ArticlePDF Available

Drug-Polymer Interaction Studies of Cytarabine Loaded Chitosan Nanoparticles

Authors:

Abstract and Figures

Assessment of possible incompatibilities between drug and excipients is an important parameter of preformulation stage during the pharmaceutical product development of active pharmaceutical ingredient (API). The potential physical and chemical interaction among the components of a delivery system can affect the chemical nature, bioavailability, stability, and subsequently therapeutic efficacy of drugs. In this study, ATR-FTIR spectroscopy was employed to investigate the possible intermolecular interaction of Cytarabine with deacetylated chitosan and tripolyphosphate in the resulting physical blends and crosslinked nanoparticulate system. Two different strategies, physical blending and ionotropic gelation, were adopted to prepare binary or tertiary mixtures and nanoparticulate formulation, respectively. The IR spectra of CB showed characteristic peaks at 3438.27 cm-1 (primary amine), 3264.74 cm-1 (hydroxyl group) and 1654.98 cm-1 (C=O stretch in cyclic ring); CS at 3361.47 cm-1 (N-H stretching), 1646.18 cm-1 (C=O of Amide I), 1582.36 cm-1 (C=O of Amide II), and sTPP at 1135.77 cm-1 (P=O). CS-sTPP chemical interaction was confirmed from the shift in the absorption band of carbonyl groups (amide I, II) to 1634.66 cm-1 and 1541.17 cm-1 in blank chitosan nanoparticles, and 1636.87 cm-1, 1543.33 cm-1 in CSNP1 (2:6:1), and at 1646.15 cm-1 and 1557.04 cm-1 in CSNP2 (1:3:1). The characteristic peaks of CB were also present in chitosan formulation with a slight shift in the amino group at 3429.43 cm-1 and 3423.21 cm-1, in the hydroxyl group at 3274.54 cm-1 and 3270.73 cm-1, CSNP1 and CSNP2, respectively. The findings counseled no significant interaction in IR absorption pattern of cytarabine functional groups after encapsulation in CS-sTP
Content may be subject to copyright.
Asadullah Madni et al., J.Chem.Soc.Pak., Vol. 39, No. 06, 2017 1045
Drug-Polymer Interaction Studies of Cytarabine Loaded
Chitosan Nanoparticles
1Asadullah Madni*, 1Prince Muhammad Kashif, 1Imran Nazir, 2Nayab Tahir, 1Mubashar Rehman,
3Muhammad Imran Khan, 1Muhammad Abdur Rahim and 1Abdul Jabar
1Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur (63100), Punjab, Pakistan.
2College of Pharmacy, University of Sargodha, Sargodha (40100), Punjab, Pakistan
3Institute of Pharmacy, Physiology and Pharmacology, University of Agriculture,
Faisalabad (38040), Punjab, Pakistan.
asadullah.madni@iub.edu.pk; asadpharmacist@hotmail.com*
(Received on 7th December 2016, accepted in revised form 7th November 2017)
Summary: Assessment of possible incompatibilities between drug and excipients is an important
parameter of preformulation stage during the pharmaceutical product development of active
pharmaceutical ingredient (API). The potential physical and chemical interaction among the
components of a delivery system can affect the chemical nature, bioavailability, stability, and
subsequently therapeutic efficacy of drugs. In this study, ATR-FTIR spectroscopy was employed to
investigate the possible intermolecular interaction of Cytarabine with deacetylated chitosan and
tripolyphosphate in the resulting physical blends and crosslinked nanoparticulate system. Two
different strategies, physical blending and ionotropic gelation, were adopted to prepare binary or
tertiary mixtures and nanoparticulate formulation, respectively. The IR spectra of CB showed
characteristic peaks at 3438.27 cm-1 (primary amine), 3264.74 cm-1 (hydroxyl group) and 1654.98
cm-1 (C=O stretch in cyclic ring); CS at 3361.47 cm-1 (N-H stretching), 1646.18 cm-1 (C=O of Amide
I), 1582.36 cm-1 (C=O of Amide II), and sTPP at 1135.77 cm-1 (P=O). CS-sTPP chemical interaction
was confirmed from the shift in the absorption band of carbonyl groups (amide I, II) to 1634.66 cm-1
and 1541.17 cm-1 in blank chitosan nanoparticles, and 1636.87 cm-1, 1543.33 cm-1 in CSNP1 (2:6:1),
and at 1646.15 cm-1 and 1557.04 cm-1 in CSNP2 (1:3:1). The characteristic peaks of CB were also
present in chitosan formulation with a slight shift in the amino group at 3429.43 cm-1 and 3423.21
cm-1, in the hydroxyl group at 3274.54 cm-1 and 3270.73 cm-1, CSNP1 and CSNP2, respectively. The
findings counseled no significant interaction in IR absorption pattern of cytarabine functional groups
after encapsulation in CS-sTPP complex, which projected the potential of chitosan nanoparticulate
system to entrap cytarabine.
Keywords: ATR-FTIR spectrometry, Chitosan, Cytarabine, Drug-polymer compatibility, Ionotropic gelation
method, Sodium tripolyphosphate
Introduction
The production of quality medicines is a
fundamental challenge for bulk drug and its suitable
delivery system. The assessment of excipients for
impurities and their chemical compatibility with drug
molecules is inevitable to assure the stability and
effectiveness of a delivery system [1]. In the
preformulation phase, drug-excipient compatibility
study signifies the chemical nature, bioavailability and
therapeutic safety of drug by evaluating the potential
physical and chemical interaction [2] Among the
various techniques, employed for assessment of
possible drug-excipient interactions, such as
differential scanning calorimetry (DSC),
thermogravimetry (TG), hot-stage microscopy (HSM),
X-ray diffractometry (XRD), diffuse reflectance
spectroscopy (DRS), self-interactive chromatography
(SIC), High Pressure Liquid Chromatography (HPLC),
thin layer chromatography (TLC), and liquid
chromatography (LC) with ultraviolet detection [3, 4],
FT-IR is most commonly employed, fast, robust and
less-expensive technique [5].
Since, the dispersive systems were
superseded by the much more powerful FT-IR
(Fourier-Transform-Infrared) spectrometers IR-
spectroscopy has developed into a widely used
routine analytical tool [6] The versatile and green
approach of ATR-FTIR technique has made for direct
analysis of possible physicochemical interactions
between active moiety and formulation components
[7] Furthermore, FTIR spectroscopy provides reliable
evidence of the appearance, modification, or
desertion of attribute peaks of various functional
groups such as C=O, C-H or N-H of the excipients
and drugs among physical mixtures and nanoparticles
formulations [8] This instrument allows measuring
all types of samples, whether they are solid, liquid or
gaseous [9]
In the clinical trial phase, the failure of a huge
number of medicaments to achieve encouraging
therapeutic outcomes is due to the lack of target
specificity, the frequent and large doses, and subsequent
*To whom all correspondence should be addressed.
Asadullah Madni et al., J.Chem.Soc.Pak., Vol. 39, No. 06, 2017 1046
adverse effects [10]. Furthermore, the drastic
physiological condition (gastric pH) and degradation of
biologically active agents hinder the distribution of drug
to a specific disease site [11] Therefore, the interesting
concept of micro and nanoparticles has developed to
overcome these disadvantages as a novel drug delivery
system. Primarily, the incorporation of active
pharmaceutical ingredients in micro/nano-particulate
matrix system is deliberating for their protection from
drastic in-vivo environment [12]. Moreover, the
controlled and sustained liberation of medicinal
substances, and conjugation or surface modification of
nanoparticles with targeting moiety also provides the
potential of treating site-specific release [12]. In these
nanoparticulate systems, a wide range of natural or
synthetic polymers (polysaccharides, polymethacrylates,
polyanhydrates, polyesters/ortho esters, poly lactic-co-
glycolic acid and polyphosphohazanes), lipids,
dendrimers, and surfactants can be employed to integrate
drugs by passive absorption and chemical conjugation
[13].
Low molecular weight chitosan (CS), a natural
cationic and hydrophilic amino-polysaccharide
macromolecule, is being used in the novel delivery
systems because of distinctive biological attributes, such
as muco-adhesion, biodegradation, haemocompatibility,
non-toxicity, and antibacterial property [14]. CS based
nano-particles/fibers, hydrogels and membranes have
been formulated to embed bioactive moieties
(hydrophilic, hydrophobic and macromolecules), due to
the specific tissue targeting and, sustained/controlled
release [15] Ionic gelation technique, among multiple
preparation methods comprising complex coacervation,
ionotropic gelation, self-assembly method through
chemical modification, solvent evaporation/diffusion, and
emulsion-droplet coalescence, is preferred for its
convenience, relative simplicity, and the rid of organic
solvents and high temperatures [16]. Chitosan
nanoparticles (CSNPs), have been widely developed for
the treatment of various conditions. For example,
nanoparticles of 5-fluorouracil, paclitaxel, catechin,
doxorubicin, leucovorin, and pravastatin are used in
cancer therapy. Nanoparticles of various other
therapeutical agents such as, donepezil, rivastigmine,
tacrine, ampicillin, chlorhexidine, levofloxacin, and
vancomycin are utilized to treat Alzheimer disease. In
order to manage Parkinson’s disease, bromocriptine,
dopamine, pramipexole, and selegiline can be
encapsulated in nanoparticulate system. Numerous
antiviral agents (saquinavir, stavudine, tenofovir, and
zidovudine) are now encapsulated into chitosan
nanocarriers in order to combat HIV. Moreover, several
antioxidants (ascorbic acid, rutoside, salvia officinalis,
and satureja montana) can also be loaded in
nanoparticulate system [17].
We envisioned the present compatibility
investigation to determine possible interaction of
Cytarabine (CB) with CS nanoparticle formulation
components. All individual components as polymers and
drug, their physical mixture, and developed
Nanoparticulate formulations have been examined by
ATR-FTIR spectrometer. Based on this observation,
future studies can be planned to evaluate the effects of
formulation parameters on the response variables. The
2D-chemical structure of cytarabine, chitosan, and
sodium tripolyphosphate was prepared on Chemdraw 8.0
Pro CambridgeSoft Corporation, USA (Fig. 1).
Experimental
Materials
Cytarabine (C9H13N3O5, PubChem CID: 6253)
was purchased from Beijing Mesochem Technology Co.,
LTD (China). Chitosan low molecular weight
(C56H103N9O39, PubChem CID: 71853) was purchased
from Sigma-Aldrich Chemie GmbH (USA), Sodium
tripolyphosphate (Na5P3O10, PubChem CID: 24455) from
MSDS ScienceLab.com, Inc. (Texas) and Acetic acid,
glacial 100% (C2H4O2, PubChem CID: 176) Merck
Millipore (USA).
Preparation of Physical Mixtures
Physical blends were prepared by intimate
trituration of equimolar quantities (10 mg) of CB, CS,
sTPP in an agate mortar for about 10 minutes to attain a
homogenized mixture [18]. The abbreviations and
equivalent ratios of resultant samples are described in
Table-1.
Table-1: Chemical composition, equivalent ratio, and sample code for ATR-FTIR analysis.
Sr. No Sample code Chemical composition Equivalent ratio
1 CB Cytarabine 1
2 CS Chitosan 1
3 sTPP Sodium Tripolyphosphate 1
4 CB-CS Cytarabine + Chitosan 1:1
5 CB-sTPP Cytarabine + Sodium Tripolyphosphate 1:1
6 CS-sTPP Chitosan + Sodium Tripolyphosphate 1:1
7 CB-CS-sTPP Cytarabine + Chitosan + Sodium Tripolyphosphate 1:1:1
8 CSNPbChitosan Nanoparticles without Cytarabi ne -
9 CSNP1Chitosan Nanoparticles loaded with Cytarabi ne -
10 CSNP2Chitosan Nanoparticles loaded with Cytarabi ne -
Asadullah Madni et al., J.Chem.Soc.Pak., Vol. 39, No. 06, 2017 1047
Fig. 1: 2D structure of Cytarabine, Chitosan, and Sodium tripolyphosphate (Chemdraw 8.0).
Table-2: Chitosan based nanoparticles with and without the loading of cytarabine.
Formulation code Cytarabi ne(mg) Chitosan(mg) SodiumTripoly-phosphate(mg) Stirring rate (rpm) Stirring time (h)
CSNPb- 60 10 700 2
CSNP120 60 10 700 2
CSNP220 60 20 700 2
Synthesis of Blank Chitosan Nanoparticles
The ionotropic gelation method was
employed to develop CS nanoparticles according to
the method reported by Calvo et al. [19] Briefly, 10
ml sTPP aqueous solution (1mg/ml) added to 20 ml
of 1% aqueous acetic acid solution containing CS (3
mg/ml) to produce blank nanoparticles under
constant stirring (700 rpm) at ambient temperature
(Table-2). The resulted nanoparticle suspension was
ultra-centrifuged using Sigma 1-14 (Sigma
Laborzentrifugen GmbH, Germany), and washed
three times with distilled water to evacuate the
unentrapped drug, and lyophilized in the freeze dryer
(CHRIST Alpha 1-2 LD plus, UK) for 24h [20]. The
interaction (inter- and intramolecular) between the
high degree of positively charged amino groups of
CS and the anionic groups of sTPP resulted from the
formation of CS based NPs [21]. The mechanism of
cross-linking described as illustration in Fig. 2 [22,
23].
Preparation of Chitosan Nanoparticles Loaded with
Cytarabine
Cytarabine loaded chitosan nanoparticles
were fabricated employing ion gelation method
reported by Kalam et al. [24] CB was dissolved into
20 ml of aqueous acetic acidic solution (1%)
containing CS before the addition of 10 ml of sTPP
(0.1%). Thereafter, the formulation was stirred
magnetically at 700 rpm for 2h, at room temperature.
Two formulations (CSNP1 and CSNP2) were
fabricated as mentioned in Table-2.
ATR-FTIR Instrumentation
Attenuated total reflection- Fourier
transform infrared (ATR-FTIR) spectroscopic
technique was employed to analyze
physical/chemical interaction between pure
components and developed formulations [25].
Chemical images of pure drug, polymer, cross linker,
their physical blends, and nanoparticles (with and
without drug) were acquired by the FTIR
spectroscopic imaging approach using ATR-FTIR
spectrometer (Bruker tensor 27, Germany). All
spectral images were collected using OPUS (v6.5)
FT-IR data collection software over a spectral range
of 4000-400 cm-1 with 16 co-added scans at a
resolution of 4 cm-1. The standard sample cell in the
FTIR is a Pike Miracle single-bounce attenuated total
reflectance (ATR) equipped with a ZnSe single
crystal. Prepared samples were placed directly on the
small crystal spot, and the arm rotated over and
turned down to press the sample down onto the
crystal face to get better contact [26].
Asadullah Madni et al., J.Chem.Soc.Pak., Vol. 39, No. 06, 2017 1048
Fig. 2: Schematic mechanism of nanoparticles preparation by crosslinking of chitosan with sodium
tripolyphosphate using ionotropic gelation (Chemdraw 8.0).
Entrapment Efficiency and Drug Release Profile
The entrapment efficiency of CB was
determined by centrifugation method. Nanoparticles
were separated from aqueous environment of
nanoparticulate system by the ultracentrifugation at
14000 rpm for 30 min. The clear superintend was
further diluted 100 times with phosphate buffer saline
(pH 7.4). The unentrapped drug was measured from
diluted solution by UV spectrophotometer at λmax of
CB (272 nm). Furthermore, standard curve was
constructed in PBS solution (pH 7.4) by plotting
graph between CB concentrations of 1 mg/ml to 15
mg/ml and UV absorbance, to calculate the amount
of entrapped drug in nanoparticulate system. The
calibration curve equation (y = 0.0414x - 0.0056) at
= 0.9996 was obtained from MS Excel 2010 and
regressed as standard for the quantitative analysis
[27].
In-vitro drug release profile of Cytarabine
from chitosan nanoparticles was evaluated by dialysis
tube method. The weighed amount of nanoparticulate
system was filled in dialysis tube and was immersed
in 400 ml of PBS at pH 7.4. The temperature of
medium was maintained at 37 ± 0.5 ºC and stirred at
100 rpm in the USP paddle assembly attached with
PTFC-2/8 Fraction Collector auto-sampler (Pharma
Test, Hainburg, Germany). During the 24 h drug
release study, subsequent series of 5 ml aliquots were
pulled back from dialysis solution at predetermined
time intervals (1, 2, 3, 4, 5, 6, 8, 10, 12, 14, 16, 18,
20, 22 and 24 h) and substituted with equal volume of
fresh PBS to sustain sink conditions. Samples were
adequately diluted, filtered and analyzed by UV-
spectrophotometer (IRMACO GmbH, Geesthacht,
Germany) at 272 nm. The released amount of CB
was calculated from the regression equation and
cumulative drug release (%) was calculated.
Results and Discussion
ATR-FTIR is recognized analytical
technique for the analysis of functional group, phase
separation behavior, and different types of bonding
forces in pharmaceutical ingredients. Considering
mid-infrared spectral modifications in the molecular
band strength and frequency shifts are acknowledged
measures for the occurrence and intensity of inter-
intramolecular forces [28]. Generally, pharmaceutical
entities comprise particular bonds and characteristic
functional groups that vibrate independently at their
equilibrium position and weekly interact with each
other, without the influence of any electromagnetic
(EM) radiation effect [29]. After the application of an
infrared (IR) radiation through a sample, the
transitions among vibrational and rotational power
levels result in the alteration of dipole moment,
because of molecular absorption of IR [30].
Moreover, IR absorption is specific to unique
molecular vibration frequency, and the resulting
spectrum epitomizes the molecular
transmission/absorption and generates a precise
fingerprint of sample [31].
Asadullah Madni et al., J.Chem.Soc.Pak., Vol. 39, No. 06, 2017 1049
Fig. 3: FTIR fingerprint of individual components (a) cytarabine, (b) chitosan, and (c) sodium
tripolyphosphate.
Table-3: Functional groups and characteristic peaks of pure ingredients.
Samples Functional Groups with Characteristic Pea ks References
CB N-H (3438.27 cm-1 & 3356.33 cm-1), O-H (3264.74 cm-1), C-H (293 5.62 cm-1), C=O (1654.98 cm-1), N-H (1576.07 c m-1), C-N
(1473.25 cm-1), C-O-H (1279.18 cm-1), and C-O-C (1109.10 cm-1 & 1069.08 cm-1). [5, 32 33]
CS N-H & O-H (3361.47 cm-1), C-H (2878.38 cm-1), O-H (2362.21 cm-1), C=O (1646.18 cm-1 & 1582.36 cm-1), CH 3 (1374.26
cm-1), C-O-C (1149.85 cm-1), C-O (1059.02 cm-1)[6, 35, 40]
STPP P=O (1135.77 cm-1) and P-O (891.19 cm-1 & 734.44 cm-1) [5, 9]
Therefore, the elucidation of structure–
property relations of pure components with their
physical blend, and nanoparticle formulations were
analyzed by ATR-FTIR spectrometer and interpreted
to indicate the possible interactions and
incompatibilities.
FT-IR spectra of pure components
The characteristic bands of cytarabine,
deacetylated chitosan, and sodium tripolyphosphate
are presented in Fig. 3 and the locations of
characteristic band values are given in Table-3. FTIR
spectra of CB, as described in Fig. 3(a), revealed the
characteristic peak of primary amine (N-H) at
3438.27 cm-1 and 3356.27 cm-1 (asymmetric and
symmetric stretch, respectively), broad absorption
peak at 3264.74 cm-1 disclosed the presence of O-H
stretching. The characteristic peak at a wave number
of 2935.62 cm-1 showed C-H asymmetrical stretch;
the sharp peaks at 1654.98 cm-1 and 1576.07 cm-1
indicated C=O stretching (aromatic) and N-H
bending (secondary amide), respectively. C-N
stretching, C-O-H bending and C-O-C stretching
(ether) were also observed at 1473.25 cm-1, 1279.18
cm-1 and 1109.10 cm-1, 1069.08 cm-1 respectively.
Other researchers in their study, indicating the purity
and identity of CB, also reported similar
characteristic peaks of pure cytarabine [32, 33].
In the FTIR fingerprint of CS, as in Fig.
3(b), the overlapped characteristic peak of N-H and
O-H stretching at 3361.47 cm-1, and absorption band
at 2878.38 cm-1 depicted asymmetrical stretching
vibrations of C-H. The further spectral interpretation
demonstrated a broad absorption peak of O-H
(stretching) overlapped with C-H at the wave number
of 2362.21 cm-1 (which occurs in the presence of a
carbonyl group). C=O stretching of amide I at
1646.18 cm-1 and amide II at 1582.36 cm-1 confirmed
Asadullah Madni et al., J.Chem.Soc.Pak., Vol. 39, No. 06, 2017 1050
the presence of N-acetylated groups of CS. The wave
numbers at 1374.26 cm-1, 1149.85 cm-1 and 1059.02
cm-1 and ensured the presence of CH3 (asymmetrical
deformation), C-O-C bridge (asymmetrical
stretching) and C-O (stretching), respectively. Others
also reported all the absorption bands of functional
groups in chitosan [34, 35]. IR spectral analysis of
sTPP, as explained in Fig. 3(c), showed strong P=O
stretching vibrations at 1135.77 cm-1, and P-O
stretching at 891.19 cm-1 and 734.44 cm-1. Similar
characteristic peaks were observed in a study
presented by Lima et al. [36].
FT-IR Spectrum of Physical Blends
Any possible interaction in the physical
mixtures of Cytarabine and formulation excipients
has been examined and depicted in Fig. 4. The binary
blend of drug with polymer (CB-CS), as presented in
Fig. 4(a), confirmed all the identical peaks with
imperceptible shift and indicating the lack of
interaction between functional groups. IR spectra of
cytarabine with a cross-linker, as shown in Fig. 4(b),
depicted no significant shift in characteristic peaks
compared to the individual components (Fig. 3)
demonstrating the absence of interactions. The
incompatibility of chitosan with sodium
tripolyphosphate also studied and the absence of
linkage between –NH2 and –P-OH was observed in
their physical mixture.
Similarly, the IR spectral analysis of drug,
polymer, and cross-linker also confirmed the
presence of all individual attribute peaks in their
physical mixture without any virtual shift, as
illustrated in Fig. 4(d). This part of the investigation
has confirmed the compatibility of the drug with its
formulation components before the development of
nanoparticles. Locations of characteristic band values
are summarized in Table-4.
FT-IR Spectrum of Chitosan Nanoparticles
Fig. 4: FTIR fingerprint of physical blends: (a) CB-CS, (b) CB-sTPP, (c) CS-sTPP, and (d) CB-CS-sTPP.
Asadullah Madni et al., J.Chem.Soc.Pak., Vol. 39, No. 06, 2017 1051
Table-4: Functional groups and characteristic peaks of physical blends.
Physical blends Characteristic bands with functional groups
CB-CS 3435-3263.85 cm-1 (N-H & O-H), 2886.19 cm-1 (C-H), 2359.57 c m-1 (O-H), 1654.63 cm-1 (C=O), 1576.90 cm-1 (N-H), 1473.25 c m-1 (C-
N), 1372.79 cm-1 (-CH3), 1279.57 cm-1 (C-O-H), and 1149-1068.94 cm -1 (C-O-C).
CB-sTPP 3438.16-3264.58 cm-1 (N-H & O-H), 2937. 26 cm-1 (C-H), 1655.20 cm-1 (C=O), 1577.01 cm-1 (N-H), 1474.06 cm-1 (C-N), 1279.57 cm-1
(C-O-H), 1136.98 cm-1 (P=O), 1109.98-1069.48 cm-1 (C-O-C), and 735.37 cm-1 (P-O).
CS-sTPP 3356.32 cm-1 (N-H & O-H), 2871.14 cm-1 (C-H), 2359.91 cm-1 (O-H), 1645.26 cm-1 (C=O), 1583.36 cm-1 (C=O), 1374.81 cm-1 (-CH3),
1138.38 cm-1 (P=O), 1056.92 cm-1 (C-O) and 734.58 cm-1 (P-O).
CB-CS-sTPP 3438-3264.47 cm-1 (N-H & O-H), 2936-2887.69 cm-1 (C-H), 2359.56 cm-1 (O-H), 1654.48 cm-1 (C=O), 1576.32 cm-1 (N-H), 1471.99 cm-
1 (C-N), 1372.43 cm-1 (-CH3), 1279.30 cm-1 (C-O-H), 1137.32 cm-1 (P=O), 1109-1069.18 cm-1 (C-O-C) and 736 cm-1 (P-O).
Fig. 5: FTIR spectra of CS nanoparticles: (a) CSNPb, (b) CSNP1, and (c) CSNP2.
The chitosan-sodium tripolyphosphate
matrix system is resulted due to interaction of amino
group (CS) and phosphate (sTPP) [37], as explained
in Fig. 2. Different chitosan nanoparticulate
formulations have been developed by the ionotropic
gelation method from different molar ratios of
formulation components (Table-2) to scrutinize the
possible interaction and compatibility of components
in the formulation. ATR based FTIR spectrum of
chitosan nanoparticles, as revealed in Fig. 5(a), the
individual chitosan spectra, a broad absorption peak
of the hydroxyl group (O-H) was observed at 3241.12
cm-1 specifying the enhanced hydrogen bonding. A
characteristic peak of O-H stretching (overlay C-H)
was perceived with a negligible shift in the wave
number from 2362.21 cm-1 to 2358.04 cm-1. The
absorption band of C=O of amide I at 1646.18 cm-1
was shifted at 1634.66 cm-1, indicating a possible
interaction with sTPP. Moreover, the bending
vibration of amide II at 1582.36 cm-1 (representing
deacetylated group) was also shifted to 1541.17 cm-1
after the ionic interaction with sTPP. This
conjugation (between ammonium and phosphoric
ions) decreases the solubility of chitosan and
accountable for the separation of CS nanoparticles
from the solution [16].
To compare the possible interaction of drug
with polymer-crosslinker complex, two different
Asadullah Madni et al., J.Chem.Soc.Pak., Vol. 39, No. 06, 2017 1052
formulations (CSNP1 and CSNP2) were developed
with different molar ratios of CB, CS and sTPP
(2:6:1 and 1:3:1). Fig. 5(b) and (c) represents IR
spectra of CB nanoparticles, which disclosed the
notable attenuation of absorption spectra with a
substantial decrease in strength of absorption peaks,
signifying the existence of interaction and entrapment
of CB in CS-sTPP matrix system [38]. The
characteristic peaks of primary amine (N-H)
symmetrical and asymmetrical at 3438.27 cm-1 and
3356.27 cm-1 of CB were shifted to lower
wavenumber in both formulations at 3429.43 cm-1
(CSNP1) and 3423.21 cm-1 (CSNP2) with the absence
of N-H asymmetrical peak, representing the
interaction between CB and chitosan. Small
dierences were noticed at the wavenumber 3264.74
cm-1 (-OH stretching) which were shifted to
wavenumber 3274.54 cm-1 (CSNP1) and 3270.73 cm-
1 (CSNP2), and absorption peak at 2935.62 cm-1 (C-H
asymmetrical stretching) which is shifted to 2920.03
cm-1 (CSNP1) and 2886.15 cm-1 (CSNP2). The
spectral peaks of interaction between CS and sTPP
were also detected in CSNP1 and CSNP2 at 1636.87
cm-1, 1543.33 cm-1 and 1646.15 cm-1, 1557.04 cm-1,
respectively. These are the indicators of CS-sTPP
conjugation and interaction. The C-O-H (bending)
vibration peaks of CB were also observed in the
nanoparticle system at 1423.25 cm-1 and 1421.72 cm-
1 for CSNP1 and CSNP2 respectively, ensuring the
loading of the drug [39]. Moreover, Cytarabine has
three further strong absorption bands at 1279.18 cm-1,
1109.10 cm-1 and 1069.08 cm-1, which were
overlapped with chitosan absorption bands (perceived
at the same position in CSNP formulations).
Furthermore, CB showed physical
interaction with CS in nanoparticles, which may be
due to the electrostatic forces, but no additional
characteristic peak or shifting of bands was observed
in the physical mixtures or nanoparticulate
formulations. This analysis ensures the entrapment of
Cytarabine in biocompatible polymer (chitosan)
matrix and its accessibility to fabricate the drug
delivery system for biological actions.
Entrapment Efficiency and Drug Release Profile
Table-6 describes the encapsulation
efficiency of nanoparticle formulations at constant
drug content. CSNP1 demonstrated high entrapment
efficiency of Cytarabine into nanooparticles was
75.24%, while CSNP2 showed low by 64.41 %.
Formulation CSNP1 showed more sustained
discharge of Cytarabine from nanoparticulate system
whereas drug release from formulation CSNP2 was
less sustained (Table-6). The values of drug release
(%) after 12 hours and 24 hours were 51.65% and
82.84% respectively from CSNP1, and 72.34% and
89.64% from CSNP2 respectively. Nanoparticulate
formulation CSNP1 revealed high EE with sustained
liberation of drug from nanoparticles following 24
hour release period (Fig. 6).
Fig. 6: Drug release behaviour of Cytarabine from
chitosan nanoparticulate system (a) CSNP1,
and (b) CSNP2.
Table-5: Characteristic bands and functional groups of chitosan nanoparticulate system.
Formulations Characteristic bands with functional groups
CSNPb
3241.12 cm-1 (N-H & O-H), 2358.04 cm-1 (C-H), 1634.66 cm -1, 1541.17 cm-1 and 1375.98 cm-1 (ammonium a nd phosphoric ion
conjugation)
CSNP1
3429.43 cm-1 (N-H), 3274.54 cm-1 (O-H), 2920.03 cm-1 (C-H), 2293.28 c m-1 (O-H), 1636.87 cm-1 & 1543.33 cm-1 (ammonium a nd
phosphoric ion conjugation) and 1423.25 cm-1 (C-O-H).
CSNP2
3423.21 cm-1 (N-H), 3270.73 cm-1 (O-H), 2886.15 cm-1 (C-H), 2282.36 c m-1 (O-H), 1646.15 cm-1 (C=O of amide I), 1557.04 cm-1
(ammonium and phosphoric ion conjugation) a nd 1421.72 cm-1 (C-O-H)
Table-6: Entrapment efficiency and drug release behaviour of CSNP1 and CSNP2.
Formulation code Entrapment Efficiency (%) Drug release after 12hr (%) Drug release after 24hr (%)
CSNP175.24 51.65 82.84
CSNP264.41 72.34 89.64
Conclusion Chitosan nanoparticle formulations alone and
Asadullah Madni et al., J.Chem.Soc.Pak., Vol. 39, No. 06, 2017 1053
in combination with cytarabine were successfully
prepared using the ionic gelation technique. Among the
pre- and post-formulations studies, ATR-FTIR has
appeared as a fundamental technique to find out
interactions among the formulation components and
compatibility to be included in a drug delivery system.
The findings from this study have revealed the absence
of possible chemical interaction or incompatibility
between Cytarabine and chitosan-tripolyphosphate
matrix system. Physical state of the cytarabine was
modified, which corresponds to the drug crystallinity
whereas formulation is thrashing its sharpness at its
characteristic peaks. Absence of detectable crystalline
domains in cytarabine-loaded nanoparticulate clearly
indicates that drug remained intact and dispersed
completely in the formulation, thus modifying the
nanoparticles to a disordered-crystalline phase. The
analysis of various physical mixtures revealed a slight
shift in band/peaks of different functional groups and
characteristic peaks of cytarabine were observed at the
same wavenumber and intensity points. However, ATR-
FTIR spectra of the nanoparticulate formulations
revealed the successful entrapment of cytarabine in CS
nanoparticles. Furthermore, the compatibility
investigations of cytarabine with excipients pretended
the absence of precarious substances/excipients.
Therefore, it is established that cytarabine can be
successfully entrapped the CS nanoparticulate
formulations and provide guide for future compatibility
assessment of CB.
References
1. Y. Wu, J. Levons, A. S. Narang, K. Raghavan and
V. M. Rao, Reactive Impurities in Excipients:
Profiling, Identification and Mitigation of Drug–
Excipient Incompatibility, AAPS PharmSciTech,
12, 1248 (2011).
2. B. Tiţa, A. Fuliaş, G. Bandur, E. Marian and D.
Tiţa, Compatibility Study Between Ketoprofen and
Pharmaceutical Excipients Used in Solid Dosage
Forms, J. Pharm. Biomed. Anal., 56, 221 (2011).
3. M. Köllmer, C. Popescu, P. Manda, L. Zhou, and
R. A. Gemeinhart, Stability of Benzocaine
Formulated in Commercial Oral Disintegrating
Tablet Platforms, AAPS PharmSciTech, 14, 1333
(2013).
4. Í. P. de Barros Lima, N. G. P. B. Lima, D. M. C.
Barros, T. S. Oliveira, C. M. S. Mendonça, E. G.
Barbosa, F. N. Raffin, T. F. A. d. Lima e Moura, A.
P. B. Gomes, M. Ferrari, and C. F. S. Aragão,
Compatibility Study Between Hydroquinone and
the Excipients used in Semi-Solid Pharmaceutical
Forms by Thermal and Non-Thermal Techniques,
J. Therm. Anal. Calorim., 120, 719 (2015).
5. D. L. Pavia, G. M. Lampman, G. S. Kriz, and J. A.
Vyvyan, Introduction to Spectroscopy, Cengage
Learning, Stamford, USA, p. 14 (2014).
6. P. R. Griffiths and J. A. De Haseth, Fourier
Transform Infrared Spectrometry, John Wiley &
Sons, Hoboken, New Jersey, (2007).
7. H. K. Stulzer, P. O. Rodrigues, T. M. Cardoso, J. S.
R. Matos, and M. A. S. Silva, Compatibility
Studies Between Captopril and Pharmaceutical
Excipients Used in Tablets Formulations, J. Therm.
Anal. Calorim., 91, 323 (2008).
8. K. Pramod, C. V. Suneesh, S. Shanavas, S. H.
Ansari, and J. Ali, Unveiling the Compatibility of
Eugenol with Formulation Excipients by
Systematic Drug-Excipient Compatibility Studies,
J. Anal. Sci. Technol., 6, 34 (2015).
9. B. C. Smith, Fundamentals of Fourier Transform
Infrared Spectroscopy, CRC press, United States of
America, (2011).
10. G. P. Andrews, T. P. Laverty and D. S. Jones,
Mucoadhesive Polymeric Platforms for Controlled
Drug Delivery, Eur. J. Pharm. Biopharm., 71, 505
(2009).
11. A. des Rieux, V. Fievez, M. Garinot, Y. J.
Schneider, and V. Préat, Nanoparticles as Potential
Oral Delivery Systems of Proteins and Vaccines: A
Mechanistic Approach, J. Control. Release, 116, 1
(2006).
12. A. Kumari, S. K. Yadav, and S. C. Yadav,
Biodegradable Polymeric Nanoparticles Based
Drug Delivery Systems, Colloids and Surfaces B:
Biointerfaces, 75, 1 (2010).
13. Z. Liu, Y. Jiao, Y. Wang, C. Zhou, and Z. Zhang,
Polysaccharides-Based Nanoparticles as Drug
Delivery Systems, Adv. Drug Deliv. Rev., 60, 1650
(2008).
14. A. Rampino, M. Borgogna, P. Blasi, B. Bellich,
and A. Cesàro, Chitosan Nanoparticles:
Preparation, Size Evolution and Stability, Int. J.
Pharm., 455, 219 (2013).
15. J. Carneiro, J. Tedim, and M. G. S. Ferreira,
Chitosan as a Smart Coating for Corrosion
Protection of Aluminum Alloy 2024: A Review,
Prog. Org. Coat., 89, 348 (2015).
16. A. Grenha, Chitosan Nanoparticles: a Survey of
Preparation Methods, J. Drug Target., 20, 291
(2012).
17. L. Bugnicourt and C. Ladavière, Interests of
Chitosan Nanoparticles Ionically Cross-Linked
with Tripolyphosphate for Biomedical
Applications, Prog. Polym. Sci., 60, 1 (2016).
18. G. Zingone and F. Rubessa, Preformulation Study
of the Inclusion Complex Warfarin-β-cyclodextrin,
Int. J. Pharm., 291, 3 (2005).
19. P. Calvo, C. Remuñan-López, J. L. Vila-Jato and
M. J. Alonso, Chitosan and Chitosan/Ethylene
Asadullah Madni et al., J.Chem.Soc.Pak., Vol. 39, No. 06, 2017 1054
Oxide-Propylene Oxide Block Copolymer
Nanoparticles as Novel Carriers for Proteins and
Vaccines, Pharm. Res., 14, 1431 (1997).
20. S. Anandhakumar, G. Krishnamoorthy, K. M.
Ramkumar and A. M. Raichur, Preparation of
Collagen Peptide Functionalized Chitosan
Nanoparticles by Ionic Gelation Method: An
Effective Carrier System for Encapsulation and
Release of Doxorubicin for Cancer Drug Delivery,
Mater. Sci. Eng., C, 70, 378 (2017).
21. A. M. M. Sadeghi, F. A. Dorkoosh, M. R. Avadi, P.
Saadat, M. Rafiee-Tehrani, and H. E. Junginger,
Preparation, Characterization and Antibacterial
Activities of Chitosan, N-trimethyl chitosan (TMC)
and N-diethylmethyl chitosan (DEMC)
Nanoparticles Loaded with Insulin using both the
Ionotropic Gelation and Polyelectrolyte
Complexation Methods, Int. J. Pharm., 355, 299
(2008).
22. J. Guan, P. Cheng, S. J. Huang, J. M. Wu, Z. H. Li,
X. D. You, L. M. Hao, Y. Guo, R. X. Li, and H.
Zhang, Optimized Preparation of Levofloxacin-
Loaded Chitosan Nanoparticles by Ionotropic
Gelation, Physics Procedia, 22, 163 (2011).
23. K. G. Desai, Chitosan Nanoparticles Prepared by
Ionotropic Gelation: An Overview of Recent
Advances, Crit. Rev. Ther. Drug Carrier Syst., 33,
107 (2016).
24. M. Abul Kalam, A. A. Khan, S. Khan, A. Almalik,
and A. Alshamsan, Optimizing Indomethacin-
Loaded Chitosan Nanoparticle Size, Encapsulation,
and Release Using Box–Behnken Experimental
Design, Int. J. Biol. Macromol., 87, 329 (2016).
25. A. Aina, M. D. Hargreaves, P. Matousek, and J. C.
Burley, Transmission Raman Spectroscopy as a
Tool for Quantifying Polymorphic Content of
Pharmaceutical Formulations, Analyst, 135, 2328
(2010).
26. A. V. Ewing, P. S. Wray, G. S. Clarke and S. G.
Kazarian, Evaluating Drug Delivery with Salt
Formation: Drug Disproportionation Studied in
Situ by ATR-FTIR Imaging and Raman Mapping,
J. Pharm. Biomed. Anal., 111, 248 (2015).
27. K. A. Janes, M. P. Fresneau, A. Marazuela, A.
Fabra, and M. A. J. Alonso, Chitosan Nanoparticles
as Delivery Systems for Doxorubicin, J. Control.
Release, 73, 255 (2001).
28. D. P. Queiroz, M. N. de Pinho and C. Dias, ATR-
FTIR Studies of poly (propylene
oxide)/polybutadiene Bi-Soft Segment
Urethane/Urea Membranes, Macromolecules, 36,
4195 (2003).
29. J. Liu, D. Ma and Z. Li, FTIR Studies on the
Compatibility of Hard–Soft Segments for
Polyurethane–Imide Copolymers with Different
Soft Segments, Eur. Polym. J., 38, 661 (2002).
30. P. Yu, J. J. McKinnon, C. R. Christensen and D. A.
Christensen, Imaging Molecular Chemistry of
Pioneer Corn, J. Agric. Food Chem., 52, 7345
(2004).
31. P. Yu, J. J. McKinnon, C. R. Christensen and D. A.
Christensen, Using Synchrotron-Based FTIR
Microspectroscopy To Reveal Chemical Features
of Feather Protein Secondary Structure:
Comparison with Other Feed Protein Sources, J.
Agric. Food Chem., 52, 7353 (2004).
32. K. Singh, H. Kaur, and S. Kumar, Design and
Development of Sustained Release Injectable in
Situ Gel of Cytarabine, Pharmacophore, 4, 252
(2013).
33. P. Sharma, B. Dube and K. Sawant, Synthesis of
Cytarabine Lipid Drug Conjugate for Treatment of
Meningeal Leukemia: Development,
Characterization and In Vitro Cell Line Studies, J.
Biomed. Nanotechnol., 8, 928 (2012).
34. A. B. Vino, P. Ramasamy, V. Shanmugam and A.
Shanmugam, Extraction, Characterization and In
Vitro Antioxidative Potential of Chitosan and
Sulfated Chitosan from Cuttlebone of Sepia
aculeata Orbigny, 1848, Asian Pac. J. Trop. Med.,
2, S334 (2012).
35. C. Song, H. Yu, M. Zhang, Y. Yang and G. Zhang,
Physicochemical Properties and Antioxidant
Activity of Chitosan from the Blowfly Chrysomya
Megacephala larvae, Int. J. Biol. Macromol., 60,
347 (2013).
36. H. A. Lima, F. M. V. c. Lia, and S. Ramdayal,
Preparation and Characterization of Chitosan-
Insulin-Tripolyphosphate Membrane for Controlled
Drug Release: Effect of Cross Linking Agent, J.
Biomater. Nanobiotechnol., 5, 211 (2014).
37. Y. Wu, W. Yang, C. Wang, J. Hu, and S. Fu,
Chitosan Nanoparticles as a Novel Delivery System
for Ammonium Glycyrrhizinate, Int. J. Pharm.,
295, 235 (2005).
38. S. Papadimitriou, D. Bikiaris, K. Avgoustakis, E.
Karavas, and M. Georgarakis, Chitosan
Nanoparticles Loaded with Dorzolamide and
Pramipexole, Carbohydr. Polym., 73, 44 (2008).
39. G. Unsoy, R. Khodadust, S. Yalcin, P. Mutlu and
U. Gunduz, Synthesis of Doxorubicin Loaded
Magnetic Chitosan Nanoparticles for pH
Responsive Targeted Drug Delivery, Eur. J.
Pharm. Sci., 62, 243 (2014).
40. M. Fernandes Queiroz, K. Melo, D. Sabry, G.
Sassaki, and H. Rocha, Does the Use of Chitosan
Contribute to Oxalate Kidney Stone Formation?,
Mar. Drugs, 13, 141 (2015).
... Briefly, the formation occurred from the reaction between the amino group of TMSPM-grafted chitosan and the phosphate group in TPP through ionic gelation. 20 Morphology and particle size. A TEM image of TMSPM-grafted chitosan nanoparticles ( Figure 3) prepared via the ionic gelation method with TPP revealed a spherical structure of the nanoparticles with average size of 10.01 ± 1.15 nm. ...
Article
Full-text available
This research investigated and developed a laminate packaging film of poly(lactic acid) (PLA) and poly(vinyl alcohol) (PVA) containing modified chitosan nanoparticles (MCSNPs) treated with 3-(trimethoxysilyl)propyl methacrylate silane (TMSPM) which functioned both as a reinforcement and as an antimicrobial filler. Electron beam irradiation caused crosslink between the PVA and MCSNP molecules via the free radical of carbonyl in the methacrylate group of TMSPM-grafted chitosan nanoparticles and the free radical of hydroxyl group in poly(vinyl alcohol). This improved the PVA hydrophobicity. In addition, the laminate with PVA-g-MCSNPs film irradiated with a 30 kGy electron beam showed the best performance properties due to improved interfacial adhesion between PLA and PVA-g-MCSNPs. As a consequence, the mechanical and thermal properties improved. The 30 kGy irradiated film also had better antibacterial activity against both Escherichia coli and Staphylococcus aureus.
... Supplementary Materials Figure S-2 shows the ATR-FTIR spectra of the raw materials (PRED, both grades of chitosan, and mannitol). Peak characteristics to PRED were seen at 1612 cm −1 , 1652 cm −1 , and 1710 cm −1 , which likely correspond to C = O stretching of the three carbonyl groups of PRED; furthermore, a broad peak was seen at 3354 cm −1 , with two subsequent peaks at 3455 cm −1 , and 3499 cm −1 , which likely correspond to the OH stretching peaks of the three OH groups of PRED, as corroborated by previous studies on the characterization of PRED by FTIR [27][28][29]. Peak characteristics to chitosan were seen at 3361 cm −1 , 1646 cm −1 , and 1582 cm −1 , the former of which likely corresponds to overlapped OH and NH stretching, with the two subsequent peaks corresponding to the C = O stretching of the two amino groups of chitosan [30,31]. Mannitol was characterized by a complex primary peak constituting secondary peaks at 3392 cm −1 , 3340 cm −1 , 3282 cm −1 , and 3234 cm −1 , likely corresponding to the OH stretching groups of mannitol [32,33]. ...
Article
The present work explores a data mining approach to study the fabrication of prednisolone-loaded chitosan nanoparticles and their properties. Eight PLC formulations were prepared using an automated adaptation of the antisolvent precipitation method. The PLCs were characterized using dynamic light scattering, infrared spectroscopy, and drug release studies. Results showed that that the effective diameter, loading capacity, encapsulation efficiency, zeta potential, and polydispersity of the PLCs were influenced by the concentration and molecular weight of chitosan. The drug release studies showed that PLCs exhibited significant dissolution enhancement compared to pure prednisolone crystals. Principal components analysis and partial least squares regression were applied to the infrared spectra and the DLS data to extract higher-order interactions and correlations between the critical quality attributes and the diameter of the PLCs. Principal components revealed that the spectra clustered according to the type of material, with PLCs forming a separate cluster from the raw materials and the physical mix. PLS was successful in predicting the ED of the PLCs from the FTIR spectra with R 2 = 0.98 and RMSE = 27.18. The present work demonstrates that data mining techniques can be useful tools for obtaining deeper insights into the fabrication and properties of PLCs, and for optimizing their quality and performance. It also suggests that FTIR spectroscopy can be a rapid and non-destructive method for predicting the ED of PLCs.
... Similarly, the corresponding peaks of all the ingredients were found wellpreserved in their physical mixture and RLX-PNPs. Exact peaks in particular FTIR region revealed the occurrence for amide-I of CTX [69]. DSC analysis was performed to check the thermal behavior of the PNPs and its components compared with physical mixture. ...
Article
Full-text available
Osteoporosis (OP), is a systemic bone disorder associated with low bone mass and bone tissue corrosion. Worsening of the disease condition leads to bone delicacy and fracture. Various drugs are available for the treatment of OP, however they have limitations including poor solubility, bioavailability and toxicity. Herein, Raloxifene-loaded polymeric nanoparticles (RLX-PNPs) were developed and investigated for the treatment of OP with possible solutions to the above mentioned problems. RLX-PNPs were prepared by modified ionic gelation method followed by determining their particle properties. FTIR, DSC and PXRD analysis of the RLX-PNPs were performed to check chemical interaction, thermal behavior and crystallinity, respectively. In-vitro release profile of RLX-PNPs was checked in lab setting, whereas its pharmacokinetics was investigated in Sprague-Dawley rats, in-vivo. Finally, the treatment potential of RLX-PNPs was analyzed in OP induced animal model. The optimized PNPs formulation indicated 134.5 nm particle size, +24.4 mV charge and 91.73% % EE. TEM analysis showed spherical and uniform sized particles with no interactions observed in FTIR analysis. In-vitro release of RLX from RLX-PNPs showed more sustained release behavior as compared to RLX-suspension. Moreover, pharmacokinetic investigations showed a significantly enhanced bioavailability of the RLX-PNPs as well as reduced serum levels of alkaline phosphatase and calcium in OP induced rats when compared with RLX-Suspension after oral administration. Findings of this study suggested that the developed RLX-PNPs have the potential to treat OP due to sustained release and improved bioavailability of the incorporated drug.
... The mobile phase used was a mixture of acetonitrile and water (50:50) flowing at 1.2 mL/min rate. A run time of 15 min was selected and cytarabine was detected at λ max 272 nm [47,48]. ...
Article
Full-text available
In the present study, pH-sensitive, biodegradable, and biocompatible Na-CMC/pectin poly(methacrylic acid) hydrogels were synthesized using an aqueous free radical polymerization technique and encapsulated by cytarabine (anti-cancer drug). The aim of the project was to sustain the plasma profile of cytarabine through oral administration. Sodium carboxymethyl cellulose (Na-CMC) and pectin were cross-linked chemically with methacrylic acid (MAA) as a monomer, using methylene bisacrylamide (MBA) as cross-linker and ammonium per sulfate (APS) as an initiator. Prepared hydrogel formulations were characterized for their texture, morphology, cytarabine loading efficiency, compositional and structural properties, thermal nature, stability, swelling response, drug release profile (pH 1.2 and pH 7.4), and in-vivo pharmacokinetic evaluation. Cytarabine-loaded hydrogels were also evaluated for their safety profile by carrying out toxicity studies in rabbits. Results demonstrated efficient encapsulation of cytarabine into the prepared network with loading ranging from 48.5-82.3%. The highest swelling ratio of 39.38 and maximum drug release of 83.29-85.27% were observed at pH 7.4, highlighting the pH responsiveness of the grafted system. Furthermore, cytarabine maximum release was noticed over 24 h, ensuring a sustained release response for all formulations. Histopathological studies and hemolytic profiles confirmed that the prepared hydrogel system was safe, biocompatible, and non-irritant, showing no symptoms of any toxicities and degeneration in organs. Moreover, pharmacokinetic estimation of the cytarabine-loaded hydrogel showed a remarkable increase in the plasma half-life from 4.44 h to 9.24 h and AUC from 22.06 µg/mL.h to 56.94 µg/mL.h. This study revealed that the prepared hydrogel carrier system has excellent abilities in delivering the therapeutic moieties in a controlled manner.
... The particle size of nano-chitosan was then measured by a particle size analyzer (PSA). Fig. 5 shows the schematic reaction of chitosan and sodium tripolyphosphate (Madni et al. 2017). Fig. 6 shows the particle size of nano-chitosan at the various ratio of chitosan to Na-TPP solution at the rotation speed of 700 rpm. ...
Article
Full-text available
Nano-chitosan can be produced from blood clams (Anadara granosa) because they contain 14-35% of chitin. The production of nano-chitosan can be conducted by a bottom-up process using sodium tripolyphosphate (Na-TPP). The aims of this study are to produce nano-chitosan from blood clamshell and to study the factor affecting the particle size of nano-chitosan such as the ratio of chitosan to Na-TPP solution (v/v) and rotation speed of the centrifuge. The research shows that The chitin content on blood clamshell is 25.42%. The yield of chitosan from chitin is about 80.92%. The degree of deacetylation of chitin from blood clamshell reaches 63.18%. The effect of the ratio of chitosan to Na-TPP solution (R) and the rotation speed of centrifuge (N) on the particle size of nano-chitosan can be expressed by equation dp = 0.12 (R)0.714 (N)0.99.
... The presence of the independent peaks of ZIF-8 and cytarabine [44] (Table S1) with the decrease in the intensity of adsorption bands that can be seen in ZIF-8 ? CYT spectra further confirms the interaction of cytarabine with ZIF-8. ...
Article
Full-text available
Metal–organic frameworks (MOFs) possess wide array in biomedicine including drug storage and delivery application. In this work, we have synthesized ZIF-8 via room temperature method and later on performed simulation studies to give an integrated understanding of the drug and its interactions with the ZIF-8. The experimental parameters such as metal/ligand molar ratio, temperature, pH, time and stirring speed were optimized, and the material synthesized was characterized for its morphology, porosity and surface area using various analytical techniques. The in vitro drug release studies were conducted at different pH conditions, and diffusion-based release kinetics was observed. Grand Canonical Monte Carlo simulation was also performed to investigate the adsorption behavior of cytarabine (anti-cancer drug molecule) drug onto MOF framework on molecular scale. The simulation studies assist in interpreting the adsorption behavior of cytarabine on ZIF-8 in terms of loading percentage, adsorption isotherms and accessible surface area with energy interactions depicting the strong or weak binding interactions between drug molecule and ZIF-8 surface. The adsorption isotherms and isosteric heat of adsorption were calculated for the configurational ensembles so generated and compared to physical measurements done through adsorption experiments. The results unveil that the integrated experimental and computational study not only accurately recognizes the adsorption mechanism and energy/binding interaction of drug molecules on MOF but also directs the experiments for design and development of novel working pairs of metal–organic ligand drug carriers. Graphical abstract
... The peak at 1496 cm À1 on the SLN-cyt and cytarabine lines indicates C-N bonds of the aromatic rings in cytarabine confirming the presence of the drug within the SLN. The ether bond stretch (C-O-C) of cytarabine was observed 1352 cm À1 (AsadullahMadni et al. 2017). At 1082 cm À1 , a C-O stretching band is present on the SLN and cytarabine combination ...
Article
Full-text available
Aim: To synthesize cytarabine loaded SLNs modified with the RGD peptide as a ligand, suitable for effective cancer therapy. Methods: SLNs were synthesized by the high shear, hot homogenization technique. A 2 level 3 factor analysis was used in optimization. Particle size, zeta potential, poly-dispersion index and surface morphology were measured. Drug encapsulation, drug release, release kinetics, nanoparticle stability and chemical structure were determined. LIVE/DEAD® Fluorescence Assay was used to qualify cytotoxicity and Tryphan Blue assay to quantify. Results: Cyt-SLNs exhibited a size of 161 ± 2.25nm, a PDI of 0.49 ± 0.15 and zeta potential of -19.8mV. Entrapment fell at 88.87 ± 0.02% and release at 83.5 ± 0.95%. The in vitro release kinetics pointed towards a diffusion based drug release mechanism. SLNs remained stable for 60 days. Cytotoxicity studies revealed that conjugation of the ligand with the RDG peptide resulted in a significant decrease in cell viability in both cell lines. Conclusion: Overall, the study suggests that RGD-SLN-cyt can be used for effective cancer therapy.
Article
In this study, sodium diphosphate tetrabasic was utilized to modify the chitosan magnetic nanoparticles. The product was then employed for removal of the malachite green dye in water. Co-precipitation method was used to create the chitosan magnetic sodium diphosphate tetrabasic nanoadsorbent. Scanning electron microscopy (SEM), Fourier transform infrared (FTIR) spectroscopy, field scanning electron microscopy (FE-SEM) and vibrating sample magnetometer (VSM) were used to characterize the synthesized magnetic nanoadsorbent. In order to find the ideal circumstances for the adsorption process, it was then researched how several parameters, such as pH, initial concentration of malachite green and the adsorbent’s dosage, affect adsorption performance. The prepared chitosan magnetic nanoadsorbent with sodium diphosphate tetrabasic modification revealed semi-crystalline nanocomposite on both (FE-SEM) and (SEM). The effective functional groups that were confirmed on FTIR to have contributed to the adsorption of malachite green include chitosan amino group (NH2). Phosphate group (PO4) of tetrabasic sodium diphosphate serves as additional active sites for adsorption. Hydroxyl groups (OH) of chitosan and magnetic nanoparticles are other effective functional groups for MG adsorption. These hydroxyl groups form hydrogen bonds with malachite green and contribute to adsorption. The chitosan nanoparticles exhibited super-paramagnetic characteristics with attestable coercivity levels (Hc) of 15 extendable up to 20 (Oe). The removal efficiency of about 99% malachite green dye (MG) was shown by the prepared nanoadsorbent. Langmuir and Freundlich isotherm models gave successful prediction regarding adsorption efficiency using the malachite green. The findings demonstrated that the adsorption pattern was properly explained by both the Freundlich (R2 = 0.902) and Langmuir (R2 = 0.994) models with Langmuir model (R2 = 0.994) being most pronounced. The Langmuir model was used to achieve the greatest capacity for malachite green retention of 3.94 mg g−1. The kinetic experiments simulated by second-order kinetic model R2 (0.990) fitted better adsorption than first-order kinetics R2 (0.910). The desorption studies to regenerate the nanoadsorbent after eluting the malachite were based on increase in polarity. Based polarity water was confirmed to be the most prominent desorbing agent for malachite green (ethanol < acetone < methanol < water).
Article
Topical drug delivery against cutaneous leishmaniasis (CL) serves as an effective alternate approach to improve efficacy and reduce systemic adverse effects. The purpose of this study was to develop and investigate the potential of rifampicin and pentamidine co-loaded polymeric nanoparticles (RIF-PTM-PNP) for targeting the Leishmania infected macrophages and its effective antileishmanial therapy. RIF-PTM-PNP were prepared by modified ionic gelation method, trailed by their incorporation into 1% Carbopol gel to prepare RIF-PTM-PNP-Gel. The gel was formulated to ensure the retention of PNP on skin for extended period of time. Formulations were statistically optimized via Box-Behnken design (BBD) based on their particle size (P.S), zeta potential (Z.P) and percent entrapment efficiency (%EE). The optimized RIF-PTM-PNP formulation was further assessed for surface morphology and components compatibility via transmission electron microscopy (TEM) and Fourier-transform infrared spectroscopy (FTIR), respectively. Similarly, RIF-PTM-PNP-Gel was evaluated for physiochemical and rheological attributes. In-vitro drug release profile of RIF-PTM-PNP was determined and compared with RIF-Solution, PTM-Solution and RIF-PTM-PNP-Gel. Moreover, ex-vivo permeation analysis of all the test groups were performed to check their permeation profiles. The in-vitro antileishmanial assay, ex-vivo cell uptake, cell viability and cell toxicity studies of the developed formulations were also accomplished. The optimized formulation indicated suitable particle properties including, 163.1 nm P.S, 0.296 PDI, +29.7 mV Z.P and drug entrapment of RIF (89.10%) and PTM (79.31%), respectively. FTIR analysis showed no interactions between components of RIF-PTM-PNP. Moreover, RIF-PTM-PNP-Gel was homogeneous, has optimal pH with desirable viscosity. The in-vitro release of RIF and PTM from RIF-PTM-PNP and RIF-PTM-PNP-Gel showed more sustained release behavior as compared to RIF and PTM solutions. Similarly, significantly higher skin penetration was observed in case of RIF-PTM-PNP and RIF-PTM-PNP-Gel, when compared with conventional gel. Additionally, RIF-PTM-PNP showed superior percent inhibition, higher macrophage uptake and lower IC50 value against promastigotes in association with drugs solutions. Furthermore, in-vivo skin irritation and histopathology study indicated a safe and non-irritant behavior of the RIF-PTM-PNP-Gel, when compared with 0.8% formalin. Also, the optimized formulation (RIF-PTM-PNP) was found stable for at least 6 months. These findings suggested the successful preparation of co-loaded RIF-PTM-PNP and their incorporation into Carbopol-based RIF-PTM-PNP-Gel for topical administration with sustained drug release, enhanced skin permeation, macrophage uptake and improved antileishmanial effect.
Article
Full-text available
The drug-excipient compatibility studies were carried out with the possible excipients viz. Tween 80, Carbopol 940, chitosan, sodium alginate, and polycaprolactone (PCL) for their possible use in the formulation of eugenol loaded nanoemulsion gels and nanoparticles. The eugenol-excipient compatibility studies were carried out by visual observations, differential scanning calorimetry (DSC), infrared spectroscopy (FTIR), and high-performance thin-layer chromatography (HPTLC). No notable change was observed in the samples on visual observation. From the results of the DSC studies, to a much extent, it was assumed that eugenol was not subjected to any interaction with the selected excipients for the proposed nanoemulsion gel and nanoparticles. But in some cases viz. polycaprolactone and Carbopol, though not to a significant level, slight deviation of the nature and position of the endothermic peaks of eugenol were observed. The results of the IR spectroscopy confirmed the compatibility of eugenol with Tween 80, Carbopol 940, chitosan, sodium alginate, and polycaprolactone. The retention factor of the HPTLC densitogram peaks for all the physical mixtures was well within the retention factor (R f ) value range observed for pure eugenol. The study results confirmed that eugenol is compatible with the selected excipients for the development of nanoemulsion gels and nanoparticles.
Article
Full-text available
Thermal techniques, such as differential scanning calorimetry (DSC), thermogravimetry (TG), derivate of TG curve, differential thermal analysis, and non-thermal techniques such as fourier transform infrared (FTIR) spectroscopy and X-ray diffractometry (XRD) were used to evaluate the possible interactions between hydroquinone (HQ) and excipients commonly used in semi-solid pharmaceutical forms. The DSC curve of HQ showed a sharp endothermic event between 173 and 179 °C indicating melting point. No evidence of interaction was observed between HQ and cetyl alcohol (CA), cetostearyl alcohol (CTA), disodium ethylenediaminetetraacetate, and decyl oleate. However, based on the thermoanalytical trials, a physical interaction was suspected between HQ and dipropylene glycol (DPG), glycerin (GLY), hydroxypropyl methylcellulose (HPMC), imidazolidinyl urea (IMD), methylparaben (MTP), and propylparaben (PPP). The FTIR results show that for DPG, GLY, HPMC, MTP, and PPP, there were no chemical interactions with HQ at room temperature, but the heating promotes interaction between HQ and HPMC. The FTIR spectra of HQ/IMD show the chemical interaction at room temperature, which was also observed with heating. The XRD results of mixtures between HQ and DPG, HPMC, IMD, MTP, and PPP indicate no interaction between these substances at room temperature, but the heating modifies the HQ crystallinity in these mixtures. All of these methods showed incompatibility between HQ and the excipient IMD.
Article
Captopril (CAP) was the first commercially available angiotensine-converting enzyme (ACE) inhibitor. In the anti-hypertensive therapy is considered the selected drug has to be therapeutically effective together with reduced toxicity. CAP is an antihypertensive drug currently being administered in tablet form. In order to investigate the possible interactions between CAP and excipients in tablets formulations, differential scanning calorimetry (DSC) and thermogravimetric (TG) analysis completed by X-ray powder diffraction (XRPD) and Fourier transform infrared spectroscopy (FTIR) were used for compatibility studies. A possible drug-excipient interaction was observed with magnesium stearate by DSC technique.
Article
In recent years, nanoparticles (NPs) based on biopolymers or peptides are gaining popularity for the encapsulation and release of drug molecules, especially for cancer therapy, due to their ability for targeted and controlled release. The use of collagen peptide (CP) for the preparation of chitosan (CN) NPs is especially interesting as it results in NPs that are stable under physiological conditions. In this work, mono-dispersed pH responsive CPCN NPs of about 100 nm were prepared via ionic gelation method by simple and mild co-precipitation of CN and CP. Investigation of NPs with Fourier transform infra-red (FTIR) spectroscopyand dynamic light scattering (DLS) measurements reveals that hydrogen bonding and electrostatic interactions are believed to be major driving forces for NP formation and drug encapsulation, respectively. Scanning electron microscopic (SEM) investigations show that hard and fine CPCN NPs transform to soft and bigger gel like particles as a function of collagen concentration. The unique “polymeric gel” structure of NPs showed high encapsulation efficiency towards doxorubicin hydrochloride (DOX) as well as pH controlled release. Anti-proliferative and cell viability analysis revealed that DOX loaded NPs showed excellent anti-proliferative characteristics against HeLa cells with favorable biocompatibility against normal cells. Such NPs have high potential for use as smart drug delivery carriers in advanced cancer therapy.
Article
The process of ionic gelation is one of the easiest ways to develop chitosan nanoparticles reported so far in the literature. Its success is mainly due to its one-shot synthesis, and to the mild environment required to produce the nanoparticles. The nanoparticle formation all along this process has been therefore thoroughly studied to lead to particles with a nanometric size, a narrow size distribution, and a spherical shape that are ideal for biomedical uses. The purpose of this review is to compile the biomedical applications that have been considered in the literature for these chitosan nanoparticles prepared by ionic gelation using tripolyphosphate as ionic agent. Their intrinsic biological properties such as non-toxicity, antimicrobial activity, mucoadhesivity and haemocompatibility are firstly discussed and compared to those of chitosan solutions. Then, the different bioactive species (drugs and biomacromolecules) incorporated in these chitosan nanoparticles, their maximal incorporation efficiency, their loading capacity, and their principal associated biomedical applications are presented.
Article
The objective of this review is to summarize recent advances in chitosan nanoparticles prepared by ionotropic gelation. Significant progress has occurred in this area since the method was first reported. The gelation technique has been improved through a number of creative methodological modifications. Ionotropic gelation via electrospraying and spinning disc processing produces nanoparticles with a more uniform size distribution. Large-scale manufacturing of the nanoparticles can be achieved with the latter approach. Hydrophobic and hydrophilic drugs can be simultaneously encapsulated with high efficiency by emulsification followed by ionic gelation. The turbulent mixing approach facilitates nanoparticle formation at a relatively high polymer concentration (5 mg/mL). The technique can be easily tuned to achieve the desired polymer/surface modifications (e.g., blending, coating, and surface conjugation). Using factorial-design-based approaches, optimal conditions for nanoparticle formation can be determined with a minimum number of experiments. New insights have been gained into the mechanism of chitosan-tripolyphosphate nanoparticle formation. Chitosan nanoparticles prepared by ionotropic gelation tend to aggregate/agglomerate in unfavorable environments. Factors influencing this phenomenon and strategies that can be adopted to minimize the instability are discussed. Ionically cross-linked nanoparticles based on native chitosan and modified chitosan have shown excellent efficacy for controlled and targeted drug-delivery applications.
Article
Indomethacin chitosan nanoparticles (NPs) were developed by ionotropic gelation and optimized by concentrations of chitosan and tripolyphosphate (TPP) and stirring time by 3-factor 3-level Box-Behnken experimental design. Optimal concentration of chitosan (A) and TPP (B) were found 0.6mg/mL and 0.4mg/mL with 120minutes stirring time (C), with applied constraints of minimizing particle size (R1) and maximizing encapsulation efficiency (R2) and drug release (R3). Based on obtained 3D response surface plots, factors A, B and C were found to give synergistic effect on R1, while factor A has a negative impact on R2 and R3. Interaction of AB was negative on R1 and R2 but positive on R3. The factor AC was having synergistic effect on R1 and on R3, while the same combination had a negative effect on R2. The interaction BC was positive on the all responses. NPs were found in the size range of 321-675nm with zeta potentials (+25 to+32mV) after 6 months storage. Encapsulation, drug release, and content were in the range of 56-79%, 48-73% and 98-99%, respectively. In vitro drug release data were fitted in different kinetic models and pattern of drug release followed Higuchi-matrix type.