (2017) 1884–1892.
[13] A. Alhalaweh, W. Kaialy, G. Buckton, H. Gill, A. Nokhodchi, S.P. Velaga, Theophylline
cocrystals prepared by spray drying: physicochemical properties and aerosolization
In summary, from the results obtained it was strongly evidenced performance, AAPS PharmSciTech 14 (2013) 265–276.
that the formed chitosan -telmisartan cocrystals have improved solubil- [14] D.R. Serrano, P. O'Connell, K.J. Paluch, D. Walsh, A.M. Healy, Cocrystal habit engi-
ity and bio availability with certain ratios of drug and polymer used. In neering to improve drug dissolution and alter derived powder properties, J.
Pharm. Pharmacol. 68 (2016) 665–677.
our case the ratios from 1:1 to 1:6 weight ratio of drug to polymer [15] R. Thipparaboina, D. Kumar, R.B. Chavan, N.R. Shastri, Multidrug co- crystals: to-
ratio support for the continual improvement of solubility of the drug, wards the development of effective therapeutic hybrids, Drug Discov. Today 21
while the same on further increasing the polymer will tend to decrease (2016) 481–490.
[16] N. Shan, M.J. Zaworotko, The role of cocrystals in pharmaceutical science, Drug
the solubility hence we chosen the 1:6 as the favourable highest drug Discov. Today 13 (2008) 440–446.
polymer ratio for excellent solubility. From the results of SEM and [17] G.R. Desiraju, Supramolecular synthons in crystal engineering—a new organic syn-
PXRD it was clearly observed the crystals structure of telmisartan was thesis, Angew. Chem. Int. Ed. Engl. 34 (1995) 2311–2327.
[18] S. Mutalik, P. Anuj, K. Manoj, A.N. Usha, Enhancement of dissolution rate and bio-
lost with higher polymer concentration. Further the selected formulation availability of aceclofenac: a chitosan-based solvent change approach, Int. J.
(1:6, TCC-6) increases the in vivo bioavailability of the drug considerably Pharm. 350 (2008) 279–290.
at high level when compared to pure telmisartan (one-fold increase) [19] M. Ganesh, U.J. Jeon, U. Ubaidulla, P. Hemalatha, A. Saravanakumar, P. Mei Mei, H.T.
Jang, Chitosan cocrystals embedded alginate beads for enhancing the solubility and
with better AUC and Cmax. A biodegradable polymer like chitosan are bioavailability of aceclofenac, Int. J. Biol. Macromol. 74 (2015) 310–317.
considered to a safe polymer for formulating pharmaceutical formulation [20] U.J. Ries, G. Mihm, B. Narr, K.M. Hasselbach, H. Wittneben, M. Entzeroth, J.C. Van
due to their bio stability and toxic free nature when compared to other Meel, W. Wienen, 6-Substituted benzimidazoles as new nonpeptide angiotensin II
receptor antagonists: synthesis, biological activity, and structure-activity relation-
cocrystals formulation of telmisartan with citric, gallic, or saccharin like
ships, J. Med. Chem. 36 (1993) 4040–4051.
co-formers. The above listed co-formers have ability to change the pH [21] E. Cagigal, L. Gonza'lez, R.M. Alonso, R.M. Jime'nez, pK(a) determination of angioten-
of microenvironment from where they get absorbed upon disassociated. sin II receptor antagonists (ARA II) by spectrofluorimetry, J. Pharm. Biomed. Anal. 26
Such consequences will not develop with bio degradable polymers. (2001) 477–486.
[22] J. Stangier, S. Capf, W. Roth, Pharmacokinetics of orally and intravenously adminis-
Further the process reported herein is very simple and easy to adopt tered telmisartan in healthy young and elderly volunteers and in hypertensive pa-
for scale up. With this we conclude that the chitosan based cocrystals tients, J. Int. Med. Res. 28 (2000) 149–167.
M. Ganesh et al. / International Journal of Biological Macromolecules 131 (2019) 879–885 885

[23] V. Sekar, V.R. Chellan, Immediate release tablets of telmisartan using super [31] R. Thipparaboina, D. Kumar, R.B. Chavan, N.R. Shastri, Multidrug co- crystals: to-
disintegrant formulation, evaluation and stability studies, Chem. Pharm. Bull. 56 wards the development of effective therapeutic hybrids, Drug Discov. Today 21
(2008) 575–577. (2016) 481–490.
[24] Y. Zhang, T. Jiang, Q. Zhang, S. Wang, Inclusion of telmisartan in mesocellular foam [32] P. Arora, A. Kaur, J. Haneef, R. Chadha, Solubility improvement of telmisartan by co-
nanoparticles: drug loading and release property, Eur. J. Pharm. Biopharm. 76 crystallization with citric acid, Int. J. Pharm. Sci. Res. 8 (2017) 3768–3775.
(2010) 17–23. [33] C. Renu, B. Swati, H. Jamshed, K. Sadhika, M. Sanjay, Cocrystals of Telmisartan:
[25] Y. Zhang, Z. Zhi, T. Jiang, J. Zhang, Z. Wang, S. Wang, Spherical mesoporous silica characterization, structure elucidation, In-vivo, and toxicity studies, Cryst. Eng.
nanoparticles for loading and release of the poorly water-soluble drug telmisartan, Comm. 16 (2014) 8375–8389.
J. Control. Release 145 (2010) 257–263. [34] H. Jamshed, C. Renu, Drug-drug multicomponent solid forms: cocrystal,
[26] J. Patel, G. Kevin, A. Patel, M. Raval, N. Sheth, Design and development of a self- coamorphous and eutectic of three poorly soluble antihypertensive drugs using
nanoemulsifying drug delivery system for telmisartan for oral drug delivery, Int. J. mechanochemical approach, AAPS PharmSciTech 18 (2017) 2279–2290.
Pharm. Educ. 1 (2011) 112–118. [35] M.N.V. Ravi Kumar, R.A.A. Muzzarelli, C. Muzzarelli, H. Sashiwa, A.J. Domb, Chitosan
[27] P. Koilkonda, A.R. Lekkala, C.M.K. Golla, S.R. Ganji, R.B. Konda, Amorphous chemistry and pharmaceutical perspectives, Chem. Rev. 104 (2004) 6017–6084.
Telmisartan , US Patent, 0111417 A1, 2006. [36] J. Shen, Z. Jiao, Z.D. Li, X.J. Shi, M.K. Zhong, HPLC determination of telmisartan in
[28] L. Zhong, X. Zhu, B. Yu, W. Su, Influence of alkalizers on dissolution properties of human plasma and its application to a pharmacokinetic study, Pharmazie 60
telmisartan in solid dispersions prepared by co-grinding, Drug Dev. Ind. Pharm. 40 (2005) 418–420.
(2014) 1660–1669. [37] R.A. Saber, A.K. Attia, W.M. Salem, Thermal analysis study of antihypertensive drugs
[29] Z. Lin, X. Zhu, X.F. Luo, W. Su, Dissolution properties and physical characterization of telmisartan and cilazapril, Adv. Pharm. Bull. 4 (2014) 283–287.
telmisartan–chitosan solid dispersions prepared by mechanochemical activation, [38] N.A. Ahmed, F.N. Viviane, S.E. Safaa, Formulation and physicochemical characteriza-
AAPS PharmSciTech 14 (2013) 541–550. tion of chitosan/acyclovir cocrystals, Pharm. Dev. Technol. 18 (2013) 856–865.
[30] P.H.L. Tran, H.T.T. Tran, B.J. Lee, Modulation of microenvironmental pH and crystal- [39] H.J. Lemmer, J.H. Hamman, Paracellular drug absorption enhancement through tight
linity of ionizable telmisartan using alkalizers in solid dispersions for controlled re- junction modulation, Expert Opin. Drug Deliv. 10 (2013) 103–114.
lease, J. Control. Release 129 (2008) 59–65.

You might also like