Open access peer-reviewed chapter

Pharmaceutical Applications of Pectin

Written By

Olufunke D. Akin-Ajani and Adenike Okunlola

Submitted: 20 August 2021 Reviewed: 26 August 2021 Published: 16 September 2021

DOI: 10.5772/intechopen.100152

From the Edited Volume

Pectins - The New-Old Polysaccharides

Edited by Martin Alberto Masuelli

Chapter metrics overview

591 Chapter Downloads

View Full Metrics

Abstract

Pectin, a natural ionic polysaccharide found in the cell wall of terrestrial plants undergoes chain–chain association to form hydrogels upon addition of divalent cations. Based on its degree of esterification, pectin has been classified into two main types. The high methoxyl pectin with a degree of esterification greater than 50%, which is mainly used for its thickening and gelling properties and the low methoxyl pectin, which is widely used for its low sugar-content in jams, both applications being in the food industry. Pectin is mostly derived from citrus fruit peels, but can also be found in other plants such as waterleaf leaves, cocoa husk, and potato pulps. Pectin has been used as an excipient in pharmaceutical formulations for various functions. This chapter will focus on the various applications to which pectin has been used in the pharmaceutical industry.

Keywords

  • Pectin
  • Degree of Esterification
  • Drug Delivery
  • Polymer Matrix
  • Excipients

1. Introduction

In the pharmaceutical industry, plants and plant products are continually used as sources of drugs and excipients. In particular, plant-derived polymers have contributed significant roles in drug delivery systems where they function as excipients [1]. Excipients refer to the non-pharmacological ingredients that are required to convert the Active Pharmaceutical Ingredient (API) into a dosage form. The International Pharmaceutical Excipients Council (IPEC, 1995) defines excipients as all substances contained in a dosage form other than the active substance or finished dosage form, which have been appropriately evaluated for safety and are included in a drug delivery system [2]. Excipients are included in drug delivery systems to assist in processing during manufacture, protect, support, enhance stability, bioavailability and patient acceptability, help in product identification, or enhance any other aspects of the drug delivery system’s overall safety and effectiveness during use or storage [2, 3, 4]. Far from being just a random combination of ingredients, a pharmaceutical formulation is a well-rationalized formulation designed to satisfy quality and performance. Excipients are essential in the drug development process, as well as the formulation and administration of stable dosage forms [2]. Excipients are required in drug formulations to guarantee the potency, safety, predictability and reproducibility of the release of the API as well as its palatability and suitability for the patients [3].

The interest in excipients of plant origin over semi-synthetic or synthetic excipients is not far-fetched: low toxicity, relative abundance, cost-effectiveness and non-irritant nature make them preferable to others sources [5]. Plant-based polymeric excipients can be used in different pharmaceutical formulations where they act as diluents or bulking agents, thickeners, binders, disintegrants, suspending agents, emulsifiers, film formers, matrix formers, release modifiers, sweeteners and mucoadhesive polymers [6, 7, 8, 9]. These natural polymers would have to fulfill the requirements of an ideal excipient to be successful candidates for use as excipients in various formulations for pharmaceutical use. The requirements for an ideal excipient includes being pharmacologically inert, non-toxic and non-irritant as well as being non-reactive with drug or with other substances present in the formulation and the packaging. In addition, they must be easy to handle, cost-effective and readily available for the sustainable manufacture of the pharmaceutical product. Numerous plant polymers fulfill many of these requirements and have found application in pharmaceutical formulations. These include Inulin; a polysaccharide obtained from plant sources like; onion, garlic, artichoke and chicory, starches which are polymeric carbohydrates with large glucose units joined by glycosidic bonds, gums, and mucilage such as: acacia gum, tragacanth gum, locust bean gum, okra mucilage, seaweed polysaccharides which include carrageenan, agar and alginates, microbial polysaccharides such as: xanthan gum and pullulan obtained by the fermentation of carbohydrate products by specific bacteria or fungus, and polysaccharides of the plant cell wall with cellulose, hemicelluloses, pectin being the main polymers of this group [10, 11, 12, 13, 14, 15, 16].

Pectin, a structural heteropolysaccharide, is considered the second most abundant component of the cell wall of all terrestrial plants [17, 18]. It is a hydrophilic polymer that is biodegradable, biocompatible and non-toxic, making it a good biomaterial for packaging, coating and various pharmaceutical applications. Pectin is normally produced during the initial stages of growth of the primary cell wall and constitutes about one-third of the dry substance of the cell wall of some monocotyledonous and dicotyledonous plants [19]. A white to light brown powder, pectin is found in numerous fruits and vegetables. The main raw materials for pectin production are dried citrus peels or apple pomace, both by-products of juice production that are often discarded as waste. Alternative sources of pectin extraction include sugar beet waste from sugar manufacturing, mango waste from mango canning factories and sunflower seeds used for extracting edible oil, waterleaf leaves, cocoa husk, and potato pulps [20, 21, 22, 23].

Pectin is the methylated ester of polygalacturonic acid which contains 1, 4-linked α-D-galacturonic acid residues and a variety of neutral sugars like arabinose, galactose, rhamnose and lesser amounts of other sugars [24, 25]. It can be classified into different types based on the degree of esterification or the number of methoxy groups that substitutes the carboxylic acid moiety on the galacturonic acid residues [26]. The degree of esterification influences the gelation mechanism, processing conditions and properties of the pectin [18, 27]. High methoxyl pectin is primarily used for gelation and has a degree of esterification greater than 50%. It requires a large amount of sugar and is acid-sensitive. Because of hydrogen bonding and hydrophobic interactions between the pectin chains, high methoxyl pectin forms a gel at low pH and a high concentration of soluble particles [28]. Low methoxyl pectin has a degree of esterification of less than 50% and is widely used in the food industry to form low sugar jams since it does not require a large amount of sugar for gelation. It shows less sensitivity toward acidity and requires Ca2+ ions to form gel [29]. Low methoxyl pectin is generally formed by the de-esterification of high methoxy pectin using acids, alkali, pectin methylesterase and ammonia in alcohol or concentrated aqueous ammonia. Monovalent cation i.e. alkali metal salts of pectin is normally soluble in water while di- and trivalent cations are partially or completely insoluble in water. When dissolved, pectin decomposes rapidly by de-esterification or depolymerization. The rate of decomposition depends on the pH and temperature of the solution. The maximum stability of pectin is at pH 4 [30]. Low pH and high temperature increase the rate of degradation due to hydrolysis of the glycosidic linkage. At alkaline pH, pectin is rapidly de-esterified and degraded even at room temperature [31].

In this Chapter, the sections that follow would review in detail, some important pharmaceutical applications of pectin and possible modifications to enhance the future uses of pectin in pharmaceutical formulations.

Advertisement

2. Pharmaceutical uses of pectin

2.1 Drug delivery systems

The polymer pectin has been put to several uses since its discovery over 200 years ago. Though its major application has been in the food industry where it has been used as a gelling agent, emulsifier, stabilizer, thickener, and more recently as a food packaging material, where they as used as edible films on fruits and vegetables etc. The most important use of pectin is based on its ability to form gels, hence its potential as an excipient; pectin has been used as a binding agent [32, 33] in tablets, carrier for drug delivery to the gastrointestinal tract from matrix tablets, and as a controlled-release matrix in tablet formulations [34, 35, 36]. It has also been used as a sustained release drug delivery system in gel beads prepared by the ionotropic gelation method [19, 37, 38], colon-specific drug delivery vehicle [39], and film-coated dosage forms. Gel formation is caused by hydrogen bonding between free carboxyl groups on the pectin molecules and between the hydroxyl groups of neighboring molecules [40]. Most of the unesterified carboxyl groups in pectin occur as partially ionized salts in a neutral or very slightly acid dispersion of pectin molecules. [41]. Those that are ionized produce a negative charge on the molecule, which together with the hydroxyl groups causes it to attract layers of water [38]. Because of their negative charge, the repulsive forces between these groups can be strong enough to preclude the creation of a pectin network. When acid is added, the carboxyl ions are converted to mostly unionized carboxylic acid groups [38]. The attraction between pectin and water molecules is lowered by a reduction in the number of negative charges, which also lowers the forces of repulsion between pectin molecules. Sugar further decreases the hydration of the pectin by competing for water [41]. These conditions decrease the ability of pectin to stay dispersed. When cooled, the unstable dispersion of less hydrated pectin forms a gel, a continuous network of pectin holding the aqueous solution. High methoxyl pectin produces gels with sugar and acid. Unlike Low methoxyl pectin, high methoxyl pectin does not contain sufficient acid groups to gel or precipitate with calcium ions, although other ions such as aluminum or copper cause precipitation under certain conditions [25]. The degree of esterification (DE) affects the rate at which gel formation takes place [38]. A higher DE causes a more rapid setting. Slow-set pectins (with DE 58–65%) gel at lower soluble solids and greater levels than rapid-set pectins (DE > 72 per cent). Low methoxyl pectins require the presence of divalent cations (usually calcium) for proper gel formation [38].

2.2 Bioadhesive systems

The ability of pectin to absorb water, swell and form bioadhesive bonds with biological tissue has found application in the preparation of mucoadhesive formulations such as patches [42]. Pectin has also been found useful as a demulcent in throat lozenges where it gives temporary relief for minor discomfort and protects irritated areas in sore mouth and sore throat [43]. The antihemorrhagic effect of pectin has also been utilized in wound healing as medical adhesives [44].

2.3 Disperse systems

Pectin has been shown to have foam stabilizing and emulsification potential since the protein and hydrophobic acetyl groups of pectin can act as anchors on the oil particle surface, thus decreasing the surface tension [45]. Other areas of use have been as an emulsifier in oil: water emulsions [46, 47], and as a viscosity enhancer in lipid digests [48]. Pectin slows gastric transit thus helps control energy intake and hence its use by weight-watchers, since the large water-binding capacity of pectin reduces contact between intestinal enzymes and food, thus prolonging gastric emptying half-life, allowing a marked reduction in quantity and frequency of eating [49]. Furthermore, its interaction with polyphenolic compounds leads to systemic anti-inflammation [50].

2.4 Health benefits

In the pharmaceutical industry, pectin has been used both for its health benefits and as an excipient. Pectin as an active agent was formerly used in diarrhea mixtures, in conjunction with kaolin and sometimes bismuth compounds, and in wound dusting powders and ulcer dressings where pectin appears to have some specific activity in promoting healing [25]. It has been found to have certain health benefits such as reducing cancer development, lowering blood cholesterol and blood glucose level through the different domains of the pectin structure, and stimulating the immune response [51, 52, 53, 54, 55].

2.5 Other applications

Pectin’s application has spread to water treatment where it is used as a biosorbent to remove heavy metals [47] and in urinary excretion of toxic minerals such as lead, cadmium, strontium, or arsenic [56, 57, 58]. In cosmetics, it is used as a plasticizer, texturizer and adhesive [59], and in biomedical applications, where it is used as a biomaterial ink to fabricate patient-specific scaffolds when cross-linked with 3-glycidyloxypropyl trimethoxysilane (GPTMS) [60]. Some examples of drug products that contain pectin are presented in Table 1.

Brand NameIngredients/strengthDosage formManufacturer
Berry Breezer Throat Drop7 mg/1LozengeTopco Associates USA
Burts Bees Throat Soothing10.5 mg/1LozengeL. Perrigo Company USA
CVS Clean Label Throat Relief Pops12 mg/1LozengeCVS Pharmacy USA
Grape Throat Relief Lollipop10 mg/1LozengeTopco Associates USA
Little Remedies Sore Throat Pops5.4 mg/1LozengeMedtech Products Inc. USA
Ludens Assorted Flavors2.8 mg/1LozengePrestige Brands Holdings, Inc. USA
Sundown Honey Soothers Lollipops0.1 g/100 gLozengeThe Nature’s Bounty Company USA
Throat Coat Lemon Ginger Echinacea5 mg/4.2 gLozengeTraditional Medicinals, Inc. USA
Axcel Kaopec SuspensionPectin (20 mg/5ml) + Kaolin (1 g/5ml)SuspensionKotra Pharma (M) Sdn. Bhd.
Benylin DM With Pectin Freezer PopsPectin (150 mg/unit) + Dextromethorphan hydrobromide (7.5 mg/unit)LiquidMcneil Consumer Healthcare Division of Johnson & Johnson Inc
Cepacol Sore Throat and CoatingPectin (5 mg/1) + Benzocaine (15 mg/1)LozengeReckitt Benckiser
Cepacol Sore Throat Plus CoatingPectin (5 mg/1) + Benzocaine (15 mg/1)LozengeCombe Incorporated
Diaret LiqPectin (150 mg/30 mL) + Kaolin (3.078 g/30 mL)LiquidProduits Francais Labs Inc. Canada
Diaret TabPectin (45 mg/tab) + Aluminum hydroxide (70 mg/tab) + Attapulgite (350 mg/tab) + Zinc phenolsulfonate (30 mg/tab)TabletProduits Francais Labs Inc. Canada
Diban CapPectin (71.4 mg) + Atropine sulfate anyhdrous (9.7 mcg) + Attapulgite (300 mg) + Hyoscyamine sulfate (0.0519 mg) + Opium (12 mg) + Scopolamine (3.3 mcg)CapsuleWyeth Ayerst Canada Inc.
Orabase PastePectin (13.3%) + Carboxymethylcellulose sodium (13.3%) + Gelatin (13.3%)PasteConvatec Inc.
Organix CompletePectin (1.7 mg/1) + Levomenthol (2.5 mg/1)LozengePro Phase Labs, Inc.
Herbon Berry BuddiesPectin (10 mg/1) + Echinacea purpurea (50 mg/1)LozengePurity Life Division of SunOpta

Table 1.

Some commercial drug products containing pectin.

Advertisement

3. Material properties of pectin

Pectin is an important biomaterial that has numerous pharmaceutical applications. Its application, however, largely depends on its material properties such as degree of esterification (DE), degree of blockiness (DB), ash value and solubility. This section will focus on these properties and how each affects the application of pectin pharmaceutically.

3.1 Degree of esterification

The DE of pectin is the ratio of esterified D-galacturonic acid (GalA) groups to total GalA groups [34, 61]. Depending on the species, tissue, and maturity of the plant, the DE can have a wide range. In general, the structure of pectin is mostly composed of homogalacturonan (HG), regions (partially 6-methylated and 2- and/or 3-acetylated poly-α(1–4)-D-galacturonic acid residues), alternating with rhamnogalacturonan I (RG-I), regions (branched α(1–2)-L-rhamnosyl-α(1–4)-D-galacturonosyl chains substituted with side chains of mainly α-L-arabinofuranose and α-D-galactopyranose) [18, 62]. The interconnection of HG “smooth” (responsible for the gelling capability) and RG-I “hairy” (play a gel-stabilizing role) regions, in relative proportions determine the flexibility and rheological properties of the polymer in solution [63, 64]. The gelling mechanism of pectin is dictated by its degree of esterification (total methoxyl content) [65]. Pectin based on the DE can be classified as high methoxyl (HM) pectin with DE > 50% or low methoxyl (LM) pectin DE < 50%, which are either the conventionally demethylated or the amidated molecule [66, 67, 68]. The two groups of pectin gel by different mechanisms. To form gels, high methoxyl pectin requires a minimum amount of soluble solids and a pH of around 3.0.

HM pectins are generally hot water-soluble, thermally reversible, and often contain dextrose (a dispersion agent) to prevent lumping. Conversely, LM pectins produce gels independent of sugar content, are less sensitive to pH compared to the HM pectins, and require the presence of a controlled amount of calcium or other divalent cations for gelation [41].

The specific application to which pectin will be put is a function of its gelling behavior, which is dependent on its DE, the monosaccharide content (HG), and the spatial disposition of the cross-linking blocks (RG) [69]. While HM pectins have been used in tablet formulations as a binder, controlled-release matrix and taste masker through complexation with bitter molecules, the LM pectins have been used as sustained-release matrices in microspheres produced by ionotropic gelation [19, 53, 69].

3.2 Degree of blockiness

Pectin, an anionic cell wall polysaccharide through its non-methyl esterified galacturonic acid units, interacts with divalent cations [40, 47]. At pH values above the pKa of pectin (2.8 to 4.1), non-methyl esterified GalA residues can be negatively charged, giving pectin the ability to interact with cations [34, 70]. Thus, the lower the DE of pectin, the higher the number of non-methyl esterified GalA residues present, the higher the cation-binding capacity. Due to LM pectin’s higher number of negatively chargeable carboxyl groups (non-methyl esterified carboxyl groups) compared to HM pectin, it exhibits a higher charge density, further showing that the cation-binding capacity of pectin increases with decreasing DE [40, 70, 71]. Studies have shown that regardless of the method used, a stronger and higher bound interaction occurs between pectin with decreasing DE and cations (Fe2+, Zn2+, or Ca2+) [47]. Furthermore, the DE and the intramolecular distribution of the non-methyl esterified carboxyl groups within the pectin determine pectin’s anionic nature and associated functionality [72, 73]. Interestingly, less described in the literature is the influence of the distribution pattern of non-methyl esterified GalA units on the cation-binding capacity of pectin compared to DE [47].

Daas et al. first quantified the relative occurrence of blocks of non-methyl esterified GalA units within a pectin chain as the degree of blockiness, DB [47, 74]. Apart from the DB, the absolute number of non-methyl esterified GalA units present in blocks can be expressed as the absolute degree of blockiness (DBabs). Both parameters (DB and DBabs) were established by exhaustive enzymatic degradation of pectin using endo-polygalacturonase (endo-PG) of Kluyveromyces fragilis, which required at least four consecutive non-methyl esterified GalA units to hydrolyze the linkage between two non-methyl esterified GalA units [70]. The DB is the proportion of galacturonic acid units (mono-, di-, and tri-) released by the enzyme to the total amount of non-methyl esterified GalA units, while DBabs is the number of GalA oligomers released in the endo-PG digest to the total number of GalA units in the pectin polymer, without adjustment of the DE [47, 74, 75]. Thus, to characterize the presence of blocks of non-methyl esterified GalA units, these parameters (DB and DBabs) are used [70]. For most of the cations (divalent cations), the binding between them and pectin is known to follow the egg-box model [47]. The egg-box model of ´binding was mainly described for pectin-Ca2+ binding but assumed to be applicable for interaction between pectin and other divalent cations [76]. However, Assifaoui et al. [77] reported that the egg-box model was more appropriate for Zn2+ binding than Ca2+ as they found that Zn2+ interacts with both carboxyl and hydroxyl groups, comparable to the egg-box model, whereas Ca2+ binds only via carboxyl groups [40]. This egg-box model yields stronger gels [78, 79].

Applications to which a high DB is required would thus mean high cation-binding capacity and hence the use of LM pectins and the converse is true.

3.3 Ash value

The ash content of pectin is a valuable tool in determining the purity as well as the gel-forming capability of the polymer. The ash content of pectin has been found to increase as the yield of pectin decreases [80]. High levels of ash in pectin may be caused by elevated concentrations of negatively charged carboxylic groups of pectin and the counterions in solution during pectin precipitation [41]. However, for gel formation, low ash content (≤ 10%) is a more favorable criterion as this will aid in determining the applicability of the polymer [47, 81, 80]. Ash content along with the anhydrouronic acid value of pectin has also been used to determine its purity [82, 83].

3.4 Solubility

Pectins are soluble in pure water. The solubility appears to depend on the valency of the cation salt; monovalent cation salts of pectin and pectic acids are usually soluble in water, while the di- and trivalent cation salts are weakly soluble or insoluble in water. Dry powdered pectin hydrates very rapidly when added to water, but tends to form clumps. These clumps are semidry packets of pectin within a highly hydrated outer coating. Dry mixing the powder with water-soluble carrier material can prevent the formation of clumps or by the use of specially treated pectin that has improved dispersibility [20, 83]. Studies have shown that pectin extracted with distilled water showed a high yield and low ash content when compared to other solvents [79]. High ash content and the drying process of the extracted pectin, however, may reduce the solubility of pectin [47]. It has been shown too that a decrease in the esterified carboxylic group reduced the solubility of extracted pectin; this insolubility of the extracted pectin is probably due to the presence of electrolytes in de-methylated pectic acid [47]. Thus, pectins with lower DE are less hydrophilic [69].

Dilute pectin solutions are Newtonian in behavior but at a moderate concentration, they exhibit the non-Newtonian, pseudo plastic behavior characteristics. Solubility, viscosity, and gelation are generally related. Whatever factors increase gel strength will increase the gelling tendency, viscosity, decrease solubility, and vice versa [84].

3.5 Antioxidant activity

Another property of pectin that could affect its application is its antioxidant activity. However, there are limited studies to show how this property may be applied to either the food industry or the pharmaceutical sector [85].

Advertisement

4. Modifications of pectin for future applications

The presence of several hydroxyl and carboxyl groups distributed along its backbone as well as a certain amount of neutral sugars presented as side chains gives pectin the capability of producing a broad spectrum of derivatives with modified or new functional properties. Various methods used for pectin modification include substitution (alkylation, amidation, quaternization, thiolation, sulfation, oxidation, etc.), chain elongation (cross-linking and grafting) and depolymerization (chemical, physical, and enzymatic degradation). Saponification (a process catalyzed by mineral acids, bases, salts of weak acids and primary aliphatic amines) can also be used to modify pectin chemically. Modification induced by pH changes can produce new fragments that have their solubility and biological activities altered [86]. Enzymatic modification of pectin has been achieved by using endo-polygalacturonase (Endo-PG), resulting in highly selective and specific structural changes in the polymer backbone. This modification leads to the cleavage of glycosidic linkages between two non-esterified α-D-galacturonic acid residues inside the HG fragment, which is depolymerisation. The enzymatic modification method can alter the macromolecular structure of pectin and can yield modified pectin with newer and improved properties and functionalities [87].

A new hydrolyzed polyacrylamide-graft-sodium alginate (PAAmg-SA) and diclofenac sodium-loaded interpenetrating polymer network (IPN) beads of pectin were developed using the ionic gelation method. The results of the investigation verified that hydrolyzed PAAm-g-SA and pectin cross-linked with aluminum ion (Al3+) and glutaraldehyde could form an optimal matrix material for the production of IPN beads to support the sustained release of diclofenac sodium [88]. In another study, for the nasal administration of tacrine hydrochloride (an anti-Alzheimer drug), mucoadhesive microparticles based on chitosan/pectin polyelectrolyte complexes were prepared. The microparticles were produced by spray drying followed by lyophilization and direct spray drying. The study thus demonstrated the potential of the chitosan/pectin polyelectrolyte complexes to function variously in mucoadhesive microparticles [89, 90]. The chitosan/pectin molar ratio influenced the water uptake and tacrine hydrochloride permeation [90, 91].

Emerging advanced manufacturing technology in the field of tissue engineering and pharmaceutical formulations is the use of 3D bioprinting technology. 3D printing is an additive manufacturing technology in which objects are constructed in a layer-by-layer manner achieved by heat fusion, ultraviolet light (UV), and chemical bonding [91]. Spritam®, a fast disintegrating orodispersible tablet containing levetiracetam for epilepsy was the first 3D printed drug product approved by the US Food and Drug Administration (FDA) in 2015 [91]. To sustain the manufacturing of these types of drugs using this new technique, biomaterials that are green and non-toxic, derived from renewable sources and can be processed through 3D bioprinting are being developed [42, 60]. Common techniques include powder bed printing, vat polymerization (VP), and fused deposition modeling (FDM) [92]. A major disadvantage of the FDM technology is the need to insert printing materials into a nozzle in the form of a solid filament, which is non-existing for many pharmaceutical materials, thus necessitating the transformation of pharmaceutical-grade materials, including active pharmaceutical ingredients (API), into FDM-suitable filaments using techniques like hot-melt extrusion (HME). However, thermolabile therapeutics are not suitable for extrusion via FDM, due to potential degradation concerns [93]. The use of bio-inks for extrusion-based bioprinting at room or body temperature has shown clinical potential in achieving personalized treatment [92]. For example, Long et al. developed a personalized 3D printed wound dressing composed of chitosan and pectin with the ability to control dimensional properties such as thickness and pore size using an extrusion-based bioprinter [91, 92], while allowing for facile lidocaine incorporation for immediate pain relief [94]. Pectin from citrus peels has also been cross-linked with (3-glycidyloxypropyl)trimethoxysilane (GPTMS) through a one-pot procedure to obtain freeze-dried porous pectin sponges with varying porosity, water uptake, and compressive modulus [42]. The addition of GPTMS improved the printability of pectin due to an increase in viscosity and yield stress [95]. Without the use of any additional support material, three-dimensional woodpile and complex anatomical-shaped scaffolds interconnected with micro and macro pores were, therefore, bioprinted [96]. Thus showing the great potential of pectin cross-linked with GPTMS as biomaterial ink to fabricate patient-specific scaffolds that could be used to promote tissue regeneration in vivo [42]. In another study, gelatin, another natural biopolymer has had its rheological properties improved to aid its bio-printing performance by using pectin as a rheology modifier of gelatin and GPTMS as a gelatin-pectin crosslinking agent [95]. Pectin played a key role in increasing the viscosity and the yield stress of low viscous gelatin solutions as shown through investigation of the rheological properties, as well as bioprinting assessments [96]. Water stable, three-dimensional, and self-supporting gelatin-pectin-GPTMS scaffolds with interconnected micro- and macro- porosity were successfully obtained by combining extrusion-based bioprinting and freeze-drying which did not require any additional temperature control to further modulate the rheological properties of gelatin solutions [95]. Patient-centric dosage forms have been produced through additive manufacturing techniques, which enable its design with precise control over dimension and microstructure, factors that are known to ultimately play key roles in modulating drug release kinetics, a feat not achieved through compression; traditional manufacturing techniques [92, 96, 97].

Advertisement

5. Conclusions

Pectin over the years has “metamorphosed” from just being a gelling agent for the production of jam and confectionaries to a biomaterial with health benefits to being useful as an excipient in drug delivery systems and more recently even personalized 3D printed medicine. This is as a result of a better understanding of its structure, mechanism by which it gels, and its properties such as degree of esterification and degree of blockiness, which has aided its classification and application.

The pharmaceutical industry has a material that can be explored in different functional dimensions as the usefulness and functionality of pectin unfolds.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Nayak A, Olatunji O, Das DB, Vladvisavljevic G. Pharmaceutical Applications of Natural Polymers. Natural Polymers. 2015; 263-313|. DOI: 10.1007/978-3-319-26414-1-9
  2. 2. Furrer P. The central role of excipients in drug formulation [Internet]. 2013. Available from https://www.europeanpharmaceuticalreview.com/article/18434/the-central-role of-excipients-in-drug-formulation-2/ [Accessed: 2021-08-14]
  3. 3. Ogaji IJ, Nep EI, Peter JDA. Advances in natural polymers as pharmaceutical excipients. Pharm Anal Acta 2011; 3:1-16
  4. 4. Robertson MI. Regulatory issues with excipients International Journal of Pharmaceutics. 1999; 187(2): 273-276. DOI: 10.1016/S0378-5173(99)00110-6
  5. 5. Reddy SB, Ponnamma D, Choudhary R and Sadasivuni KK. A Comparative Review of Natural and Synthetic Biopolymer Composite Scaffolds. Polymers 2021, 13:1105:1-51. DOI: 10.3390/polym13071105
  6. 6. Kulkarni GT, Gowthamarajan K, Dhobe FY, Suresh B. Development of controlled release spheroids using natural polysaccharide as release modifier. Drug Deliv 2005; 12:201-206
  7. 7. Beneke CE, Viljoen AM, and Hamman JH. Polymeric Plant-derived Excipients in Drug Delivery. Molecules. 2009; 14(7):2602-2620. DOI: 10.3390/molecules14072602
  8. 8. Seki H, Tamura K, Muranaka T. Plant-derived isoprenoid sweeteners: recent progress in biosynthetic gene discovery and perspectives on microbial production. Biosci Biotechnol Biochem 2018; 82(6):927-934
  9. 9. Okunlola A. Optimization of formulations of chloroquine phosphate tablets containing Ofada rice (Oryza glaberrina) starch as a binder: A Grey-Taguchi analysis. J.Excip. Food. Chem 2020; 11(3):62 – 75
  10. 10. Vervoort L, Van den Mooter G, Augustijns P, Busson R, Toppet S. Inulin hydrogels as carrier for colonic drug targeting: I. Synthesis and characterization of methacrylated inulin and hydrogel formation. Pharm Res. 1997; 14: 1730-1736
  11. 11. Okunlola A and Adedeji AT. Cross-linked plantain starch-urea as a polymer in matrix tablets of ambroxol hydrochloride. Br. J.Pharm. 2020; 5(1): 1- 13
  12. 12. Odeku OA, Patani B. Evaluation of dika nut mucilage (Irvingia gabonensis) as a binding agent in metronidazole tablet formulation. Pharm Dev Technol 2005; 10:439-446
  13. 13. Ndjouenkeu R, Akingbala J, Oguntimein G. Emulsifying properties of three African food hydrocolloids: Okra (Hibiscus esculentus), dika nut (Irvingia gabonensis) and khan (Belschmiedia sp.). Plant Foods Hum. Nutr. 1997;51:245-255
  14. 14. Balasubramaniam J, Rao VU, Vasudha M, Babu J, Rajinikanth PS. Sodium alginate microspheres of metformin HCl: Formulation and in vitro evaluation. Curr Drug Deliv 2007; 4:249-56
  15. 15. Santos H, Veiga F, Pina ME, Sousa JJ. Compaction compression and drug release properties of diclofenac sodium and ibuprofen pellets comprising xanthan gum as a sustained release agent. Int J Pharm. 2005; 295:15-27
  16. 16. Cheng K, Lim LY. Insulin-loaded calcium pectinate nanoparticles: Effects of pectin molecular weight and formulation pH. Drug Dev Ind Pharm 2004; 30:359-367
  17. 17. Noreena A, Nazlic Z, Akrama J, Rasulb I, Manshaa A, Yaqoobc N, Iqbald R, Tabasuma S, Zubera M, Ziaa KM. Pectins functionalized biomaterials; a new viable approach for biomedical applications: A review. Int. J.Bio. Macromol 2017; 101: 254-272
  18. 18. Mohnen D. Pectin structure and biosynthesis. Curr. Opin.Plant Biol. 2008; 11: 266-277
  19. 19. Okunlola, A. and Akindele, L.O. Application of response surface methodology and central composite design for the optimization of metformin microsphere formulations using tangerine (citrus tangerina) pectin as copolymer. Br.J.Pharm. Res. 2016; 11(3): 1 - 14
  20. 20. Rolin C, Pectin, In: Industrial gums, RL Whistler and JN BeMiller (eds), 1993, 3rd eds. New York: Academic Press
  21. 21. Andarwulan N, Faridah DN, Prabekti YS, Fadhilatunnur H, Mualim L, Aziz SA, Cisneros-Zevallos L. Dietary Fiber Content of Waterleaf (Talinum triangulare (Jacq.) Willd) Cultivated with Organic and Conventional Fertilization in Different Seasons. American Journal of Plant Sciences, 2015; 6:334-343. DOI:10.4236/ajps.2015.62038
  22. 22. Priyangini F, Walde SG, Chidambaram R. Extraction optimization of pectin from cocoa pod husks (Theobroma cacao L.) with ascorbic acid using response surface methodology. Carbohydr. Polym 2018; 202:497-503. DOI: 10.1016/j.carbpol.2018.08.103
  23. 23. Yang JS, Mu TH, Ma M. Extraction, structure and emulsifying properties of pectin from potato pulp. Food Chem. 2018; 244: 197 – 205
  24. 24. Hoff HE, Castro MD. Chemical composition of the potato cell wall. J. Agric. Food Chem. 1969; 17: 1328-1331
  25. 25. May CD, Handbook of Hydrocolloids, in: P.A. Williams, G.O. Phillips (Eds.),Woodhead Publishing Limited, Cambridge, UK, 2003, p. 169-188
  26. 26. Scheller HV, Jensen JK, Sørensen SO, Harholt J, Geshi N. Biosynthesis of pectin Physiol. Plant. 2007; 129:283-295
  27. 27. Sriamornsak JP. Application of pectin in oral drug delivery. Expert Opin. Drug Deliv. 2011; 8:1009-1023
  28. 28. Perez, S., Rodriguez-Carvajal MA, Doco T. Doco, A complex plant cell wall polysaccharide: rhamnogalacturonan II. A structure in quest of a function. Biochimie 2003; 85: 109-121
  29. 29. Oakenfull, A. Scott, J (1984). Hydrophobic interaction in the gelation of high methoxy pectins. Food Sci. 1984; 49:1093-1098
  30. 30. Raeisi F, Raeisi E. Mini review of polysaccharide nanoparticles and drug delivery process. Advances in Applied NanoBio-Technologie. 2020; 1(2):
  31. 31. Rolin C, Pectins and their manipulation, in: C. Seymour, P. Knox (Eds.), Commercial Pectins Preparation. Oxford Blackwell Boca Raton FL; 2002. p. 222-241
  32. 32. Slany J. Evaluation of tablets with pectin as a binding agent. Farmaceuticky Obzor, 1981a;50: 491 – 498
  33. 33. Slany J et. al. Study of functional action of citrus pectins in tablets. Ceska a Slovenska Farmacie, 1981b; 30: 195-200
  34. 34. Krusteva S, Lambov N, Velinov G. Pharmaceutical investigation of a bioerodible nystatin system. Pharmazie. 1990; 45:195-197
  35. 35. Naggar VF, El-Khawas M, Ismail FA, Boraie NA. Pectin, a possible matrix for oral sustained-release preparations of water-soluble drugs. STP Pharma Sci. 1992; 2:227-234
  36. 36. Srisagul Sungthongjeen, Pornsak Sriamornsak, Tasana Pitaksuteepong, Atawit Somsiri, and Satit Puttipipatkhachorn. Effect of degree of esterification of pectin and calcium amount on drug release from pectin-based matrix tablets. AAPS PharmSciTech 2004; 5(1) Article 9 (http://www.aapspharmscitech.org)
  37. 37. Aydin Z, Akbuga J. Preparation and evaluation of pectin beads. Int JPharm 1996; 137: 133-136
  38. 38. Sriamornsak P. Chemistry of pectin and its pharmaceutical use: A review. Silpakorn University International Journal 2003; 3(1-2): 206-228
  39. 39. Wicker, L., Kim, Y., Kim M. J. Thirkield, B. Lin, Z. and Jung, J. Pectin as a bioactive polysaccharide—Extracting tailored function from less. Food Hydrocoll. 2014; 42:251-259
  40. 40. Oakenfull, D.G. The chemistry of high-methoxyl pectins. In: The chemistry and technology of pectin, ed. R.H. Walter. New York: Academic Press; 1991
  41. 41. Sundar Raj AA, Rubila S, Jayabalan R, Ranganathan TV. (2012). A review on pectin: Chemistry due to general properties of pectin and pharmaceutical uses. Scientific reports, 1: 2, 1-4
  42. 42. Kaur A and Kaur G. Mucoadhesive buccal patches based on interpolymer complexes of chitosan–pectin for delivery of carvedilol. Saudi Pharm J. 2012; 20(1): 21-27. DOI: 10.1016/j.jsps.2011.04.005
  43. 43. Srivastav P and Malviya R. Sources of pectin, extraction and its application in pharmaceutical industry – An overview. Indian J.Nat. Prod.Resour 2011; 2(1): 10 -18
  44. 44. Mishra RK, Majeed, BA and Banthia AK Development and characterization of pectin/gelatin hydrogel membranes for wound dressing. Int.J.Plast. Technol. 2011; 15(1): 82-95
  45. 45. Leroux J, Langendorff V, Schick G, Vaishnav V, Mazoyer J. Emulsion stabilizing properties of pectin. Food Hydrocoll. 2003; 17(4): 455 – 462
  46. 46. Dickinson, E. Hydrocolloids at interfaces and the influence on the properties of dispersed systems. Food Hydrocolloids, 2003; 17(1):25-39
  47. 47. Celus M, Kyomugasho C, Van Loey AM, Grauwet T, Hendrickx ME. Influence of pectin structural properties on interactions with divalent cations and its associated functionalities. Compr. Rev. Food Sci. Food Saf. 2018; 17: 1576 – 1594. DOI: 10.1111/1541-4337.12394
  48. 48. Cervantes-Paz B, Ornelas-Paz J, de J, Ruiz-Cruz S, Rios-Velasco C, Ibarra-Junquera V, Gardea-Bejar AA. Effects of pectin on lipid ´ digestion and possible implications for carotenoid bioavailability during pre-absorptive stages: A review. Food Res. Int. 2017; 99:917-927
  49. 49. Schwartz SE, Levine RA, Singh A, Scheidecker JR, Track NS. Sustained pectin ingestion delays gastric emptying. Gastroenterology. 1982; 83(4):812-817
  50. 50. Wicker L, Kim Y. Pectin and health. in Encyclopedia of Food and Health, 2016; 289-293. https://doi.org/10.1016/B978-0-12-384947-2.00532-8
  51. 51. Jackson CL, Dreaden TM, Theobald LK, Tran NM, Beal TL, Eid M, Gao MY, Shirley RB, Stoffel MT, Kumar MV, Mohnen D. 2007. Pectin Induces Apoptosis in Human Prostate Cancer Cells: Correlation of Apoptotic Function with Pectin Structure. Glycobiology. 2007; 17:805-819
  52. 52. Behall K, Reiser S. Effects of Pectin on Human Metabolism, in “Chemistry and Functions of Pectins”. In: Fishman, M.L., Ren, J.J. (Eds.).American Chemical Society, Washington, DC. 1986:p. 248-265
  53. 53. Brown L, Rosner B, Willett WW, Sacks FM. Cholesterol-Lowering Effects of Dietary Fiber: A Meta-Analysis. Am.J. Clin.Nutri. 1999; 69:30-42
  54. 54. Inngjerdingen KT, Patel TR, Chen X, Kenne L, Allen S, Morris GA., et al. Immunological and Structural Properties of a Pectic Polymer from Glinus oppositifolius. Glycobiology 2007; 17:1299-1310
  55. 55. Begum R, Azizb MG, Uddinb MB, Yusof YA. Characterization of Jackfruit (Artocarpus heterophyllus) Waste Pectin as Influenced by Various Extraction Conditions. Agriculture and Agricultural Science Procedia. 2014; 2:244 – 251
  56. 56. Braudo EE, Danilova IV, Dianova VT, Kobak VV, Plashchina IG, Sidorov EV, Bogatyrev AN. Thermodynamic approach to the selection of polyuronide sequestrants for preventive and medicinal nutrition. Die Nahrung, 1996; 40:205-208
  57. 57. Eliaz I, Hotchkiss AT, Fishman ML, Rode D. The effect of modified citrus pectin on urinary excretion of toxic elements. Phytotherapy Res. 2006; 20:859-864
  58. 58. Zhao ZY, Liang L, Fan X, Yu Z, Hotchkiss AT, Wilk BJ, Eliaz I. The role of modified citrus pectin as an effective chelator of lead in children hospitalized with toxic lead levels. Alternative Therapies. 2008; 14:34-38
  59. 59. Pe’rez-Martı’nez JD, Sa’nchez-Becerril M, Ornelas-Paz JJ, Gonza’lez-Cha’vez MM, Ibarra-Junquera V, Escalante-Minakata P. The effect of extraction conditions on the chemical characteristics of pectin from opuntia Ficus indica cladode flour. J Polym Environ. 2013; 21: 1040 – 1051. DOI: 10.1007/s10924-013-0602-2
  60. 60. Lapomarda A, De Acutis D Chiesa I, Fortunato GM, Montemurro F, De Maria C, Belmonte MM , Gottardi R, Vozzi G. Pectin-GPTMS-Based Biomaterial: toward a sustainable bioprinting of 3d scaffolds for tissue engineering application. Biomacromolecules. 2019; 21(2):319-327. DOI: 10.1021/acs.biomac.9b01332
  61. 61. Mukhiddinov Z.K., Khalikov DK, Abdusamiev F.T., Avloev C.C. Isolation and structural characterization of a pectin homo and rhamnogalacturonan. Talanta 2000; 53: 171-176
  62. 62. Presentato A, Piacenza E, Scurria A, Albanese L, Zabini F, Meneguzzo F, Nuzzo D, Pagliaro M, Martino DC, Alduina R, Ciriminna R. A New Water-Soluble Bactericidal Agent for the Treatment of Infections Caused by Gram-Positive and Gram-Negative Bacterial Strains. Antibiotics. 2020; 9: 586; DOI:10.3390/antibiotics9090586
  63. 63. Morra M, Cassinelli C, Cascardo G, Nagel MD, Volpe CD, Siboni S, Maniglio D, Brugnara M, Ceccone G, Schols HA, Ulvskov P. Effects on Interfacial Properties and Cell Adhesion of Surface Modification by Pectic Hairy Regions. Biomacromolecules. 2004; 5: 2094-2104
  64. 64. Ciriminna R, Fidalgo A, Delisi R, Tamburino A, Carnaroglio D, Cravotto G, Ilharco LM, Pagliaro M. Controlling the Degree of Esterification of Citrus Pectin for Demanding Applications by Selection of the Source ACS Omega 2017; 2:7991-7995 DOI: 10.1021/acsomega.7b01109
  65. 65. Harding SE, Tombs MP, Adams GG, Paulsen BS, Inngjerdingen KT, Barsett H. An Introduction to Polysaccharide Biotechnology; CRC Press: Boca Raton, FL, 2017; p 99-126
  66. 66. Bagherian H, Zokaee Ashtiani F, Fouladitajar A, Mohtashamy M. Comparisons between conventional, microwave- and ultrasound-assisted methods for extraction of pectin from grapefruit. Chem Eng Process Process Intensif. Elsevier B.V.; 2011; 50: 1237-1243. DOI: 10.1016/j.cep.2011.08.002
  67. 67. Brain C, Jhonston B. Spreabale honey, US patents, 4,532,143. [Internet]. US 4532143 A, 1985. p. US patents, 4,532,143 (1985). Available: http://www.lens.org/lens/patent/US_4532143_A/fulltext
  68. 68. Sayah MY, Chabir R, Benyahia H, Rodi Kandri Y, Ouazzani Chahdi F, Touzani H, et al. Yield, Esterification Degree and Molecular Weight Evaluation of Pectins Isolated from Orange and Grapefruit Peels under Different Conditions. PLoS ONE 2016; 11(9): e0161751. doi:10.1371/journal. pone.0161751
  69. 69. Yavuz-Düzgün M, Zeeb B, Dreher J, Özçelik B, Weiss J. The Impact of Esterification Degree and Source of Pectins on Complex Coacervation as a Tool to Mask the Bitterness of Potato Protein Isolates. Food Biophysics 2020; 15:376-385. DOI:10.1007/s11483-020-09631-1
  70. 70. Ralet MC, Dronnet V, Buchholt HC, Thibault JF. Enzymatically and chemically de-esterified lime pectins: Characterisation, polyelectrolyte behaviour and calcium-binding properties. Carbohydr, Res., 2001; 336:117-125
  71. 71. Thibault JF, Rinaudo M. Interactions of mono- and divalent counterions with alkali- and enzyme-deesterified pectins in salt-free solutions. Biopolymers, 1985; 24:2131-2143
  72. 72. Vincken JP, Schols HA, Oomen RJFJ, Beldman G, Visser RGF, Voragen AGJ. Pectin- The hairy thing. Evidence that homogalacturonan is a side chain of rhamnogalacturonan I. In AGJ Voragen, HA Schols, R Visser (Eds.), Advances in pectin and pectinases research. Dordrecht, The Netherlands: Kluwer Academic Publishers 2003a; p.47-59
  73. 73. Cameron RG, Kim Y, Galant AL, Luzio GA, Tzen JTC. Pectin homogalacturonans: Nanostructural characterization of methyl esterified domains. Food Hydrocoll. 2015; 47:184-190
  74. 74. Daas PJH, Meyer-Hansen K, Schols, HA, De Ruiter GA, Voragen AGJ. Investigation of the non-esterified galacturonic acid distribution in pectin with endopolygalacturonase. Carbohydrate Research. 1999; 318:135-145
  75. 75. Guillotin SE, Bakx EJ, Boulenguer P, Mazoyer J, Schols HA, Voragen, AGJ. Populations having different GalA blocks characteristics are present in commercial pectins, which are chemically similar but have different functionalities. Carbohydr. Polym. 2005; 60:391-398
  76. 76. Grant GT, Morris ER, Rees DA, Smith PJC, Thom D. Biological interactions between polysaccharides and divalent cations: The egg-box model. FEBS Letters, 1973; 32:195-198
  77. 77. Assifaoui A, Lerbret A, Uyen HTD, Neiers F, Chambin O, Loupiac C, Cousin F. Structural behaviour differences in low methoxy pectin solutions in the presence of divalent cations (Ca2+ and Zn2+): A process driven by the binding mechanism of the cation with the galacturonate unit. Soft Matter, 2015; 11:551-560
  78. 78. Fraeye I, Doungla E, Duvetter T, Moldenaers P, Van Loey A, Hendrickx M. Influence of intrinsic and extrinsic factors on the rheology of pectin-calcium gels. Food Hydrocoll. 2009; 23:2069-2077
  79. 79. Kyomugasho C, Christiaens S, Van de Walle D, Van Loey AM, Dewettinck K, Hendrickx ME. Evaluation of cation-facilitated pectin-gel properties: Cryo-SEM visualisation and rheological properties. Food Hydrocoll. 2016; 61:172-182
  80. 80. Azad AKM, Ali MA, Sorifa Akter S, Jiaur Rahman I, Ahmed MIsolation and characterization of pectin extracted from lemon pomace during ripening Journal of Food and Nutrition Sciences 2014; 2(2): 30-35 DOI: 10.11648/j.jfns.20140202.12
  81. 81. Ismail NSM, Ramli N, Hani NM, Meon Z. Extraction and characterization of pectin from dragon fruit (Hylocereus polyrhizus) using various extraction conditions. Sains Malaysiana. 2012; 41: 41-45
  82. 82. Madhav, A., Pushpalatha, P.B. Characterization of pectin extracted from different fruit wastes. Int J Trop Agric. 2002; 40: 53-55
  83. 83. Cagatay Ceylan, Oguz Bayraktar, Erhan Atci, Şahin Sarrafi Extraction and characterization of pectin from fresh globe artichoke and canned artichoke waste. GIDA 2017; 42 (5): 568-576 DOI: 10.15237/gida.GD17015
  84. 84. Hercules Incorporated. Food gum products description: General description of pectin. 1999
  85. 85. Sundar Raj AA, Rubila S, Jayabalan R, Ranganathan TV. .A review on pectin: Chemistry due to general properties of pectin and pharmaceutical uses. Scientific reports, 2012; 1: 2, 1-4
  86. 86. Nguyen MBN, Pirak T. Physicochemical properties and antioxidant activities of white dragon fruit peel pectin extracted with conventional and ultrasound-assisted extraction, Cogent Food & Agriculture, 2019; 5:1, 1633076, DOI:10.1080/23311932.2019.1633076
  87. 87. Zhang W, Xu P, Zhang H. Pectin in cancer therapy: A review. Trends Food Sci. Technol. 2015; 44: 258-271
  88. 88. Karaki N, Aljawish A, Humeau C, Muniglia L, Jasniewski J. Enzymatic modification of polysaccharides: Mechanisms, properties, and potential applications: A review. Enzyme Microb. Technol. 2016; 90:1-18
  89. 89. Giri T.K., Thakur D., Alexander A., Ajazuddin, Badwaik H., Tripathy M., Tripathi D.K. Biodegradable IPN hydrogel beads of pectin and grafted alginate for controlled delivery of diclofenac sodium. J. Mater. Sci. Mater. Med. 2013; 24:1179-1190. DOI: 10.1007/s10856-013-4884-7
  90. 90. Minzanova ST, Mironov VF, Arkhipova DM, Khabibullina AV, Mironova LG, Zakirova YM, Milyukov VA. Biological Activity and Pharmacological Application of Pectic Polysaccharides: A Review. Polymers 2018;10:1407; doi:10.3390/polym10121407 31
  91. 91. Saladini B., Bigucci F., Cerchiara T., Gallucci M.C., Luppi B. Microparticles based on chitosan/pectin polyelectrolyte complexes for nasal delivery of tacrine hydrochloride. Drug Delivery Transl. Res. 2013; 3:33-41. DOI: 10.1007/s13346-012-0086-y
  92. 92. Ivone R, Yang Yand Jie Shen J (2021). Recent Advances in 3D Printing for Parenteral ApplicationsThe AAPS Journal 2021; 23:87. DOI: 10.1208/s12248-021-00610-z
  93. 93. Azad MA, Olawuni D, Kimbell G, Badruddoza AZM, Hossain MS, Sultana T. Polymers for extrusion-based 3D printing of pharmaceuticals: a holistic materials–process perspective. Pharmaceutics. 2020; 12:1-34
  94. 94. Araújo MRP, Sa-Barreto LL, Gratieri T, Gelfuso GM, Cunha-Filho M. The digital pharmacies era: how 3D printing technology using fused deposition modelling can become a reality. Pharmaceutics. 2019;11:3
  95. 95. Long J, Etxeberria AE, Nand AV, Bunt CR, Ray S, Seyfoddin A. A 3D printed chitosan-pectin hydrogel wound dressing for lidocaine hydrochloride delivery. Mater Sci Eng C [Internet]. 2019; 104:109873. Available from. https://doi.org/10.1016/j.msec.2019.109873
  96. 96. Lapomarda A, Pulidori E, Cerqueni G, Chiesa I, DeBlasiM, MA, Montemurro F, Duce C, Belmonte M, Tiné MR, Vozzi G, De Maria C. Pectin as Rheology Modifier of a Gelatin-Based Biomaterial Ink. Materials 2021; 14(11): 1-16
  97. 97. Mathew E, Pitzanti G, Larrañeta E, Lamprou DA. Three-dimensional printing of pharmaceuticals and drug delivery devices. Pharmaceutics. 2020; 12(3):1-9

Written By

Olufunke D. Akin-Ajani and Adenike Okunlola

Submitted: 20 August 2021 Reviewed: 26 August 2021 Published: 16 September 2021