Next Article in Journal
The Influence of Edible Oils’ Composition on the Properties of Beeswax-Based Oleogels
Next Article in Special Issue
Modern Herbal Nanogels: Formulation, Delivery Methods, and Applications
Previous Article in Journal
Recent Advances in Zwitterionic Hydrogels: Preparation, Property, and Biomedical Application
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis and Characterization of Acrylamide/Acrylic Acid Co-Polymers and Glutaraldehyde Crosslinked pH-Sensitive Hydrogels

1
Department of Pharmaceutics, Faculty of Pharmacy, Gomal University, Dera Ismail Khan 29050, Pakistan
2
Pharmaceutical Technology Unit, Faculty of Pharmacy, AIMST University, Bedong 08100, Kedah, Malaysia
3
Department of Pharmacy, Gono Bishwabidyalay, Mirzanagar, Savar, Dhaka 1344, Bangladesh
4
Institute of Health and Sports, Victoria University, Melbourne 3011, Australia
5
Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea
6
Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
7
Biology Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61421, Saudi Arabia
8
Zoology Department, Faculty of Science, Damanhour Univesity, Damanhour 22511, Egypt
9
Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
10
Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
11
Department of Pharmaceutical Sciences, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
12
Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 20 December 2021 / Revised: 31 December 2021 / Accepted: 5 January 2022 / Published: 9 January 2022
(This article belongs to the Special Issue Recent Advances on Nanogel Engineering for Drug Delivery)

Abstract

:
This project aims to synthesize and characterize the pH-sensitive controlled release of 5-fluorouracil (5-FU) loaded hydrogels (5-FULH) by polymerization of acrylamide (AM) and acrylic acid (AA) in the presence of glutaraldehyde (GA) as a crosslinker with ammonium persulphate as an initiator. The formulation’s code is named according to acrylamide (A1, A2, A3), acrylic acid (B1, B2, B3) and glutaraldehyde (C1, C2, C3). The optimized formulations were exposed to various physicochemical tests, namely swelling, diffusion, porosity, sol gel analysis, and attenuated total reflection-Fourier transform infrared (ATR-FTIR). These 5-FULH were subjected to kinetic models for drug release data. The 5-FU were shown to be soluble in distilled water and phosphate buffer media at pH 7.4, and sparingly soluble in an acidic media at pH 1.2. The ATR-FTIR data confirmed that the 5-FU have no interaction with other ingredients. The lowest dynamic (0.98 ± 0.04% to 1.90 ± 0.03%; 1.65 ± 0.01% to 6.88 ± 0.03%) and equilibrium swelling (1.85 ± 0.01% to 6.68 ± 0.03%; 10.12 ± 0.02% to 27.89 ± 0.03%) of formulations was observed at pH 1.2, whereas the higher dynamic (4.33 ± 0.04% to 10.21 ± 0.01%) and equilibrium swelling (22.25 ± 0.03% to 55.48 ± 0.04%) was recorded at pH 7.4. These findings clearly indicated that the synthesized 5-FULH have potential swelling characteristics in pH 6.8 that will enhance the drug’s release in the same pH medium. The porosity values of formulated 5-FULH range from 34% to 62% with different weight ratios of AM, AA, and GA. The gel fractions data showed variations ranging from 74 ± 0.4% (A1) to 94 ± 0.2% (B3). However, formulation A1 reported the highest 24 ± 0.1% and B3 the lowest 09 ± 0.3% sol fractions rate among the formulations. Around 20% drug release from the 5-FULH was found at 1 h in an acidic media (pH1.2), whereas >65% of drug release (pH7.4) was observed at around 25 h. These findings concluded that GA crosslinked 5-FU loaded AM and AA based hydrogels would be a potential pH-sensitive oral controlled colon drug delivery carrier.

1. Introduction

The worldwide prevalence of colonic disease is increasing with over one hundred thousand colorectal cancer patients newly diagnosed every year [1]. Traditional non-targeted therapy has unwanted side-effects and offers low efficacy owing to the systemic absorption of the drug before it reaches the disease site [2]. Therefore, colon-targeted drug delivery systems for the local treatment of colonic diseases are urgently sought [3]. Depending on the colonic milieu, colon targeted drug delivery systems specifically release the drug where it is needed, thus circumventing the untimely release of the drug in the upper gastrointestinal (GI) tract [4]. It is important to take into consideration the area neighboring the disease site(s) as well as the colon physiology, since the GI tract experiences constant changes in motility, fluid composition, pH from the small to the large intestine, and enzymatic activity [5]. Numerous formulation strategies have been investigated to develop colonic drug delivery (i.e., enzyme-sensitive systems, magnetically triggered systems, and pH-responsive systems) [6]. Hydrogels are a three-dimensional network of crosslinked polymer that can absorb and hold a significant amount of water inside the gap between the chains [7]. Hydrogels are extensively explored in a wide range of applications including medical, biological, and pharmaceutical fields due to their high-water content, swelling characteristics, biocompatibility, high permeability, and nontoxicity [8]. Besides this, stimuli-sensitive hydrogels are gaining attention as smart drug delivery systems as they can respond to the surrounding environment (e.g., temperature, pH, and enzymes). pH-sensitive hydrogels are preferable to other systems due to their higher controllability [9,10]. Several types of pH-responsive hydrogels have been investigated for site specific drug delivery owing to significant pH variations in the GI tract, where the drug release characteristics change with the variations in the medium pH because of relaxation of the chain [11].
AA is a pH-sensitive polyelectrolyte, which is superabsorbent as well as possessing concentration-dependent mucoadhesive properties and has been exclusively explored in colon drug delivery systems [12]. The advantage of AA is that variation in the composition of the polymers results in different drug release characteristics depending on the surrounding pH [13]. Temperature-sensitive and pH-responsive behavior has also been displayed by the copolymers AA and the interpenetrating networks (IPNs) [14]. Also, pH-sensitive macromolecular structures designed by the poly(AA) (PAA) have been explored for use in site-specific drug delivery systems [15]. Owing to the ionization of the carboxylic acid (CA) functionalities, the PAA based hydrogels swell considerably at pH 7.4 since its pKa value ranges from 4.5–5 [16].
AM has been employed in various biomedical applications including protein drug delivery due to its biocompatibility, pH-responsiveness and mucoadhesivity [17]. Success has been reported using an AM grafted N-succinyl chitosan-based hydrogel with a pH dependent release of insulin when administered orally [18].
GA is used as a crosslinking agent for the preparation of hydrogels [19]. It is also employed for cell and enzyme immobilization due to its higher aqueous solubility, ease of accessibility, low cost and efficiency of biomaterials stabilization [20]. GA contains two aldehyde groups which can form immediate covalent bonds with the functionalities available (i.e., imidazoles, phenols, hydroxyl, and thiols) [21]. It has been reported that at a pH of around 7, GA reacts more quickly with amine groups [22] as well as forming better chemical and thermal crosslinks than those of other aldehydes [23].
5-FU is frequently used as one of the mainstays in potential antitumor drugs in clinical chemotherapy, specifically for the treatment of colon cancer [24]. It has a short biological half-life (10–20 min), causing partial absorption where? Upper intestine? since it is a sparingly water-soluble pyrimidine antimetabolite. The drug is rapidly metabolized by dihydro pyrimidine dehydrogenase after oral delivery [25]. The clinical use of 5-FU affects the normal cells, leading to the adverse effects on GI tracts, the central nervous system and bone marrow, owing to the lack of tumor selectivity [26,27]. In recent studies, pH-sensitive hydrogels have been investigated by researchers for controlled delivery of 5-FU to reduce these side effects and to enhance the antitumor activity [28]. PAA and its derivatives-based pH-responsive hydrogels can circumvent the effect of the highly acidic surroundings on drugs [29]. The pKa of PAA is around 3.5, therefore at low pH, the hydrogels based on PAA become contracted resulting in the premature release of the drug in the stomach. Consequently, at higher pH, they remain swollen, leading to drug release in the intestine. Nevertheless, avoiding premature drug release before entering the colon section is very challenging due to the pH resemblance between the small intestine and the colon [30].
The goal of this study is to synthesize pH-responsive hydrogels using AM, AA and GA. Hydrogels were prepared by a simple crosslinking method, employing 5-FU as a model drug. The ATR-FTIR spectroscopy was used to investigate the interaction between the drug and the excipients. Swelling characteristics, diffusion coefficient, drug loading, in vitro release and release kinetics were also studied.

2. Results and Discussion

2.1. Synthesis Hydrogel

The mechanism of polymerization of acrylic acid and acrylamide in the presence of glutaraldehyde (GA) cross-linker was adapted from Wang et al. and partially modified the synthesis reaction mechanism shown in Scheme 1. Acrylic acid (1), acrylic acid sodium (2) and acrylamide (3) can react to Poly(acrylic acid-co-acrylamide) (4) using KPS as a radical initiator. The persulfate initiator was decomposed under heating to generate sulfate anion radical. The cross-linker glutaraldehyde (5) was added after polymerization finished in order to avoid the crosslinking reaction of Poly(acrylic acid-co-acrylamide) (4) during polymerization. Through heat-treatment at 110–160 °C the amide groups and hydroxyl groups can react with aldehyde groups from glutaraldehyde (5) to form poly(AA-co-AM) (6) network structure. Grafted hydrogels exhibit pH-sensitive swelling properties and pH-controlled drug release behavior.

2.2. Solubility Study

The 5-FU is a nucleobase analogue having a pKa value of (8 ± 0.1). It has a water solubility of 12.2 g/L at 20 °C. (repeated) It is freely soluble in a phosphate buffer of pH 7.4 as well as in distilled water compared to pH 1.2 (Table 1). The solubility study is an important parameter which conforms to the dissolution experiments.

2.3. FTIR

FTIR spectral analysis of 5-FU and 5-FULH is shown in Figure 1. Pure 5-FU, drug loaded, and unloaded hydrogel samples were assessed using FTIR in the array of 400–4000 cm−1. FTIR represents the distinct peak at 3177 cm−1, 2927 cm−1, 1658 cm−1 in pure 5-FU spectra and 3196 cm−1, 2924 cm−1 and 1654 cm−1 in 5-FULH. In the case of pure 5-FU, the bands at 3412 cm−1, 1165 cm−1 show evidence of N-H stretching (free) and –C-F band; after drug loading some of the bands disappear and N-H stretching (free) appears at the same wave number, while –C-F band appears at 1165 cm−1. This indicates that the 5-FU is molecularly dispersed into the prepared hydrogels [32]. In the case of unloaded hydrogel, see Figure 1c, the AM peak appears at 1647 cm−1 for primary amide (C=O) stretching. The peak for the alkene group (HC=CH) appears at 3065 cm−1. In the band of GA, the peak assigned to the C-OH stretching at 1243 cm−1 is very clear, suggesting that the desired material has been successfully prepared [33]. Owing to the electrostatic interaction among the various functional groups of AM, AA and GA, the characteristic peaks of AM, AA and GA are shifted to developed 5-FULH. Figure 1b shows that the loading of 5-FU by the developed hydrogel was successful and no interaction between the 5-FU and hydrogel ingredients was detected [34].

2.4. Swelling Studies

The release of the drug from the 5-FULH occurs once the polymer network is dissolved, followed by drug diffusion from the surface of the structure which is associated with the swelling behavior of the 5-FULH [35]. The advantage of hydrogel is that it can swell in the surrounding medium due to its chemical structure, which allows affinity with the water molecules [36]. Swelling studies of 5-FULH were conducted using various pH mediums of 1.2, 6.8, and 7.4. Equilibrium and dynamic swelling proportions of different 5-FULH are depicted in Table 2.

2.5. Effect of AM

Figure 2 and Figure 3 describe the swelling attitude of 5-FULH with various concentrations of AM. The swelling values of these gels were measured at 37 °C and medium pH of 1.2, 6.8, and 7.4. The swelling increased with an increase in AM concentration and a raise in medium pH. This swelling behavior of the polymer was due to its hydrophilic nature. The physical nature and hydrogen bond formation restricts the release of the drug.

2.6. Effect of AA

Figure 4 and Figure 5 show the swelling attitude of 5-FULH with various concentrations of AA. The swelling values of these gels were measured at 37 °C and medium pH of 1.2, 6.8, and 7.4. The swelling increased with an increase in AA concentration and a raise in medium pH. This swelling behavior of the polymer was again due to its hydrophilic nature. The physical nature and hydrogen bond formation again restricts the release of the drug.

2.7. Effect of GA

Introduction of a crosslinking agent such as GA affects the swelling behavior of 5-FULH. Figure 6 and Figure 7 show GA at different concentrations and the effect over the swelling of hydrogel at 37 °C in medium pH of 1.2, 6.8, and 7.4. Results show that when the concentration of GA increases, the swelling ratio decreases. This might be due to increased cross linking of GA. Generally, mobility of the polymer chain is affected by the crosslinking, ensuring the low solubility of the polysaccharide [37]. GA promotes the degree of crosslinking which in turn results in folding of the polymeric chains and the subsequent attainment of reticulation point, thus affecting the aqueous absorption capacity [38].

2.8. Porosity Measurement

The porosity values of 5-FULH range from 34% to 62% with different weight ratios of AM, AA, and GA. It was found that the higher the concentration of polymer and monomer, the higher the porosity values. While enhancing the crosslinker concentration, the porosity values decrease. Table 3 (B3) shows a higher % of gel fraction (94 ± 0.2%) with a higher % of porosity (62 ± 0.06%); a similar type of relationship was found in previous studies. The controlled release of the drug, around 65% at 25 h, could be attributed to the reduction of water entry and subsequent diffusion of the drug from the hydrogel network [39].

2.9. Gel Fraction

Table 3 shows the results of gel and sol fraction. In relation to the value of gel fraction, it was observed that the gel fraction depended on the AM and AA. The gelling strength of the prepared 5-FULH may increase with the higher content of AM, AA, and GA. As the concentration of the AA is increased, the polymerization reaction is also enhanced due to the accessibility of more binding sites. The higher gel fraction is attributed to the increased bulk density of the hydrogel structure. A firm and robust hydrogel network is established due to the higher concentration of polymer, resulting in a greater degree of crosslinking which in turn leads to a lower porous structure of hydrogels [40]. However, sol fraction is decreased since the concentration of AA and GA is increased due to the inverse relationship with gel fraction [41].

2.10. Drug Loading

Drug loading was determined by using swelling, extraction, and weight values. The amount of 5-FU was presented as g/g of dry gel and the data were presented in Table 4.

2.11. Cumulative Drug Release Measurement

From the polymeric 5-FULH, it can be seen that the release pattern of a formulation depends on the swelling behavior of these hydrogels. This section discusses some of the factors which influence the release of 5-FU from the prepared hydrogels.

2.12. Effect of AM

These 5-FULH were designed by varying AM concentrations at 37 °C and pH of 1.2 and 7.4. The release of 5-FU is very low in an acidic medium, increasing to 67% in a basic medium. This might be due to greater swelling in a basic medium and less swelling in an acidic medium. Mundargi et al. reported that the release of 5-FU increases when AM content increases in the matrix [42]. Figure 8 shows that drug release was varied based on the pH of the medium i.e., around 20% drug release was found at 1 h in an acidic media (pH 1.2), whereas >65% of drug release (pH 7.4) was observed at around 25 h from or with? the 5-FULH containing a higher amount of AM due to increased polymer chain flexibility, as shown in Figure 9. This release behavior of the drug was in agreement with the previous study where single polymeric chains in pure AM allowed higher drug transportation compared to the co-polymeric chain [43].

2.13. Effect of AA

Figure 10 and Figure 11 present the release profile of 5-FU under the influence of AA. The higher the concentration of AA, the higher the release of 5-FU. Enhancing the quantity of polymer AA increases the release rate of 5-FU due to the development of channels and pores in the hydrogel matrix resulting from relaxation of the polymer network. This result is in agreement with previous studies where drug release was enhanced with the higher pH and the amount of AA [44].

2.14. Effect of GA

Figure 12 and Figure 13 describe the release of 5-FU under different concentrations of GA. The release of 5-FU in an acidic medium was almost the same irrespective of different concentrations of GA. The release of 5-FU was lower with a higher concentration of GA when it comes to basic medium pH 7.4. This could be attributed to the higher H-bonding and lower swelling of 5-FULH with a high concentration of crosslinking agent. It has been reported that the gelation ratio is faster in GA crosslinked hydrogel, therefore higher GA concentration increases the drug loading capacity [45]. As seen in Figure 13, the 5-FU release percentages from the 5-FULH at pH 7.4 were more than 50% of the pH 1.2 at 18% in every formulation. The present findings are in line with the previous data which investigated 5-FU loaded GA blended pH-responsive hydrogel and found 5-FU release of around 64.0% to 85.99% at pH 7.4 but only 13.33% to 19.64% at pH 1.2 [46].
The protonation of sulfonate ions occurs at acidic pH 1.2, which results in the generation of strong hydrogen bonds and physical interaction between the functional groups of the hydrogel, causing a subsequent reduction in swelling [47]. However, at pH 7.4, deprotonation of sulfonate ions occurs, owing to the enhanced electronic density on the polymeric structure and less physical interaction among the –SO−3 moieties, thereby generating a greater degree of swelling of AA 5-FULH [48]. All of these factors cause reduction in intermolecular hydrogen bonding, resulting in higher swelling [49]. Likewise, the COOH groups of AA are protonated at pH 1.2 which leads to a fall in swelling. However, at higher pH 7.4, carboxylic acid (CA) functionalities are deprotonated, resulting in higher swelling of the 5-FULH [50]. Also, the osmotic pressure of the ions is increased due to the increase in protonation of the COOH groups [51]. The carboxylic acid (CA) moieties were converted to the salt form which led to maximum swelling, as the pH of the medium was elevated from a lower to a higher number [52].

2.15. Release Kinetics

Several kinetic models were employed to help understand the release mechanism of the 5-FU from the hydrogels, and the regression coefficients (r) were selected for the evaluation of the most suitable drug release [53]. The (r) values of most samples were greater for zero-order kinetics compared to the first-order kinetics, which could be ascribed to the variation in the amounts of AA and GA. From the results of the Higuchi model, it was obvious that the release of the drug follows the diffusion-controlled release mechanism. Among the models, the (r) values of 5-FULH for zero order (0.9632–0.9954) was comprehensively higher than the first order (0.9378–0.9932). The results indicated that the release pattern of the drug corresponded to the zero-order kinetics.

3. Conclusions

The 5-FULH were processed for oral delivery using AM, AA and GA crosslinked. These prepared 5-FULH were physio-chemically characterized. ATR-FTIR shows no such interaction between the excipients and the model drug (i.e., 5-FU). Swelling studies show that minimum swelling was achieved at acidic pH and maximum at alkaline Ph, which depends on the monomer, polymer, and crosslinking agent. By increasing AM and AA concentrations, swelling increased, whereas by increasing GA, it decreased. In vitro release studies show that the most drug was released on alkaline pH, whereas the least drug was released on acidic pH. The release of the drug also increases with increasing AM and AA content but decreases with an increase in cross linker. The drug release kinetics models followed a non-Fickian order of release. These findings concluded that AM and AA based 5-FULH would be appropriate for controlled drug delivery with pH reactive characteristics.

4. Materials and Methods

4.1. Materials

5-Fluorouracil (5-FU) was obtained from Biolabs Pharma Pvt. Ltd., (Biolabs Pharma, Islamabad, Pakistan). Acrylamide (AM), acrylic acid (AA), glutaraldehyde (GA), disodium hydrogen phosphate, sodium hydroxide, and monobasic potassium phosphate were purchased from Sigma Aldrich (Sigma Aldrich, Darmstadt, Germany). Hydrochloric acid and acetic acid were supplied by Icon Chemicals (Icon Chemicals, Ludhiana, Punjab, India).

4.2. Pre-Formulation Studies and Standard Curve

The stock solution was prepared using 10 mg of 5-FU dissolved in 100 mL of phosphate buffer pH 7.4. A series of dilutions were made in descending order using phosphate buffer pH 7.4. Samples from such mixtures were taken and spectrophotometrically analyzed at 256 nm (Shimadzu 601, Japan). The results were taken in triplicate and plotted.
Y = M C + B
where:
Y = the absorbency of the solution, M = the angle of standard curve of identified concentration, C = concentration that must be calculated and B = the curve’s cut off.

4.3. Solubility Study of Drug

The solubility study of 5-FU was conducted using different solvents of varying pH at 37 °C.

4.4. Preparation of 5-FULH

The 5-FULH were produced by a simple crosslinking method, with slight modifications as presented in Table 5. In the first step a specific amount of distilled water was put into a beaker and placed on a magnetic stirrer and heated. Thereafter the AM was poured into the beaker and heated up till a clear solution was achieved. Similarly, by applying slight modifications in temperature as well as in revolution time, an AA solution was obtained. These solutions were then thoroughly mixed with continuous stirring on a magnetic stirrer until a clear homogeneous liquid was produced. Finally, by adding distilled water, the volume of the solution was made up to a hundred milliliters. Consequently, in a drop wise method and with regular stirring, cross linker was added to the mixture, to produce a uniform clear solution. This solution was then poured into the deoxygenized test tubes and left to congeal in them for four days. The 5-FULH, once they were formed, were then removed from the tubes. With the help of sharpened blades, these gels were cleaved as well as properly scrubbed with Methanol. The discs were then put into petri dishes and desiccated in an oven at 40 °C for seventy-two hours [54,55].

4.5. Hydrogel’s Characterizations

4.5.1. ATR-FTIR

Physical interactions between drug and excipients were investigated using ATR-FTIR (Perkin Elmer, Waltham, MA, USA). The samples were placed on ATR-FTIR and scanned from 400–4000 cm−1 [56].

4.5.2. Dynamic Swelling Study

Dried slices of these preparations were properly weighed and immersed into buffers of various pH (i.e., pH 7.4, pH 6.8, and pH 1.2). By removing discs from the medium, readings were noted over regular time periods [57,58]. The surface water was removed from the 5-FULH using tissue paper. The discs were dipped again in the pH 6.8 and 7.4 buffer, after having been weighed at definite intervals [59]. The following formula was used to obtain the dynamic S:
S = W s W d
where:
Ws = weight of swollen gel at specific time, Wd = weight of dry hydrogel, and S = swelling ratio.

4.5.3. Equilibrium Swelling Study

ES studies were also performed on the 5-FULH, which were prepared for frequent swelling. The % ES was determined using the following equation [60,61].
E S   ( % ) = W s W d W d × 100
where:
ES = equilibrium swelling, Ws = weight of swollen gel at specific time, and Wd = weight of dry hydrogel.

4.5.4. Diffusion Coefficient (DC)

DC is the quantity of solvent that was absorbed/diffused across the unit area in unit time through a concentration gradient [62,63]. The diffusion coefficient was calculated by Formula (4):
D = π ( h · θ 4 · Q e q ) 2
where:
D = DC of the 5-FULH, Qeq = the hydrogel’s swelling at equilibrium, θ = the angle of straight portion of swelling curves and h = early sample thickness.

4.5.5. Sol Gel Fraction Analysis

Sol-gel analysis technique was performed for predisposing reactant quantity used during the preparation of the 5-FULH. The prepared 5-FULH were dried without washing and placed in deionized water at ambient temperature until constant weight was attained. After that, extracted hydrogel was taken out and dried in an oven at 60 °C [64,65].
Sol   fraction   ( SF )   ( % ) = M 2 M 1 M 1 × 100
where:
M2 = Final/extracted gel wt, M1 = Initial wt of dry gel and
G e l   f r a c t i o n = 100 S F

4.5.6. Porosity Measurement

Porosity is an important consideration mainly affecting the swelling attributes of the 5-FULH. The % porosity was calculated by solvent replacement technique. Hydrogel discs were dried and soaked in ethanol (100%) overnight. Extra ethanol was removed using blotting paper and then the 5-FULH were weighed [66]. The porosity was calculated and attained by using the following equation:
Porosity   ( % ) = M t M o ρ v × 100
where:
Mt = weight before immersion, Mo = weight after immersion, ρ = density of absolute ethanol and v = volume of hydrogel.

4.5.7. Drug Loading

Samples which showed maximum swelling were used for drug loading and release studies. The drug loading into the discs of hydrogel was achieved by soaking them for one week in a solution of the drug. A 1% w/v 5-FU in pH 7.4 solution was used for drug loading. After achieving the equilibrium value, the swelled 5-FULH were removed from the drug solution, blotted with filter paper, first dried at room temperature, and then placed in an oven at 40–45 °C for one week to remove the absorbed solvent. To determine the percentage of drug-loading, weighed drug loaded samples were extracted repeatedly using a phosphate buffer solution of pH 7.4 up to exhaustion, and the concentration of the drugs in pooled extract was determined spectrophotometrically at λ max 256 nm. The quantity of drug loaded into the 5-FULH was also determined by the swelling method [67].

4.5.8. In Vitro Release Study

To assess the amount of released drug from formulated 5-FULH, in vitro release studies were performed, using dissolution apparatus (pharma test; Pt-Dt 7). These hydrogel discs were positioned in a dissolution medium of 500 mL at 37 °C temp in a pH of 1.2 and 7.4 and shaken at a hundred revolutions per minute. Samples were selected at specific time periods and replaced. These samples were then evaluated at 256 nm using a UV-spectrophotometer (Shimadzu) [68].

4.5.9. Release Kinetics

To investigate the discharge of medicament from the gels and its mechanism, various kinetic models were applied [58], which are given below.
First order kineticsln (1 − F) = K1t
Zero order kineticsMo − Mt = Kot
Higuchi modelQ = K2t1/2
Korsmyer Peppas ModelMt/Mœ = Ktn

4.6. Statistical Analysis

Findings from all of the experiments were made in triplicate and presented as the means ± S.D. These samples were analyzed statistically with the help of one-way ANOVA and t-test. At p < 0.05, differences were considered as significant.

Author Contributions

Conceptualization, A.K.A., and M.S.; methodology, M.A.K., A.K.A., M.S., A.N., and M.A.; software, M.A., A.K.A., and M.S.; validation, A.K.A.; formal analysis, M.A., and A.K.A.; investigation, M.A.K, M.A., and M.S.; resources, M.S., A.N., and M.A.; data curation, A.K.A.; writing—original draft preparation, M.A.K, A.K.A., M.A., and M.S.; review and editing, A.K.A., P.P., M.K.H., R.S.B., A.F.E.-k., M.K., S.G.B., M.M.A.-D., and M.H.R.; visualization, A.K.A.; supervision, M.S., A.N., and M.A.; project administration, A.K.A., and M.S.; Funding, A.K.A., and M.M.A.-D. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Taif University Researchers Supporting Program (Project number: TURSP-2020/269), Taif University, Saudi Arabia. The authors would like to thank the Deanship of Scientific Research at King Khalid University, Abha, KSA, for funding this work under grant number (R.G.P.1/62/42).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data will be available upon request.

Acknowledgments

This work was supported by the Taif University Researchers Supporting Program (Project number: TURSP-2020/269), Taif University, Saudi Arabia. The authors would like to thank the Deanship of Scientific Research at King Khalid University, Abha, KSA, for funding this work under grant number (R.G.P.1/62/42).

Conflicts of Interest

Authors have declared no conflict of interest with other forms of study.

References

  1. Olga, H.; Maryam, A.S.B.; Karim, S.S.; Brigitta, L.; Alf, L.; Claus-Michael, L. Drug delivery to the inflamed intestinal mucosa-targeting technologies and human cell culture models for better therapies of IBD. Adv. Drug Deliv. Rev. 2021, 175, 113828. [Google Scholar] [CrossRef]
  2. Ajitkumar, B.N.; Kumar, L. Treating colon cancers with a non-conventional yet strategic approach: An overview of various nanoparticulate systems. J. Control. Release 2021, 336, 16–39. [Google Scholar] [CrossRef]
  3. Patole, V.C.; Pandit, A.P. Mesalamine-loaded alginate microspheres filled in enteric coated HPMC capsules for local treatment of ulcerative colitis: In vitro and in vivo characterization. J. Pharm. Investig. 2018, 48, 257–267. [Google Scholar] [CrossRef]
  4. Hani, U.; Honnavalli, Y.K.; Begum, M.Y.; Yasmin, S.; Osmani, R.A.M.; Ansari, M.Y. Colorectal cancer: A comprehensive review based on the novel drug delivery systems approach and its management. J. Drug Deliv. Sci. Technol. 2021, 63, 102532. [Google Scholar] [CrossRef]
  5. Hua, S.; Marks, E.; Schneider, J.J.; Keely, S. Advances in oral nano-delivery systems for colon targeted drug delivery in inflammatory bowel disease: Selective targeting to diseased versus healthy tissue. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 1117–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lee, S.H.; Bajracharya, R.; Min, J.Y.; Han, J.-W.; Park, B.J.; Han, H.-K. Strategic Approaches for Colon Targeted Drug Delivery: An Overview of Recent Advancements. Pharmaceutics 2020, 12, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Bignotti, F.; Baldi, F.; Grassi, M.; Abrami, M.; Spagnoli, G. Hydrophobically-Modified PEG Hydrogels with Controllable Hydrophilic/Hydrophobic Balance. Polymers 2021, 13, 1489. [Google Scholar] [CrossRef]
  8. Chai, Q.; Jiao, Y.; Yu, X. Hydrogels for Biomedical Applications: Their Characteristics and the Mechanisms behind Them. Gels 2017, 3, 6. [Google Scholar] [CrossRef] [Green Version]
  9. Mudassir, J.; Ranjha, N.M. Dynamic and equilibrium swelling studies: Crosslinked pH sensitive methyl methacrylate-co-itaconic acid (MMA-co-IA) hydrogels. J. Polym. Res. 2008, 15, 195–203. [Google Scholar] [CrossRef]
  10. Elliott, J.E.; Macdonald, M.; Nie, J.; Bowman, C.N. Structure and swelling of poly (acrylic acid) hydrogels: Effect of pH, ionic strength, and dilution on the crosslinked polymer structure. Polymer 2004, 45, 1503–1510. [Google Scholar] [CrossRef]
  11. Ranjha, N.M.; Ayub, G.; Naseem, S.; Ansari, M.T. Preparation and characterization of hybrid pH-sensitive hydrogels of chitosan-coacrylic acid for controlled release of verapamil. J. Mater. Sci. Mater. Med. 2010, 21, 2805–2816. [Google Scholar] [CrossRef]
  12. Akhlaq, M.; Azad, A.K.; Ullah, I.; Nawaz, A.; Safdar, M.; Bhattacharya, T.; Uddin, A.B.; Abbas, S.A.; Mathews, A.; Kundu, S.K.; et al. Methotrexate-loaded gelatin and polyvinyl alcohol (Gel/PVA) hydrogel as a pH-sensitive matrix. Polymers 2021, 13, 2300. [Google Scholar] [CrossRef] [PubMed]
  13. Malviya, R.; Sundram, S.; Fuloria, S.; Subramaniyan, V.; Sathasivam, K.V.; Azad, A.K.; Sekar, M.; Kumar, D.H.; Chakravarthi, S.; Porwal, O.; et al. Evaluation and Characterization of Tamarind Gum Polysaccharide: The Biopolymer. Polymers 2021, 13, 3023. [Google Scholar] [CrossRef] [PubMed]
  14. Lo, Y.L.; Hsu, C.Y.; Lin, H.R. pH-andthermo-sensitivepluronic/poly(acrylic acid) in situ hydrogels for sustained release of an anticancer drug. J. Drug Target. 2013, 21, 54–66. [Google Scholar] [CrossRef] [PubMed]
  15. Bajpai, A.K.; Kankane, S. Evaluation of water sorption property and in vitro blood compatibility of poly(2-hydroxyethyl methacrylate) (PHEMA) based semi interpenetrating polymer networks (IPNs). J. Mater. Sci. Mater. Med. 2008, 19, 1921–1933. [Google Scholar] [CrossRef]
  16. Kheirandish, S.; Jabbari, E. Effect of surface polarity on wettability and friction coefficient of silicone rubber/poly(acrylic acid) hydrogel composite. Colloid Polym. Sci. 2006, 284, 1411–1417. [Google Scholar] [CrossRef]
  17. Rizwan, M.; Yahya, R.; Hassan, A.; Yar, M.; Azzahari, A.D.; Selvanathan, V.; Sonsudin, F.; Abouloula, C.N. pH Sensitive Hydrogels in Drug Delivery: Brief History, Properties, Swelling, and Release Mechanism, Material Selection and Applications. Polymers 2017, 9, 137. [Google Scholar] [CrossRef]
  18. Sudhakar, K.; Fuloria, S.; Subramaniyan, V.; Sathasivam, K.V.; Azad, A.K.; Swain, S.S.; Sekar, M.; Karupiah, S.; Porwal, O.; Sahoo, A.; et al. Ultraflexible Liposome Nanocargo as a Dermal and Transdermal Drug Delivery System. Nanomaterials 2021, 11, 2557. [Google Scholar] [CrossRef] [PubMed]
  19. Wu, L.; Brazel, C.S. Modifying the release of proxyphylline from PVA hydrogels using surface crosslinking. Int. J. Pharm. 2008, 349, 144–151. [Google Scholar] [CrossRef]
  20. Kozlovskaya, V.; Kharlampieva, E.; Sukhishvili, S.A. 1.124-Polymer Films Using LbL Self-Assembly. Compr. Biomater. 2011, 1, 417–430. [Google Scholar] [CrossRef]
  21. Kim, K.-J.; Lee, S.-B.; Han, N.-W. Kinetics of crosslinking reaction of PVA membrane with glutaraldehyde. Korean J. Chem. Eng. 1994, 11, 41–47. [Google Scholar] [CrossRef]
  22. Okuda, K.; Urabe, I.; Yamada, Y.; Okada, H. Reaction of glutaraldehyde with amino and thiol compounds. J. Ferment. Bioeng. 1991, 71, 100–105. [Google Scholar] [CrossRef]
  23. Khan, T.A.; Azad, A.K.; Fuloria, S.; Nawaz, A.; Subramaniyan, V.; Akhlaq, M.; Safdar, M.; Sathasivam, K.V.; Sekar, M.; Porwal, O.; et al. Chitosan-Coated 5-Fluorouracil Incorporated Emulsions as Transdermal Drug Delivery Matrices. Polymers 2021, 13, 3345. [Google Scholar] [CrossRef] [PubMed]
  24. Chandran, S.P.; Natarajan, S.B.; Chandraseharan, S.; Mohd Shahimi, M.S.B. Nano drug delivery strategy of 5-fluorouracil for the treatment of colorectal cancer. J. Cancer Res. Pract. 2017, 4, 45–48. [Google Scholar] [CrossRef]
  25. Swamy, B.Y.; Chang, J.H.; Ahn, H.; Lee, W.-K.; Chung, I. Thermoresponsive N-vinyl caprolactam grafted sodium alginate hydrogel beads for the controlled release of an anticancer drug. Cellulose 2013, 20, 1261–1273. [Google Scholar] [CrossRef]
  26. Anirudhan, T.S.; Mohan, A.M. Novel pH switchable gelatin-based hydrogel for the controlled delivery of the anticancer drug 5-fluorouracil. RSC Adv. 2014, 4, 12109–12118. [Google Scholar] [CrossRef]
  27. Eswaramma, S.; Rao, K.S.V.K.; Rao, K.M. Diffusion and Controlled Release Characteristics of Ph-Sensitive Poly(2-(Dimethyl Amino)ethyl Methacrylate-co-2-hydroxyethylacrylate) Hydrogels. Int. J. Polym. Mater. Polym. Biomater. 2016, 65, 134–142. [Google Scholar] [CrossRef]
  28. Che, Y.; Li, D.; Liu, Y.; Ma, Q.; Tan, Y.; Yue, Q.; Meng, F. Physically crosslinked pH-responsive chitosan-based hydrogels with enhanced mechanical performance for controlled drug delivery. RSC Adv. 2016, 6, 106035–106045. [Google Scholar] [CrossRef]
  29. Ma, Z.; Ma, R.; Wang, X.; Gao, J.; Zheng, Y.; Sun, Z. Enzyme and pH responsive 5-flurouracil (5-FU) loaded hydrogels based on olsalazine derivatives for colon-specific drug delivery. Eur. Polym. J. 2019, 118, 64–70. [Google Scholar] [CrossRef]
  30. Latif, M.S.; Azad, A.K.; Nawaz, A.; Rashid, S.A.; Rahman, M.; Al Omar, S.Y.; Bungau, S.G.; Aleya, L.; Abdel-Daim, M.M. Ethyl Cellulose and Hydroxypropyl Methyl Cellulose Blended Methotrexate-Loaded Transdermal Patches: In Vitro and Ex Vivo. Polymers 2021, 13, 3455. [Google Scholar] [CrossRef]
  31. Wang, M.Y.; Wang, Y.P.; Yu, Y.T.; Wang, Y.M.; Ni, J.H. Glutaraldehyde-Cross-Linked Poly(acrylic acid-co-acrylamide)/Poly(vinyl alcohol) Superabsorbent Membranes. Mater. Sci. Forum 2014, 789, 194–200. [Google Scholar] [CrossRef]
  32. Suhail, M.; Fang, C.W.; Khan, A.; Minhas, M.U.; Wu, P.C. Fabrication and In Vitro Evaluation of pH-Sensitive Polymeric Hydrogels as Controlled Release Carriers. Gels 2021, 7, 110. [Google Scholar] [CrossRef] [PubMed]
  33. Akhtar, M.F.; Ranjha, N.M.; Hanif, M. Effect of ethylene glycol dimethacrylate on swelling and on metformin hydrochloride release behavior of chemically crosslinked pH–sensitive acrylic acid–polyvinyl alcohol hydrogel. DARU J. Pharm. Sci. 2015, 23, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Al-Tabakha, M.M.; Khan, S.A.; Ashames, A.; Ullah, H.; Ullah, K.; Murtaza, G.; Hassan, N. Synthesis, Characterization and Safety Evaluation of Sericin-Based Hydrogels for Controlled Delivery of Acyclovir. Pharmaceuticals 2021, 14, 234. [Google Scholar] [CrossRef]
  35. Ugwu, O.B.; Barminas, J.T.; Nkafamiya, I.I.; Akinterinwa, A. Synthesis and Characterization of Crosslinked Hydrogel of Konkoli (Maesopsis eminii) Grafted Polymethylacrylamide for a Preliminary Study as an Insulin Delivery System. J. Biosci. Med. 2016, 4, 36–47. [Google Scholar] [CrossRef] [Green Version]
  36. Cavalcante, L.A.; Batista, B.S.; Fonseca, A.P.G.; Balteiro, A.J.D.; Amaral, F.M.B.; Mendes, F.; Macêdo, A.A.M. Effect of pH on Swelling Profile of Glutaraldehyde Crosslinked Adenanthera pavonina L. Galactomannan. In IOP Conference Series: Materials Science and Engineering, Proceedings of the 5th International Conference on Architecture, Materials and Construction (ICAMC 2019), Lisbon, Portugal, 2–4 December 2019; IOP Publishing: Bristol, UK, 2020; Volume 809, p. 012023. [Google Scholar] [CrossRef]
  37. Kunjiappan, S.; Theivendran, P.; Baskararaj, S.; Sankaranarayanan, B.; Palanisamy, P.; Saravanan, G.; Arunachalam, S.; Sankaranarayanan, M.; Natarajan, J.; Somasundaram, B.; et al. Modeling a pH-sensitive Zein-co-acrylic acid hybrid hydrogels loaded 5-fluorouracil and rutin for enhanced anticancer efficacy by oral delivery. 3 Biotech 2019, 9, 185. [Google Scholar] [CrossRef]
  38. Shah, M.K.; Azad, A.K.; Nawaz, A.; Ullah, S.; Latif, M.S.; Rahman, H.; Alsharif, K.F.; Alzahrani, K.J.; El-Kott, A.F.; Albrakati, A.; et al. Formulation Development, Characterization and Antifungal Evaluation of Chitosan NPs for Topical Delivery of Voriconazole In Vitro and Ex Vivo. Polymers 2022, 14, 135. [Google Scholar] [CrossRef]
  39. Ali, L.; Ahmad, M.; Usman, M. Evaluation of crosslinked hydroxypropyl methylcellulose graft-methacrylic acid copolymer as extended-release oral drug carrier. Cell. Chem. Technol. 2015, 49, 143–151. [Google Scholar]
  40. Dergunov, S.A.; Nam, I.K.; Mun, G.A.; Nurkeeva, Z.S.; Shaikhutdinov, E.M. Radiation synthesis and characterization of stimuli-sensitive chitosan–polyvinyl pyrrolidone hydrogels. Radiat. Phys. Chem. 2005, 72, 619–623. [Google Scholar] [CrossRef]
  41. Mundargi, R.C.; Rangaswamy, V.; Aminabhavi, T.M. A novel method to prepare 5-fluorouracil, an anti-cancer drug, loaded microspheres from poly (N-vinyl caprolactam-co-acrylamide) and controlled release studies. Des. Monomers Polym. 2010, 13, 325–336. [Google Scholar] [CrossRef]
  42. Kulkarni, A.R.; Soppimath, K.S.; Aminabhavi, T.M.; Dave, A.M.; Mehta, M.H. Glutaraldehyde crosslinked sodium alginate beads containing liquid pesticide for soil application. J. Control. Release 2000, 63, 97–105. [Google Scholar] [CrossRef]
  43. Sohail, K.; Khan, I.U.; Shahzad, Y.; Hussain, T.; Ranjha, N.M. pH-sensitive polyvinylpyrrolidone-acrylic acid hydrogels: Impact of material parameters on swelling and drug release. Braz. J. Pharm. Sci. 2014, 50, 173–184. [Google Scholar] [CrossRef]
  44. Özbaş, Z.; Gürdağ, G. Synthesis and Characterization of 5-Fluorouracil-Loaded Glutaraldehyde Crosslinked Chitosan Hydrogels. Süleyman Demirel Üniv. Fen Bilim. Enst. Derg. 2016, 20, 460–467. [Google Scholar] [CrossRef] [Green Version]
  45. Ju, H.K.; Kim, S.Y.; Lee, Y.M. pH/temperature-responsive behaviors of semi-IPN and comb-type graft hydrogels composed of alginate and poly (N-isopropylacrylamide). Polymer 2001, 42, 6851–6857. [Google Scholar] [CrossRef]
  46. Ju, Y.-H.; Huynh, L.-H.; Kasim, N.S.; Guo, T.-J.; Wang, J.-H.; Fazary, A.E. Analysis of soluble and insoluble fractions of alkali and subcritical water treated sugarcane bagasse. Carbohydr. Polym. 2011, 83, 591–599. [Google Scholar] [CrossRef]
  47. Bashir, S.; Teo, Y.Y.; Naeem, S.; Ramesh, S.; Ramesh, K. pH responsive N-succinyl chitosan/Poly (acrylamide-co-acrylic acid) hydrogels and in vitro release of 5-fluorouracil. PLoS ONE 2017, 12, e0179250. [Google Scholar]
  48. Bashir, S.; Teo, Y.Y.; Ramesh, S.; Ramesh, K. Synthesis, characterization, properties of N-succinyl chitosan-g-poly (methacrylic acid) hydrogels and in vitro release of theophylline. Polymer 2016, 92, 36–49. [Google Scholar] [CrossRef]
  49. Atta, A.M. Swelling behaviors of polyelectrolyte hydrogels containing sulfonate groups. Polym. Adv. Technol. 2002, 13, 567–576. [Google Scholar] [CrossRef]
  50. Ali, A.E.-H. Removal of heavy metals from model wastewater by using carboxymehyl cellulose/2-acrylamido-2-methyl propane sulfonic acid hydrogels. J. Appl. Polym. Sci. 2012, 123, 763–769. [Google Scholar] [CrossRef]
  51. Gupta, N.V.; Shivakumar, H.G. Investigation of swelling behavior and mechanical properties of a pH-sensitive superporous hydrogel composite. Iran. J. Pharm. Res. 2012, 11, 481–493. [Google Scholar] [PubMed]
  52. Bukhari, S.M.H.; Khan, S.; Rehanullah, M.; Ranjha, N.M. Synthesis and Characterization of Chemically Cross-Linked Acrylic Acid/Gelatin Hydrogels: Effect of pH and Composition on Swelling and Drug Release. Int. J. Polym. Sci. 2015, 2015, 187961. [Google Scholar] [CrossRef]
  53. Azad, A.K.; Al-Mahmood, S.M.A.; Chatterjee, B.; Wan Sulaiman, W.M.A.; Elsayed, T.M.; Doolaanea, A.A. Encapsulation of black seed oil in alginate beads as a pH-sensitive carrier for intestine-targeted drug delivery: In vitro, in vivo and ex vivo study. Pharmaceutics 2020, 12, 219. [Google Scholar] [CrossRef] [Green Version]
  54. Mirzaei, B.E.; Ramazani, S.A.A.; Shafiee, M.; Danaei, M. Studies on glutaraldehyde crosslinked chitosan hydrogel properties for drug delivery systems. Int. J. Polym. Mater. Polym. Biomater. 2013, 62, 605–611. [Google Scholar] [CrossRef]
  55. Suhail, M.; Hsieh, Y.-H.; Khan, A.; Minhas, M.U.; Wu, P.-C. Preparation and In Vitro Evaluation of Aspartic/Alginic Acid Based Semi-Interpenetrating Network Hydrogels for Controlled Release of Ibuprofen. Gels 2021, 7, 68. [Google Scholar] [CrossRef] [PubMed]
  56. Suhail, M.; Khan, A.; Rosenholm, J.M.; Minhas, M.U.; Wu, P.-C. Fabrication and Characterization of Diclofenac Sodium Loaded Hydrogels of Sodium Alginate as Sustained Release Carrier. Gels 2021, 7, 10. [Google Scholar] [CrossRef]
  57. Akhlaq, M.; Azad, A.K.; Fuloria, S.; Meenakshi, D.U.; Raza, S.; Safdar, M.; Nawaz, A.; Subramaniyan, V.; Sekar, M.; Sathasivam, K.V.; et al. Fabrication of Tizanidine Loaded Patches Using Flaxseed Oil and Coriander Oil as a Penetration Enhancer for Transdermal Delivery. Polymers 2021, 13, 4217. [Google Scholar] [CrossRef] [PubMed]
  58. Azad, A.K.; Doolaanea, A.A.; Al-Mahmood, S.M.A.; Kennedy, J.F.; Chatterjee, B.; Bera, H. Electro-hydrodynamic assisted synthesis of lecithin-stabilized peppermint oil-loaded alginate microbeads for intestinal drug delivery. Int. J. Biol. Macromol. 2021, 185, 861–875. [Google Scholar] [CrossRef]
  59. Akhlaq, M.; Idrees, N.; Nawaz, A.; Jalil, A.; Zafar, N.; Adeel, M.; Afridi, H.H. HPMC-coacrylic acid dexibuprofen once-daily oral hydrogels. J. Macromol. Sci. Part A 2020, 57, 663–674. [Google Scholar] [CrossRef]
  60. Ahmad, U.; Sohail, M.; Ahmad, M.; Minhas, M.U.; Khan, S.; Hussain, Z.; Kousar, M.; Mohsin, S.; Abbasi, M.; Shah, S.A.; et al. Chitosan based thermosensitive injectable hydrogels for controlled delivery of loxoprofen: Development, characterization and in-vivo evaluation. Int. J. Biol. Macromol. 2019, 129, 233–245. [Google Scholar] [CrossRef]
  61. Ranjha, N.M.; Qureshi, U.F. Preparation and characterization of crosslinked acrylic acid/hydroxypropyl methyl cellulose hydrogels for drug delivery. Int. J. Pharm. Pharm. Sci. 2014, 6, 400–410. [Google Scholar]
  62. Peppas, N.A.; Hilt, J.Z.; Khademhosseini, A.; Langer, R. Hydrogels in Biology and Medicine: From Molecular Principles to Bionanotechnology. Adv. Mater. 2006, 18, 1345–1360. [Google Scholar] [CrossRef]
  63. Peppas, N.A.; Bures, P.; Leobandung, W.; Ichikawa, H. Hydrogels in pharmaceutical formulations. Eur. J. Pharm. Biopharm. 2006, 50, 27–46. [Google Scholar] [CrossRef]
  64. Peppas, N.A.; Mikos, A.G. Preparation methods and structure of hydrogels. In Hydrogels in Medicine and Pharmacy; Peppas, N.A., Ed.; CRC Press: Boca Raton, FL, USA, 1986; Volume 1, pp. 2–23. [Google Scholar]
  65. Amini-Fazl, M.S.; Mohammadi, R.; Kheiri, K. 5-Fluorouracil loaded chitosan/polyacrylic acid/Fe3O4 magnetic nanocomposite hydrogel as a potential anticancer drug delivery system. Int. J. Biol. Macromol. 2019, 132, 506–513. [Google Scholar] [CrossRef]
  66. Ranjha, N.M.; Mudassir, J.; Akhtar, N. Methyl methacrylate-co-itaconic acid (MMA-co-IA) hydrogels for controlled drug delivery. J. Sol-Gel Sci. Technol. 2008, 47, 23–30. [Google Scholar] [CrossRef]
  67. Reddy, P.R.S.; Rao, K.S.V.K.; Rao, K.M.; Reddy, N.S. Synthesis of Novel Hydrogels based Poly(4-Hydroxyphenylazo-3-N-(4-hydroxyphenyl)maleimide) for Specific Colon Delivery of Chemotherapeutic Agent. J. Appl. Pharm. Sci. 2015, 5, 021–028. [Google Scholar] [CrossRef] [Green Version]
  68. Feng, L.; Yang, H.; Dong, X.; Lei, H.; Chen, D. pH-sensitive polymeric particles as smart carriers for rebar inhibitors delivery in alkaline condition. J. Appl. Polym. Sci. 2018, 135, 45886. [Google Scholar] [CrossRef]
Scheme 1. General mechanism for polymerization of AA and AM in the presence of glutaraldehyde cross-linker [31].
Scheme 1. General mechanism for polymerization of AA and AM in the presence of glutaraldehyde cross-linker [31].
Gels 08 00047 sch001
Figure 1. ATR-FTIR spectra of (a) Pure 5-FU, (b) 5-FULH, and (c) Unloaded hydrogels.
Figure 1. ATR-FTIR spectra of (a) Pure 5-FU, (b) 5-FULH, and (c) Unloaded hydrogels.
Gels 08 00047 g001
Figure 2. Effect of AM on dynamic swelling of 5-FULH.
Figure 2. Effect of AM on dynamic swelling of 5-FULH.
Gels 08 00047 g002
Figure 3. Effect of AM on equilibrium swelling of 5-FULH.
Figure 3. Effect of AM on equilibrium swelling of 5-FULH.
Gels 08 00047 g003
Figure 4. Effect of AA on dynamic swelling of 5-FULH.
Figure 4. Effect of AA on dynamic swelling of 5-FULH.
Gels 08 00047 g004
Figure 5. Effect of AA on equilibrium swelling of 5-FULH.
Figure 5. Effect of AA on equilibrium swelling of 5-FULH.
Gels 08 00047 g005
Figure 6. Effect of GA over dynamic swelling of 5-FULH.
Figure 6. Effect of GA over dynamic swelling of 5-FULH.
Gels 08 00047 g006
Figure 7. Effect of GA over the equilibrium swelling of 5-FULH.
Figure 7. Effect of GA over the equilibrium swelling of 5-FULH.
Gels 08 00047 g007
Figure 8. At pH 1.2, effect of AM on 5-FU release.
Figure 8. At pH 1.2, effect of AM on 5-FU release.
Gels 08 00047 g008
Figure 9. At pH 7.4, effect of AM on 5 FU release profile.
Figure 9. At pH 7.4, effect of AM on 5 FU release profile.
Gels 08 00047 g009
Figure 10. Effect of AA on the release profile of 5-FU at pH 1.2.
Figure 10. Effect of AA on the release profile of 5-FU at pH 1.2.
Gels 08 00047 g010
Figure 11. Effect of AA on the release profile of 5-FU at pH 7.4.
Figure 11. Effect of AA on the release profile of 5-FU at pH 7.4.
Gels 08 00047 g011
Figure 12. Effect of GA on the release profile of 5-FU at pH 1.2.
Figure 12. Effect of GA on the release profile of 5-FU at pH 1.2.
Gels 08 00047 g012
Figure 13. Effect of GA over 5-FUrelease profile at pH 7.4.
Figure 13. Effect of GA over 5-FUrelease profile at pH 7.4.
Gels 08 00047 g013
Table 1. Solubility study of 5-FU at 37 °C.
Table 1. Solubility study of 5-FU at 37 °C.
S.NoSolventRemarks
1Distilled WaterSoluble
2Phosphate buffer pH 7.4Soluble
3Acidic media pH 1.2Sparingly soluble
Table 2. % Dynamic and equilibrium swelling study of 5-FULH.
Table 2. % Dynamic and equilibrium swelling study of 5-FULH.
F. CodepH 1.2pH 6.8pH 7.4
Dynamic SwellingEquilibrium SwellingDynamic SwellingEquilibrium SwellingDynamic SwellingEquilibrium Swelling
A10.98 ± 0.041.65 ± 0.011.85 ± 0.0110.12 ± 0.024.33 ± 0.0429.67 ± 0.05
A20.99 ± 0.032.98 ± 0.033.89 ± 0.0316.27 ± 0.035.99 ± 0.0339.68 ± 0.03
A31.32 ± 0.014.11 ± 0.054.13 ± 0.0528.15 ± 0.017.71 ± 0.0347.98 ± 0.02
B10.99 ± 0.023.55 ± 0.023.35 ± 0.0212.89 ± 0.045.77 ± 0.0233.46 ± 0.07
B21.21 ± 0.045.02 ± 0.015.02 ± 0.0117.65 ± 0.057.44 ± 0.0347.96 ± 0.05
B31.43 ± 0.026.88 ± 0.036.68 ± 0.0327.89 ± 0.0310.21 ± 0.0155.48 ± 0.04
C11.90 ± 0.033.99 ± 0.014.99 ± 0.0118.67 ± 0.026.88 ± 0.0242.78 ± 0.06
C21.33 ± 0.014.64 ± 0.023.14 ± 0.0216.74 ± 0.016.10 ± 0.0334.53 ± 0.05
C31.10 ± 0.013.45 ± 0.042.05 ± 0.0411.87 ± 0.045.66 ± 0.0222.25 ± 0.03
Table 3. Diffusion coefficient, porosity %, gel fraction %, and sol fraction % of formulated 5-FULH.
Table 3. Diffusion coefficient, porosity %, gel fraction %, and sol fraction % of formulated 5-FULH.
F. CodeDC (cm2/s)Porosity %Gel Fraction %Sol Fraction %
A10.19 ± 0.0234 ± 0.0374 ± 0.424 ± 0.1
A20.09 ± 0.0342 ± 0.0486 ± 0.322 ± 0.2
A30.05 ± 0.0155 ± 0.0290 ± 0.415 ± 0.1
B10.08 ± 0.0141 ± 0.0385 ± 0.517 ± 0.3
B20.07 ± 0.0343 ± 0.0491 ± 0.311 ± 0.2
B30.03 ± 0.0262 ± 0.0694 ± 0.209 ± 0.3
C10.15 ± 0.0139 ± 0.0583 ± 0.416 ± 0.4
C20.09 ± 0.0339 ± 0.0489 ± 0.411 ± 0.4
C30.07 ± 0.0247 ± 0.0393 ± 0.309 ± 0.3
Table 4. Amount of 5-Flurouracil loaded in different formulations of acrylamide/acrylic acid hydrogels.
Table 4. Amount of 5-Flurouracil loaded in different formulations of acrylamide/acrylic acid hydrogels.
F. CodeAmount of 5-FU Loaded (g/g of Dry Gel)
By SwellingBy ExtractionBy Weight
A10.03940.03730.0347
A20.04020.03920.0369
A30.04160.04230.0391
B10.03720.03930.0357
B20.03490.03850.0341
B30.03310.03770.0328
Table 5. Composition of formulated 5-FULH.
Table 5. Composition of formulated 5-FULH.
F. CodeAMAAGATEMEDAPSD/Water
A14.08 g0.48 g23 µL235 µL0.20 g13 mL
A25.88 g0.48 g23 µL235 µL0.20 g13 mL
A37.5 g0.48 g23 µL235 µL0.20 g13 mL
B14.08 g0.96 g23 µL235 µL0.20 g13 mL
B24.08 g1.5 g23 µL235 µL0.20 g13 mL
B34.08 g2 g23 µL235 µL0.20 g13 mL
C14.08 g0.48 g19 µL235 µL0.20 g13 mL
C24.08 g0.48 g13 µL235 µL0.20 g13 mL
C34.08 g0.48 g7 µL235 µL0.20 g13 mL
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Khan, M.A.; Azad, A.K.; Safdar, M.; Nawaz, A.; Akhlaq, M.; Paul, P.; Hossain, M.K.; Rahman, M.H.; Baty, R.S.; El-kott, A.F.; et al. Synthesis and Characterization of Acrylamide/Acrylic Acid Co-Polymers and Glutaraldehyde Crosslinked pH-Sensitive Hydrogels. Gels 2022, 8, 47. https://doi.org/10.3390/gels8010047

AMA Style

Khan MA, Azad AK, Safdar M, Nawaz A, Akhlaq M, Paul P, Hossain MK, Rahman MH, Baty RS, El-kott AF, et al. Synthesis and Characterization of Acrylamide/Acrylic Acid Co-Polymers and Glutaraldehyde Crosslinked pH-Sensitive Hydrogels. Gels. 2022; 8(1):47. https://doi.org/10.3390/gels8010047

Chicago/Turabian Style

Khan, Munir Ahmad, Abul Kalam Azad, Muhammad Safdar, Asif Nawaz, Muhammad Akhlaq, Pijush Paul, Md. Kamal Hossain, Md. Habibur Rahman, Roua S. Baty, Attalla F. El-kott, and et al. 2022. "Synthesis and Characterization of Acrylamide/Acrylic Acid Co-Polymers and Glutaraldehyde Crosslinked pH-Sensitive Hydrogels" Gels 8, no. 1: 47. https://doi.org/10.3390/gels8010047

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop