Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Apr 3.
Published in final edited form as: ACS Appl Bio Mater. 2023 May 1;6(5):1684–1700. doi: 10.1021/acsabm.3c00050

Hydrogels for Mucosal Drug Delivery

Taj Yeruva 1, Sydney Yang 2, Shadin Doski 3, Gregg A Duncan 4,*
PMCID: PMC11966650  NIHMSID: NIHMS2066432  PMID: 37126538

Abstract

Mucosal tissues are often a desirable site of drug action to treat disease and engage the immune system. However, systemically administered drugs suffer from limited bioavailability in mucosal tissues where technologies to enable direct, local delivery to these sites would prove useful. In this Spotlight on Applications article, we discuss hydrogels as an attractive means for local delivery of therapeutics to address a range of conditions affecting the eye, nose, oral cavity, gastrointestinal, urinary bladder, and vaginal tracts. Considering the barriers to effective mucosal delivery, we provide an overview of the key parameters in the use of hydrogels for these applications. Finally, we highlight recent work demonstrating their use for inflammatory and infectious diseases affecting these tissues.

Keywords: hydrogels, drug delivery, mucosa, infectious disease, inflammatory disease

Graphical Abstract

graphic file with name nihms-2066432-f0008.jpg

1. INTRODUCTION

The human body is covered with mucosa amounting to ~400 m2 in total surface area, roughly 20 times greater in surface area than the skin. Mucosal layers keep bodily tissues hydrated and function as a protective layer by blocking foreign substances and pathogens. Mucus secreted at epithelial surfaces primarily consists of mucin glycoproteins that form a hydrogel mesh network via disulfide bonds and entanglement.1,2 When subjected to high physiological shear stresses (e.g., blinking, coughing), mucus undergoes a transition from solid to viscous fluid-like behavior where the viscosity of mucus approaches that of water.35 This allows for the rapid clearance, on the order of seconds to minutes, of mucus and its contents. Thus, drugs locally delivered as a suspension that become entrapped within the mucus gel are likely to be cleared from the body before they exert a therapeutic effect. In disease-affected tissues, further changes in mucin concentration and hydration due to inflammation can greatly affect mucus viscoelasticity and permeability toward therapeutic agents. Ointments and cream formulations can help to slow drug clearance but are often cumbersome to apply topically, particularly in the eye, nose, and oral cavity. To overcome these challenges, sustained drug delivery at mucosal sites can be achieved using hydrogels for local administration. Beyond their ability to act as vehicles for therapeutic cargo, hydrogels also can provide a protective barrier mimicking the native mucosal lining and may have a synergistic benefit in conditions where mucosal barrier integrity has been compromised such as in inflammatory bowel disease (IBD).

Over the past roughly three decades, mucosal delivery using polymers and hydrogel systems has advanced significantly owing to the pioneering work of Claus-Michael Lehr, Kinam Park, Nicholas Peppas, Joseph Robinson, and others.69 Through this work, the principles of mucoadhesion were established that govern how polymeric materials associate to a mucus-coated epithelium.10 The identification of mucoadhesive biomaterials has enabled the design of advanced hydrogel drug delivery systems to enhance the bioavailability of topical or locally administered drug products.11 While mucoadhesion remains an important design criterion, a new formulation strategy has emerged considering the pathological changes to mucosal epithelium, such as in IBD, where the mucosal interface is altered due to inflammation such that mucoadhesive strategies are not likely to be as effective. In addition to this, new design strategies have emerged that exploit physiological cues to enhance hydrogel mucoadhesion and trigger drug release at sites of injury. The purpose of this review is to highlight both previously known and newly discovered formulation strategies that should be considered in designing hydrogels for mucosal delivery. While hydrogel biomaterials can also be formulated as colloidal particle suspensions, we have focused our discussion on formulation strategies for bulk hydrogels and refer the reader to these recent review articles for additional considerations on microgel drug delivery.12,13

Each mucosal tissue is unique owing to its functions and required protective mechanisms. To directly interrogate the microscopic properties that give rise to mucus barrier function, microrheology can be used to measure the viscoelasticity experienced by micro- and nanoscaled molecules and pathogens.5 Using this approach, the pore sizes of the mucus layer have been found to vary throughout different mucosal tissues ranging from 20–200 nm, 100–300 nm, and 300–500 nm within the gastrointestinal, respiratory, and reproductive tracts, respectively.1416 This in turn presents unique challenges in delivering drugs in different mucosal tissues, and understanding these differences is essential to the design of effective hydrogel delivery systems. As such, we discuss in this review the tissue-specific mucosal barrier properties most relevant to hydrogel-based drug delivery before introducing specific formulation strategies that have proven useful in enhancing their therapeutic effect. While many opportunities have been explored using hydrogels in drug delivery applications, we highlight recent studies that demonstrate how hydrogel formulation strategies can be harnessed for local therapeutic delivery in inflammatory and infectious disease applications.

2. MUCOSAL TISSUES

2.1. Ocular.

The tear film, which is about 2–6 μm in thickness, is the outermost layer of the ocular surface, acts as a protective film to underlying epithelia, and provides a smooth refractive surface for clear vision.17,18 As depicted in Figure 1, the tear film is composed of a three-layered structure: an outermost lipid layer, middle aqueous layer, and innermost mucosal layer. The mucus layer is composed of soluble mucins and gel-forming mucins.19 The interface between tear film and cornea is composed of the glycocalyx and membrane tethered mucins, which are expressed on the apical surface of the corneal epithelium. These mucins play a vital role in pathogen recognition and clearance. Alterations in the percentages of different mucins can be observed in eye related diseases such as dry eye syndrome, ocular allergies, and conjunctivitis. In addition to defense against pathogens, the mucosal layer of the ocular surface also acts as a barrier to drug absorption. In vitro studies conducted on rose bengal permeation across corneal cell monolayer and stratified corneal cells with mucosal differentiation showed a blockage of rose bengal penetrance in the presence of mucins.20,21 In addition to the mucosal layer, the epithelium underlying the cornea is another barrier to drug absorption due to tight junctions that prevent paracellular transport.

Figure 1.

Figure 1.

Illustration of lipid, aqueous, and mucinous layers of the tear film. Inspired by refs 22 and 23.

A major limitation of ocular drug delivery is the rapid clearance of topically administered drugs. Tear drainage occurs as soon as eye drops are applied through the nasolacrimal duct to remove excess fluid on the ocular surface. In addition, rapid tear turnover of 0.5–2.2 μL min−1 and blinking reflexes of 6 to 15 times per minute lead to drug elimination at the rate of 0.5–1.0 min−1, reducing contact time with the ocular surface. Low contact time, along with permeation barriers such as the mucosal layer and underlying epithelium, leads to a low bioavailability of 0.1 to 5%. Increasing the contact time of a drug by using hydrogel formulations can help to increase absorption through the ocular surface, leading to higher bioavailability. Hydrogels offer a promising solution as a delivery system that can increase the retention time of drugs. However, ideally ocular hydrogel formulations should not blur vision. Several hydrogel delivery systems have been developed and proven effective for topical ophthalmic administration in animal models to treat a range of conditions such as cataracts, glaucoma, and bacterial conjunctivitis.2430

2.2. Upper Respiratory Tract.

The nasal cavity consists of three major compartments: the vestibular, respiratory, and olfactory regions. The vestibular region is the outermost region and is composed of nasal hairs, squamous epithelial cells, and ciliated cells.31 The middle region is the respiratory region, the most vascularized region, composed of mucus-secreting goblet cells, basal cells, ciliated, and nonciliated columnar cells. The innermost region is the olfactory region, which consists of tubular Bowman’s glands, which secrete a mucus layer, and olfactory sensory neurons. The nasal tissue has historically been used as a route for local drug delivery as it is easily accessible and also highly vascularized. Compared to the other mucosal tissues, the nasal tissue has a low enzymatic concentration, adding to its appeal as a route for drug delivery.32 However, given the high density of ciliated cells and low viscosity of mucus produced in the nostrils, rapid mucociliary clearance poses a significant challenge to intranasal drug delivery. Recently, it was shown that mucus clearance velocity on the nasal tract can be up to 4 times faster (~10–15 mm/min) than that observed in the lower respiratory tract.33 Thus, the retention of drugs in the nasal tract may be improved using hydrogels that locally adhere and slowly release therapeutics to target cells. As such, it should be noted that hydrogels used as intranasal delivery systems must be designed such that they do not impede normal clearance mechanisms or obstruct airway passages. To reach the lower respiratory tract, micro- and nanoparticulate systems provide a suitable drug delivery approach, and given our focus here on bulk hydrogels, delivery to these regions of the lung is not discussed. Several hydrogel-based formulations have been developed for intranasal delivery of therapeutics to treat conditions primarily affecting the nasal tract such as allergic rhinitis34 as well as other distal tissues for conditions such as diabetes and chronic pain.35,36 It is also proven to be an effective route for drug delivery to the brain, allowing the blood–brain barrier (BBB) to be bypassed where formulations using thermosensitive and in situ forming hydrogels have improved drug retention and achieved highly efficient nose-to-brain delivery.3740 For example, a mucoadhesive chitosan-based hydrogel was intranasally administered to rabbits and demonstrated significantly improved delivery of donepezil hydrochloride, used in the treatment of Alzheimer’s, with a roughly ~2-fold increase in total drug content reaching the brain as compared to orally administered tablets.41 It has also been shown that nasal immunization produces strong IgA and IgG antibody responses in the nose and other mucosal tissues (e.g., the cervicovaginal tract), making it an attractive route for vaccination against infectious diseases.42 As such, hydrogel delivery systems have also proven useful for the intranasal delivery of immunotherapies and vaccines to treat allergic asthma and prevent upper respiratory infections.4345

2.3. Oral/Buccal Cavity.

The oral or buccal cavity includes the inner linings of the lips, cheeks, palates, and tongue.46 These tissues are composed of multiple layers including the mucus layer, epithelium, and connective tissues.47 Epithelial layers express increased keratinization in areas, i.e., the palates and gingiva, subject to mechanical forces from mastication or swallowing.46 Mucosal regions of the oral cavity that require more elasticity and flexibility are nonkeratinized. Secreted mucins create a heterogeneous mucus layer and are present within the salivary film.48,49 Mucus secretions localized within the oral cavity have different affinities for bacteria and can interact with microorganisms individually or in tandem.48,50 Saliva secretions cover mucosal tissues in a film approximately 70–100 μm in thickness with a clearance rate of 0.3–0.4 mL/min.51,52

The oral mucosa has a large surface area with high blood supply and vascularization, which aids in rapid drug absorption for systemic uptake. Drug delivery through the oral cavity is sometimes used as an alternative to oral ingestion to bypass drug degradation by the gastrointestinal environment. However, the barrier function of the oral mucosa can be compromised in pathological conditions. For example, keratosis leads to increases in keratinized mucosa tissues and reduces drug permeability. Lesions within the oral cavity are characterized by tissue necrosis resulting in epithelial layer thinning and keratosis/hyperkeratosis.53 Current treatments include topical applications of corticosteroids and analgesics via tablets or patches to treat the affected mucosa tissue. Within the oral cavity, administered drugs are prone to degradation in a high enzymatic environment as well as degradation from experiencing constant mechanical forces. In addition, saliva clearance rapidly removes drugs from the mucosal surface. Mechanical forces from mouth movements also affect topical treatments.48 Thus, hydrogel-based delivery systems can be useful to resist salivary clearance and degradation leading to improved bioavailability.5456

2.4. Gastrointestinal.

Oral administration for gastrointestinal (GI) drug delivery is the most common mode of drug administration owing to its convenience. The rectal delivery route provides another alternative for local GI drug delivery. Orally administered drugs are swallowed and absorbed in the GI tract, whereas rectally administered drugs allow for local absorption of drugs. The GI tract has a total surface area of 250–400 m2 that can be divided into five compartments: the esophagus, stomach, intestine, colon, and rectum. Each compartment is unique in its structure and physiology, posing challenges and opportunities for drug delivery.

The esophagus has the shortest transit time of <1 min. This leads to poor drug absorption, making localized delivery to the esophagus quite challenging. The esophageal epithelium is further lined by a thin mucus layer that is the primary protective barrier against stomach acid reflux and ingested foreign materials including therapeutics. Similar to the change in epithelial thickness in esophagitis, an adherent mucin layer of ~90 μm is observed in Barrett’s esophagus but absent in healthy esophagus.57 To facilitate esophageal drug absorption, drug formulations with viscous liquids or gels are given to increase esophageal surface contact time. A study reported reversal of esophagitis in a dog model of Barrett’s esophagus when treated with mucoadhesive hydrogel formulated using extracellular matrix derived from porcine esophageal mucosa.58 Another study reported use of a hyaluronic acid-based hydrogel for treatment of esophageal fistulas.59

Orally ingested material travels through the esophagus to the stomach. The stomach’s transit time ranges between 1 and 4 h. It is highly acidic with a pH of 1–2 with a high concentration of gastric enzymes, such as pepsin, to aid in food digestion. Despite these harsh conditions, the stomach is protected from self-digestion through a thick mucus layer. As shown in Figure 2, the stomach is covered by two layers of mucus primarily composed of mucin 5AC (Muc5AC), with an unstirred inner layer firmly attached to epithelial cells and a removable outer layer. The mucosal lining of the stomach maintains a pH gradient across its layers with pH 2 at its luminal surface and pH 7 at its epithelial surface to protect the underlying epithelium.60 This is achieved through both epithelial cells continuously secreting bicarbonate as well as gel-forming mucins to replenish and stabilize the inner mucus layer of the stomach.61 Thus, orally administered drugs must withstand these harsh conditions to achieve biopharmaceutical efficacy where drug delivery devices (e.g., capsules) are essential. In addition, osmotic pressure along the GI wall, shear stresses due to the flow of gastric juice, and peristalsis of GI muscles can cause mechanical damage to drug carriers. If the drug withstands the harsh conditions of the lumen, the mucosal layer becomes the next barrier to drug absorption. Gastroretentive systems that prolong gastric residence time are used to allow drug absorption in the upper GI tract and to prolong drug release in a controlled manner. Examples of gastroretentive systems include floating devices,6265 expandable dosage forms,6669 and mucus or tissue adhesive systems.7073 Mucoadhesive systems are limited by the mucus turnover time of 1–2 days and epithelial turnover time of 4–5 days across the entire GI tract.

Figure 2.

Figure 2.

Schematic representation of the mucus organization in the gastrointestinal tract. OLM = outer loose mucus, IAM = inner adherent mucus. The mucus thicknesses in different regions are based on previous reports in rats. Figure inspired from refs 61, 74, and 75.

Intestinal transit time typically ranges between 1 to 6 h and has a drastic pH increase to 7.0–7.4. The intestine consists of a single, loosely cross-linked layer of mucus built by mucin 2 (Muc2), as depicted in Figure 2. However, the intestine is kept sterile through continuous flushing of the mucus layer and liquid secretions. The colon has the longest transit time of 1–3 days and a pH ranging between 6–6.7. Like the stomach, the colon has two mucus layers with an inner adherent layer and outer loose layer, both composed of Muc2. Interestingly, the same mucin behaves differently in the small intestine and colon. The inner mucus layer of the colon is formed by densely cross-linked sheets of mucus and is devoid of bacteria achieved through size exclusion.76 It is renewed at the epithelial surface by goblet cell secretions and converts to the outer mucus layer at the luminal surface. This outer mucus layer expands 4 to 5-fold in volume and is less viscous as a result of mucin network cleavage by endogenous proteases creating a habitat for commensal microbiota. In pathological conditions such as ulcerative colitis, a defective inner mucus layer can lead to bacterial contact with the epithelium, triggering an inflammatory cascade.77,78 Intestinal and colonic drug delivery can be achieved through pH dependent, time dependent, and pressure dependent systems by considering the rapid pH change, prolonged transit time, and high-pressure environment of the colon.7985 The rectum comprises the last 5 to 6 in. of the large intestine and temporarily stores fecal material before excretion through the anus. Within the rectal environment, an average fluid volume of 1–3 mL is maintained at a neutral pH of 7–8 with minimal buffering capacity. The lumenal facing layer of the rectum is covered with a single layer of mucus and is in direct contact with feces.86 Rectal drug delivery offers rapid absorption and a homeostatic environment with relatively low enzymatic activity. Thus, these ideal properties make rectal drug delivery an appealing noninvasive alternative to both local and systemic oral drug delivery. Hydrogels are a promising platform for rectal drug delivery, and several studies were reported based on mucoadhesive and in situ forming hydrogels.8790 To provide a target retention time for hydrogels to enable delivery to the GI tract and other mucosal tissues, Table 1 provides approximate transit times of therapeutic drugs across a mucosal layer with maximum thickness on the order of millimeters. It should be noted that the diffusion coefficient and transit times will vary at different mucosal sites depending on mucus thickness and pore size. The rate of drug release from the hydrogel system should also be considered in determining the retention time. However, overall, these values provide a target retention time for hydrogels to deliver the majority of a given therapeutic payload to mucosal sites.

Table 1.

Transit Time of Therapeutics across Mucosal Barriersa

Molecule Dmucus (10−7 cm2/s) tmax, h
Fluorescein 53    0.7
Polypeptide 18    2.1
Lactalbumin 12    3.1
Ovalbumin 8.8  4.2
Fc fragment 6.7  5.6
Fab fragment 6.7  5.6
BSA 5.7  6.6
F(ab′)2 fragment 4.4  8.5
Human IgA 5.1  7.3
Human IgG 2.9 12.9
Human IgM 2.8 13.3
Human S-IgA  0.49 76.2
a

To estimate target retention times for hydrogel-based delivery systems, we calculate the maximum transit time for a representative set of small molecules and proteins across mucosal barriers. The diffusion coefficient (Dmucus) used for each molecule was previously determined in midcycle human cervical mucus at 25 °C.91 We assume a maximum mucosal thickness (L) of 1.64 mm, which is the maximum thickness observed in the human body based on endoscopic imaging within the GI tract.92 Considering diffusion in the direction perpendicular to the tissue only (i.e., z-direction), we calculate the transit time (t) across the mucosal barrier as tmax = L2/2Dmucus.

2.5. Cervicovaginal.

The vaginal cavity has a large surface area with a dense network of blood vessels, making it an excellent route of drug administration for both systemic and local delivery. This administration route has been traditionally used for the local delivery of antibacterial, antifungal, spermicidal, and labor inducing agents.93 It also allows uterine targeting due to the first uterine pass effect when applied in the upper third of the vagina.94,95 This can be taken advantage of for hormone replacement therapies to maximize the desired effect and minimize systemic side effects. In recent years, many studies investigated its potential for microbicide delivery to prevent sexually transmitted infections96 and for localized treatment of cervical cancer.9799 However, the vaginal route is underexplored due to gender specificity and is limited by the possibility of leakage, poor distribution across vaginal walls, and low residence time due to epithelial shedding and mucosal clearance.

Like other mucosal tissues, mucus lining the cervicovaginal canal lubricates the female genital tract along with acting as a first line of defense against the outside environment. Cervical mucus (CM) is secreted by the goblet cells in the crevices of the cervix, travels down to the vaginal cavity, and mixes with host secretions, with cellular debris becoming cervicovaginal mucus (CVM) with a lower viscosity.100 CM and CVM composition changes throughout the menstrual cycle and allows the movement of sperm toward the ovule depending on the menstrual phase. A cervical mucus plug is formed during pregnancy to maintain the sterility of the uterus from the vaginal canal. CVM is enriched with lipids, salts, immune cells, antimicrobial peptides, and immunoglobulins and hosts the vaginal microbiome, which plays a major role in vaginal health.101 The microstructure and bulk rheology of fresh undiluted CVM exhibits relatively minor changes across a broad pH range of 1 to 9102 and can provide a similar barrier to drug permeation across pH fluctuation before and after mixing with seminal fluids, which are ~4.5 and ~7.2, respectively.103 The average pore size of CVM is 340 ± 70 nm, which is higher than human viruses. However, viruses and bacteria are trapped in the mucus barrier through interactions with mucin glycans, antibodies, or other proteins present in the mucus network.14 They are shed out of the body during mucosal clearance. However, mucosal barrier function is compromised due to microbial dysbiosis as mucolytic enzymes are released in conditions such as bacterial vaginosis.104,105 This allows the viruses and bacteria to pass through the mucus layer and reach the underlying epithelium to cause infections including HIV.106 The vaginal route is particularly effective in such cases as the drug is present in an effective concentration at the site of pathogen entry. Different dosage forms such as gels, suppositories, rings and films are preferred by women in different regions based on individual or partner choice, cultural norms and socioeconomic status of the region.107,108 Among those, hydrogel formulations can be designed to provide ease of application, increased drug residence time, and uniform distribution in the vaginal cavity.

2.6. Urinary Bladder.

The urinary bladder temporarily stores urine and facilitates voiding through urination.109 The luminal surface of the bladder is made of a mucosal layer called the urothelium, a stratified, transitional epithelium that acts as a thick barrier to protect the bladder walls from the toxic contents of urine. The apical membrane of urothelium is covered with glycocalyx containing both adherent and soluble glycoproteins, glycolipids, galectins and proteoglycans. This surface mucus layer has nonspecific antiadherent properties to defend against invading pathogens,110 and its disruption has shown to increase bacterial infections.111 Different drug delivery routes and modalities are studied to treat bladder related disorders such as bladder cancer, interstitial cystitis, and urinary tract infections. Intravesical delivery, where drugs are directly instilled into the urinary bladder via catheter, is a common route for systemically administered drugs but suffers from poor drug distribution to the bladder. In addition, intravesical delivery achieves local delivery through minimizing systemic toxicity. However, the efficacy of intravesical delivery is limited by poor urothelium permeability, continuous dilution by urine, and subsequent disposal out of the bladder.112

Hydrogel systems are increasingly reported for intravesical delivery as use of hydrogels minimizes drug dilution and offers other advantages, including mucoadhesion and floating properties, which can enhance drug absorption, retention, and sustained release in the bladder. A study reported an in situ gelling, mucoadhesive hydrogel formulated with high molecular weight chitosan and β-glycerophosphate for treatment of bladder cancer that showed superior resistance to urine washout when tested ex vivo using porcine urinary bladder.113 Another study reported floating hydrogels made of Poloxamer 407 (P407) for the treatment of bladder related diseases. This gel was prepared by simple shaking and locking of the bubbles that were formed as a result of P407 foamability. The hydrogel demonstrated prolonged residence time and continuous drug release for up to 10 h when tested in vivo using a rabbit acute bladder injury model.114 Several other hydrogels investigated for use as intravesical drug delivery systems and therapeutic bulking agents were reported as promising drug delivery systems for treating urinary bladder disorders.115118

3. FORMULATION STRATEGIES

3.1. Mucoadhesion.

Mucoadhesion has been widely exploited to enhance the retention of drug delivery systems in mucosal tissues.119121 For these applications, there are several factors that one should consider to achieve mucoadhesion. Here, we will briefly describe general considerations for mucoadhesive biomaterial design and would recommend several recent review articles for details on the theory and mechanisms of mucoadhesion.11,122,123 It has been found in prior work that polymers containing mucin-like chemical moieties (e.g., NH2, OH, SO4) generally favor mucin-biomaterial interactions via hydrogen bonding.122 Cationic polymers may nonspecifically interact with mucus through charge-mediated interactions with net-negatively charged mucins.11 Thiol-containing biopolymers may form covalent bonds to mucin cysteine residues.124 The degree of swelling can also impact mucosal adhesion as hydration aids in expansion of polymer chains to expose adhesive functional groups and facilitate entanglement with mucin biopolymers.125 Increasing polymer molecular weight also favors interpretation and chain entanglement with mucus. Conversely, increasing hydrogel cross-linking density can limit interfacial interpenetration with the mucus gel layer. However, overall, a consensus has yet to be reached on specific material properties for optimal mucoadhesion due, at least in part, to a lack of standardization in measuring mucoadhesive capacity.126 To address this, a study was performed to directly compare mucoadhesion of different natural and synthetic polymers to excised porcine small intestin,e providing insights into how material type, functional groups (e.g., cysteine-modified), and preparation method influence performance.127 It should also be noted that changes in pH and ionic strength can significantly alter mucin–polymer interactions, making it critical for biomaterials designed for mucoadhesion to be tested at physiological conditions.

Carbomers such as Carbopol934P, Carbopol974NF, and polycarbophil (NoveonAA1) can be used as excipients to improve mucoadhesion in polymer films. Using these excipients, a systematic study was conducted to determine formulation requirements for optimal mucoadhesion of poly(vinyl alcohol) (PVA) and polyvinylpyrrolidone (PVP) films. Formulations using PVP showed higher mucoadhesion than those utilizing PVA, likely due to more favorable intermolecular interactions between PVP and carbomer excipients. Mucoadhesive PVP films were further optimized at pH between 5.5–6.8 and carbomer concentrations between 2 and 10 wt % where polycarbophil most significantly enhanced mucoadhesive strength.128 Chitosan is another widely used mucoadhesive polymer as it contains primary amine groups that drive mucoadhesive interactions that are further reinforced by hydrophobic interactions and hydrogen binding.129 To simulate the different ionic environments within the GI tract, Lehr et al. showed a significant decrease in the mucoadhesivity of chitosan in artificial gastric fluid (pH 1.2) as compared to artificial intestinal fluid (pH 7.5), presumably due to chitosan’s solubility in acidic conditions. Interestingly, polycarbophil was also shown to significantly enhance the mucoadhesive capacity of chitosan under all conditions tested.130

Mucoadhesive polymers can also be chemically modified to enhance their performance. For example, alginate, a natural biopolymer known to possess mucoadhesive properties, was modified with cysteine (SH) groups. SH-modified alginate possessed a ~4-fold higher work of adhesion when applied on freshly excised mucosa as compared to unmodified alginate.131 Similarly, hyaluronic acid (HA) functionalized with sulfhydryl cysteine groups (HA-SH) significantly outperformed unmodified HA in terms of tensile strength, total work of adhesion, and maximum detachment force in nasal, buccal, and vaginal tissue.132 To increase the mucoadhesive capacity of chitosan, Kim et al. utilized a mussel-inspired adhesion approach and synthesized catechol-functionalized chitosan (Chi-C).133 As shown in Figure 3, catechols can oxidize to form irreversible covalent bonds with thiols and amines within mucus, leading to greater bioavailability and retention time of Chi-C in the GI tract following oral administration.

Figure 3.

Figure 3.

Mussel-inspired bioadhesion as a strategy to enhance mucoadhesion. (a) Schematic showcasing mechanisms of binding using catechol conjugated chitosan (Chi-C). Catechol moieties increase consolidation through covalent bonding with the amines and cysteines present in the mucins. (b) In vivo imaging system (IVIS) imaging of extracted organs of mice and (c) relative fluorescence intensity in the GI tract following oral administration of fluorescent labeled chitosan (Chi-FITC), poly(acrylic acid) (PAA-FTSC), and Chi-C (Chi-C-FITC). Reproduced with permission from ref 133. Copyright 2015 Elsevier.

Additional functional groups have more recently been investigated for enhancing mucoadhesive properties. Modified polymers with conjugated functional groups have been used to take advantage of the thiol-mediated chemistries to promote mucoadhesion by covalently binding to cysteine residues on mucin glycoproteins.134 Maleimide-functionalized PEGylated polymers such as alginate-polyethylene glycol-maleimide (Alg-PEGM) and poly(lactide-co-glycolide)-block-polyethylene glycol-maleimide (PLGA-PEG-Mal) showed a propensity to aggregate with mucins and showed increased retention to the urinary bladder mucosa, respectively.135,136 Acrylate-chitosan and polyethylene glycol diacrylate polymers have also been explored.137,138 Similarly, methacrylate-functionalized materials have been demonstrated to exhibit enhanced adhesion to mucosal tissues.139,140 Furthermore, designing copolymer hydrogels containing acrylate and methacrylate groups may strengthen mucoadhesion.141,142 In addition, aldehyde-functionalized polymers have been known to enhance mucoadhesion. More recent studies have synthesized aldehyde copolymers with mucoadhesion force to mucosa comparable to that of chitosan and other functional groups.143,144

3.2. Viscoelasticity.

Mucus viscoelasticity is directly related to mucus barrier function. In the respiratory tract, healthy mucus has low viscoelasticity to enable clearance by cilia.145 In many pathological conditions, mucus is marked by increased viscosity due to the hyperexpression of mucins, mucus dehydration, and accumulation of bacterial and cellular debris that alter mucin content and composition. In the lung, this leads to prolonged accumulation of mucus gels and provides an environment permissible to chronic bacterial infections. Similarly in drug delivery applications, hydrogel viscoelasticity can be tuned to slow their clearance from mucosal tissues and extend drug release time. Increasing hydrogel elasticity can be used to increase flow resistance, reduce clearance rate, and enhance drug contact time.146,147 For example, the addition of alginate as a gelling agent to a hydroxypropyl methylcellulose (HPMC) hydrogel was demonstrated to increase hydrogel viscosity and thereby increase drug retention and availability for ophthalmic drug delivery.148 Increased elasticity also enables hydrogels to better withstand external mechanical forces that are exerted on native tissues. Chitosan composite hydrogels showed resistance to deformation when exposed to stresses.149 Conversely, decreased viscosity enhances hydrogel flow, which ideally is used for the injection at target tissues. Due to having a low viscosity, hyaluronic acid and chitosan-based hydrogels are used for submucosal injections.150,151

3.3. Stimuli Sensitive.

Stimuli responsive hydrogels, also called smart hydrogels, respond to environmental stimuli by dramatically changing their structural and mechanical properties to allow in situ gelation or therapeutic release on demand. Environmental stimuli for hydrogel delivery systems can be from endogenous sources such as pH, temperature, enzymes, and redox conditions or exogenous sources such as light, ultrasound, electric field, and magnetic field. Hydrogels engineered to respond to specific stimuli allow precise spatiotemporal control of drug delivery to help minimize off-target effects, reduce inherent toxicity of drugs due to prolonged tissue exposure, and improve drug efficacy.

To design hydrogels with smart on-demand drug release capabilities, drugs are either covalently attached to the hydrogel backbone via a stimuli sensitive linker or physically entrapped in a hydrogel that has a stimuli sensitive backbone. For example, a plant-based antioxidant compound (rutin) was attached to the hydrogel carrier via borate ester linkage for pH and reactive oxygen species (ROS) responsive drug release for the treatment of IBD.152 In another study, an HIV fusion inhibitor (Enfuvirtide) was attached to the hydrogel backbone via a prostate specific antigen (PSA) labile linker for seminal fluid responsive delivery to prevent heterosexual HIV transmission in women.153,154 Carboxymethyl chitosan-based hydrogels show pH responsive swelling, and a study has shown higher bovine serum albumin (BSA) release in basic pH as opposed to acidic pH where the BSA was physically entrapped in the hydrogel.155 In another study, salmon calcitonin (sCT) was physically entrapped in dextran hydrogels for colon specific delivery, where higher sCT release was observed in simulated colonic fluid compared to phosphate buffer solution due to degradation of the hydrogel backbone by dextranases.156 Some amount of drug leaching is observed from hydrogels with physically entrapped drugs even in the absence of stimuli, which can be a drawback for some applications.

In addition to nonstimuli dependent chemical strategies to produce hydrogels in situ,157 thermosensitive hydrogels have been extensively investigated for mucosal delivery.158162 Solutions of polymers such as pluronic F127 and poly(N-isopropylacrylamide) transition from liquid to solid at or above body temperature to enable in situ gelation for uniform spreading and ease of application on mucosal surfaces. pH and ion concentration were also studied as triggers for in situ gelation at the ocular surface.163166 The difference in pH across the upper and lower GI tract can also be used for localized delivery to the intestine with pH triggered hydrogel delivery systems.167169 Stimuli responsive delivery can also be used to increase penetration across physiological barriers. Iontophoretic hydrogels were studied for delivering therapeutics across deep layers of the eye, which is hard to achieve noninvasively.170 Another interesting application of stimuli sensitive hydrogels is gastric resident dosage forms with controlled residence time. A tough triggerable hydrogel (TTH) was developed that can be ingested orally and swells significantly after ingestion, enabling prolonged gastric residence and controlled drug release. As shown in Figure 4, TTH delivery systems are cross-linked using reversible Ca2+ ionic bonds and disulfide bonds, which can be triggered using biocompatible chelators or reducing agent to dissolve rapidly for quick transit through the GI tract.68

Figure 4.

Figure 4.

Design and function of tough triggerable hydrogel (TTH) platform for delivery to the GI tract. (a) Overview of TTH design showing hydrogel backbone synthesized by separately cross-linking alginate and polyacrylamide using triggerable Ca2+ ionic and disulfide bonds. (b) TTH triggered from gel to viscous solution by EDTA and GSH. (c) Photographs of initial TTH strip before administration in the pig gastric cavity and the retrieved TTH strips from control and triggered pig after 1 h residence. Reprinted with permission from ref 68 under the terms and conditions of CC-BY open access license.

3.4. Hypotonic.

Fluid absorption and secretion at mucosal surfaces can occur through gradients in tonicity and osmotic driven flow. Delivery of hypertonic formulation leads to fluid secretion into the lumen and subsequent product leakage, whereas hypotonic formulations allow rapid fluid absorption and uniform product distribution at the mucosal surface. In addition, usage of hypertonic vehicles has also shown toxicity due to disruption of epithelial integrity in human colon epithelium and vaginal epithelial explants.171,172 Thus, hypotonic formulations may offer improved safety and better therapeutic efficacy. For example, doxorubicin administered in isotonic solution (PBS) has shown vaginal surface coverage of 23%, whereas drug administration in hypotonic solution (ultrapure water) showed 83% vaginal surface coverage.173 However, improved distribution and rapid absorption lead to poor retention in the vagina, which is not ideal for local delivery. To address this, mucus-penetrating nanoparticles can be formulated and administered in hypotonic solution, leading to improved distribution as well as retention due to slow release of the drug following uniform spreading and accumulation of nanoparticles at the vaginal epithelium.173 Similarly, widespread and highly uniform nanoparticle distribution was observed in the colon when delivered locally as a hypotonic enema formulation.174 Hence, enhanced drug distribution, tissue absorption, and retention can be expected if drugs are mucosally administered in hypotonic hydrogels. Accordingly using thermoreversible Pluronic hydrogels, a hypotonic eye-drop hydrogel formulation was developed for drug delivery to the eye. In this work, it was found that Pluronic gels spread uniformly at the ocular surface due to osmotically driven water absorption. This comfortably fits under eyelids, holds drug at the ocular surface over an extended period, and resists clearance from blinking.175

3.5. Floating Hydrogels.

Floating hydrogel systems have been used to increase residence times in the gastric and urinary bladder regions, as floating hydrogels are resistant to stomach and bladder emptying. These systems float on top of the stomach and bladder contents, allowing for controlled drug release and improved bioavailability. Several approaches have been developed to formulate floating hydrogel delivery systems. One such system involves using a mixture of Poloxamer 407 (P407) and NaHCO3 for the intravesical delivery of adriamycin to treat bladder cancer. In this system, a liquid mixture of P407, NaHCO3, and adriamycin is instilled into the bladder through a catheter, which then gels in situ at body temperature. When the urine is acidified, NaHCO3 forms CO2 bubbles at the hydrogel surface, enabling the hydrogel to float.176 Perfluoropentane has also been used to produce temperature-sensitive floating hydrogels. Perfluoropentane has a lower boiling point than body temperature (29 °C) and converts to a gas state in the bladder, causing the hydrogel to swell and float.177 Emulsion gelation is another technique used to make floating hydrogels. Here, oil droplets are encapsulated within hydrogels during cross-linking to decrease the hydrogel density in aqueous solution to form floating hydrogels.178,179 Other reported floating hydrogel designs include hydrophobic hydrogels, superporous hydrogels, and hydrogel rafts.63,65,180182

3.6. Inflammation-Targeted Adhesion.

At sites of inflammation in diseases like IBD, it has been found that the mucus barrier is disrupted and inflammatory lesions are left unprotected from potentially harmful microbes in the gut.183 At these sites of disease, the epithelium is devoid of mucus and acquires a net-positive charge primarily due to the accumulation of positively charged proteins such as transferrin and antimicrobial peptides.184 As a result, an emerging design parameter has been in the use of hydrogels (and other biomaterials) that specifically target these sites of inflammation. This can be achieved with the use of net-negatively charged hydrogels that will bind to the net-positively charged inflammatory lesion. In mouse models of colitis, an ascorbyl palmitate (AP) microfiber hydrogel possessing a net-negative charge was found to be retained within the colon for up to 12 h, as presented in Figure 5.185 Of note, these gels were poorly retained in healthy animals due to their lack of mucoadhesion. Thus, this presents a new strategy using hydrogel systems to localize the release of therapeutic compounds at disease sites and further minimize off-target effects. While primarily demonstrated in the GI tract to date, future studies should determine how broadly applicable this approach is to other inflamed mucosal tissues.

Figure 5.

Figure 5.

Mode of action and in vivo retention of inflammation targeting (IT) hydrogels. (a) Schematic of ascorbyl palmitate hydrogel fiber formation and (b) adherence of negatively charged IT hydrogel fibers to positively charged inflamed epithelium. (c, d) Preferential adhesion of IT hydrogel fibers to inflamed colon epithelium ex vivo and in vivo in an induced (dextran sodium sulfate (DSS) and genetic (T-bet−/−Rag2−/−, TRUC) mouse model of IBD. Reproduced with permission from ref 185. Copyright 2015 The American Association for the Advancement of Science.

4. APPLICATIONS IN INFLAMMATORY AND INFECTIOUS DISEASES

4.1. Mucosal Delivery of Small Molecules.

Local delivery of anti-inflammatory drugs to mucosal tissues can reduce systemic exposure and may help avoid side effects (e.g., immunosuppression, hepatotoxicity) by reducing the dose required. Hydrogel-based delivery systems can greatly improve outcomes in IBD and other inflammatory conditions by locally concentrating the therapeutic payload within mucosal tissues. In addition to their inflammation-targeting properties, AP hydrogels are innately amphiphilic, making them effective for loading of lipophilic, anti-inflammatory compounds. This amphiphilicity enables the self-assembly of micelles into a microfiber hydrogel and encapsulation of lipophilic dexamethasone. As noted above, these hydrogels adhere to inflamed murine intestinal mucosa where sustained release of dexamethasone significantly reduces colitis severity.185 A similar strategy has been used to formulate hydrogels for the encapsulation and buccal delivery of dexamethasone.186 Mometasone, another lipophilic corticosteroid, has been delivered through encapsulation and release via thermosensitive copolymer hydrogel microspheres to the sinonasal cavity. Thermosensitivity of the copolymer hydrogel system allowed the hydrogel to conform to the complex architecture of the sinonasal cavity before gelation at body temperature and adherence to the mucosal layer. Treatment with mometasone-loaded hydrogels reduced inflammation in a rabbit model of chronic rhinosinusitis.187 This prior work demonstrates how amphiphilic hydrogels can be effectively used to deliver lipophilic drugs at targeted sites that otherwise would have poor bioavailability and/or greater metabolic clearance.

Antibiotic misuse and overuse is a widespread issue leading to antibiotic resistant and multidrug resistant bacteria. In addition to this, systemic administration of antibiotics has many drawbacks including low bioavailability and unwanted side effects such as drowsiness, diarrhea, and nausea.188 To circumvent these challenges, there has been a large body of work using hydrogels for antibiotic delivery to mucosal tissues. A general strategy employed is to load the antibiotic into a carrier such as liposomes or solid lipid nanoparticles (SLNs) and then load the carrier into a hydrogel for extended release. Chen et al. successfully loaded clarithromycin (CAM) into liposomes, which were then loaded into a 4-arm PEG hydrogel.188 For in vivo studies, CAM-liposome loaded PEG hydrogels were injected into the maxillary sinus of rabbits to treat acute bacterial rhinosinusitis (ABRS). They reported a significant decrease in the burden of Streptococcus pneumoniae with minimal signs of inflammation or other side effects, as shown in Figure 6. Hydrogels have also been used to deliver antibiotics to treat oral periodontitis disease.189192 Interestingly, Wróblewska et al. compared the efficacy of alginate hydrogels to that of organogels (nonaqueous, lipophilic) and bigels (biphasic hydrogel and organogel) loaded with metronidazole (MZT).192 Alginate hydrogels were found to have the highest antibacterial activity, cumulative permeation through porcine buccal mucosa, and retention time with the mucosa. Hydrogels have also been formulated for delivery of antifungals to mucosal sites. To target Candida albicans in the oral cavity, Şenel et al. successfully loaded chlorhexidine gluconate (Chx) into chitosan hydrogels.193 Chx-loaded gels achieved retention times over 3 h and showed significant in vitro antifungal activity against C. albicans. To target C. albicans in the vaginal cavity, Mirza et al. loaded itraconazole into SLNs and then into a gel composed of a mucoadhesive excipient (Carbopol 934) and Pluronic F127.194 The hydrogel formulations outperformed a commercially available antimicrobial cream (Gynazole), sustaining microbicidal activity over 21 days in a rat model of C. albicans infection.

Figure 6.

Figure 6.

Hydrogel-based mucosal drug delivery for bacterial rhinosinusitis. (a) Schematic of the hydrogel design and (b) in vivo administration process through local injection to treat acute bacterial rhinosinusitis in rabbits. (c, d) RT-PCR and Western blots showing the decrease in inflammatory markers with the use of CAM liposome loaded gels in vivo. Reproduced with permission from ref 188. Copyright 2020 John Wiley and Sons.

4.2. Mucosal Delivery of Biologics.

Biological therapeutics, such as monoclonal antibodies, have increased dramatically in recent years with rapid advances in recombinant protein production and genetic engineering. However, the bioavailability of parenterally administered biologics at the mucosal tissues is generally low and often requires administration of very high doses to treat diseases, such as IBD. Recent evidence also suggests mucosal delivery of vaccines may offer greater protective benefits where new strategies for intranasal delivery are desired. This motivates the use of hydrogels to enable low dosage treatments of glycan, protein, or nucleic acid drugs through direct delivery to the mucosal surface. Biological cargoes are also susceptible to rapid enzymatic degradation once administered where hydrogel carriers can provide additional protection.

In a previous study, hydrogels were shown to be effective in the local treatment of IBD using heparin, a naturally occurring glycosaminoglycan and anticoagulant. Specifically, heparin was loaded into an injectable protein hydrogel (HEP-Ag-BSA) and delivered rectally in an induced mouse model of colitis. Due to its net-negative charge, HEP-Ag-BSA hydrogels displayed preferential binding to inflamed tissue and local sustained release of heparin, leading to hindrance of inflammation and microthrombosis that catalyzes IBD pathogenesis, as shown in Figure 7.195 For vaccine applications, an intranasal gel was developed for delivery of broadly acting subunit influenza vaccine. Intranasal gel precursors consisting of branched polyethylenimine (PEI) and oxidized dextran (Ox-DEX) were administered by dual-barrel syringe, and PEI-Ox-DEX hydrogel was formed by in situ gelation via Schiff’s base cross-linking. Antigens were loaded into the gel by mixing with gel precursor solutions. PEI-Ox-DEX hydrogel has shown prolonged mucoadhesion and nasal retention, leading to robust mucosal immune responses.196 A similar study was conducted to evaluate the effect of chitosan-hydrogel vaccine against influenza infection in mice. Mice intranasally immunized with in situ gelling chitosan hydrogel loaded with influenza-specific antigens demonstrated robust mucosal immune responses and protection after challenge with influenza virus.45 Hydrogels have also been shown to be effective in delivering gene therapy for ocular wound repair. In this study, Connexin43 antisense oligonucleotide (ASON) was delivered to rats after corneal injury using a chitosan-based in situ gelling system. This study demonstrated the effective delivery of the ASON to the wounded tissue.197 Gene therapy can also be achieved by loading the hydrogel with nucleic acid-loaded nanoparticles (e.g., liposomes, polyplexes) as cargo to further enhance mucosal gene delivery.198

Figure 7.

Figure 7.

Injectable protein hydrogels for mucosal delivery of biologics in IBD. (a) Schematic showing gelation of BSA-SH, heparin, and Ag+ through disulfide, coordination, and ionic bonds. (b) IVIS imaging of rhodamine labeled HEP-Ag-BSA treated colons showing lesion adhesive property at inflamed site. (c) Illustration of HEP-Ag-BSA administration and its effect on mouse colon with DSS induced colitis. (d) Comparison of colon lengths following in vivo treatment with HEP-Ag-BSA, demonstrating inflammation modulation capability. Reprinted with permission from ref 195 under the terms and conditions of CC-BY open access license.

5. CONCLUSION

Delivery of therapeutics to the mucosa can have tremendous therapeutic effect for individuals with chronic inflammatory diseases and difficult-to-treat infections. A local delivery approach may also spare patients from unintended side effects and reduce overall costs by lowering dose requirements. To make mucosal delivery a viable option in the clinic, hydrogels may help to address the challenges associated with overcoming physiological barriers to effective local drug delivery. Using the formulation strategies outlined here, hydrogels can be designed to encapsulate a wide array of therapeutics, retain the payload at the mucosal site of interest, and release the drug to target cells in a sustained and/or stimuli responsive manner. For inflammatory and infectious disease applications, future work could explore the use of hydrogels for mucosal delivery of cellular,199 extracellular vesicle,200 and microbial therapies.201 In addition, hydrogel-based systems with intrinsic regenerative and/or immune-modulating properties could be particularly useful as mucosal therapies for these applications.202,203 With careful consideration of the unique challenges to effective hydrogel delivery in mucosal tissues, abundant opportunities exist to create new highly efficacious treatments.

ACKNOWLEDGMENTS

This work was supported by a Burroughs Wellcome Fund Career Award at the Scientific Interface (to G.A.D.), the National Science Foundation Graduate Research Fellowship Program (to S.Y.), and the National Institutes of Health (R21 EB030834, R01 HL160540, and U19 AI162130).

Footnotes

Complete contact information is available at: https://pubs.acs.org/10.1021/acsabm.3c00050

The authors declare no competing financial interest.

NOTE ADDED AFTER ASAP PUBLICATION

The caption of Figure 1 was modified in the version of this article published May 1, 2023. The corrected caption published May 2, 2023.

Contributor Information

Taj Yeruva, Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States.

Sydney Yang, Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States.

Shadin Doski, Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States.

Gregg A. Duncan, Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States.

REFERENCES

  • (1).Bansil R; Turner BS Mucin Structure, Aggregation, Physiological Functions and Biomedical Applications. Curr. Opin. Colloid Interface Sci 2006, 11 (2–3), 164–170. [Google Scholar]
  • (2).Bansil R; Turner BS The Biology of Mucus: Composition, Synthesis and Organization. Adv. Drug Delivery Rev 2018, 124, 3–15. [DOI] [PubMed] [Google Scholar]
  • (3).Button B; Goodell HP; Atieh E; Chen Y-C; Williams R; Shenoy S; Lackey E; Shenkute NT; Cai L-H; Dennis RG; Boucher RC; Rubinstein M Roles of Mucus Adhesion and Cohesion in Cough Clearance. Proc. Natl. Acad. Sci. U.S.A 2018, 115 (49), 12501–12506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (4).Girod S; Zahm J; Plotkowski C; Beck G; Puchelle E Role of the Physiochemical Properties of Mucus in the Protection of the Respiratory Epithelium. Eur. Respir. J 1992, 5 (4), 477–487. [PubMed] [Google Scholar]
  • (5).Lai SK; Wang Y-Y; Wirtz D; Hanes J Micro- and Macrorheology of Mucus. Adv. Drug Delivery Rev 2009, 61 (2), 86–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (6).Lehr CM Bioadhesion Technologies for the Delivery of Peptide and Protein Drugs to the Gastrointestinal Tract. Crit. Rev. Ther. Drug Carrier Syst 1994, 11 (2), 119–160. [PubMed] [Google Scholar]
  • (7).Peppas NA; Sahlin JJ Hydrogels as Mucoadhesive and Bioadhesive Materials: A Review. Biomaterials 1996, 17 (16), 1553–1561. [DOI] [PubMed] [Google Scholar]
  • (8).Park H; Robinson JR Mechanisms of Mucoadhesion of Poly (Acrylic Acid) Hydrogels. Pharm. Res 1987, 4, 457–464. [DOI] [PubMed] [Google Scholar]
  • (9).Peppas NA; Robinson JR Bioadhesives for Optimization of Drug Delivery. J. Drug Targeting 1995, 3 (3), 183–184. [DOI] [PubMed] [Google Scholar]
  • (10).Shaikh R; Raj Singh TR; Garland MJ; Woolfson AD; Donnelly RF Mucoadhesive Drug Delivery Systems. Journal of pharmacy and Bioallied Sciences 2011, 3 (1), 89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (11).Khutoryanskiy VV Advances in Mucoadhesion and Mucoadhesive Polymers. Macromol. Biosci 2011, 11 (6), 748–764. [DOI] [PubMed] [Google Scholar]
  • (12).Wang Y; Guo L; Dong S; Cui J; Hao J Microgels in Biomaterials and Nanomedicines. Adv. Colloid Interface Sci 2019, 266, 1–20. [DOI] [PubMed] [Google Scholar]
  • (13).Agrawal G; Agrawal R Functional Microgels: Recent Advances in Their Biomedical Applications. Small 2018, 14 (39), 1801724. [DOI] [PubMed] [Google Scholar]
  • (14).Lai SK; Wang Y-Y; Hida K; Cone R; Hanes J Nanoparticles Reveal That Human Cervicovaginal Mucus Is Riddled with Pores Larger than Viruses. Proc. Natl. Acad. Sci. U.S.A 2010, 107 (2), 598–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (15).Schuster BS; Suk JS; Woodworth GF; Hanes J Nanoparticle Diffusion in Respiratory Mucus from Humans without Lung Disease. Biomaterials 2013, 34 (13), 3439–3446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (16).Round AN; Rigby NM; Garcia de la Torre A; Macierzanka A; Mills ENC; Mackie AR Lamellar Structures of MUC2-Rich Mucin: A Potential Role in Governing the Barrier and Lubricating Functions of Intestinal Mucus. Biomacromolecules 2012, 13 (10), 3253–3261. [DOI] [PubMed] [Google Scholar]
  • (17).Kayal A The Physiology of Tear Film. In Dry Eye Syndrome; Ferreri FM, Ed.; IntechOpen: Rijeka, 2021; Ch. 2; DOI: 10.5772/intechopen.98945. [DOI] [Google Scholar]
  • (18).King-Smith PE; Fink BA; Fogt N; Nichols KK; Hill RM; Wilson GS The Thickness of the Human Precorneal Tear Film: Evidence from Reflection Spectra. Investigative Ophthalmology & Visual Science 2000, 41 (11), 3348–3359. [PubMed] [Google Scholar]
  • (19).Hodges RR; Dartt DA Tear Film Mucins: Front Line Defenders of the Ocular Surface; Comparison with Airway and Gastrointestinal Tract Mucins. Exp. Eye Res 2013, 117, 62–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (20).Argüeso P; Tisdale A; Spurr-Michaud S; Sumiyoshi M; Gipson IK Mucin Characteristics of Human Corneal-Limbal Epithelial Cells That Exclude the Rose Bengal Anionic Dye. Investigative Ophthalmology & Visual Science 2006, 47 (1), 113–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (21).Feenstra RPG; Tseng SCG What Is Actually Stained by Rose Bengal? Archives of Ophthalmology 1992, 110 (7), 984–993. [DOI] [PubMed] [Google Scholar]
  • (22).Butovich IA; Millar TJ; Ham BM Understanding and Analyzing Meibomian Lipids—A Review. Current Eye Research 2008, 33 (5–6), 405–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (23).Hodges RR; Dartt DA Regulatory Pathways in Lacrimal Gland Epithelium. International Review of Cytology; Academic Press: 2003; Vol. 231, pp 129–196; DOI: 10.1016/S0074-7696(03)31004-6. [DOI] [PubMed] [Google Scholar]
  • (24).Shi H; Wang Y; Bao Z; Lin D; Liu H; Yu A; Lei L; Li X; Xu X Thermosensitive Glycol Chitosan-Based Hydrogel as a Topical Ocular Drug Delivery System for Enhanced Ocular Bioavailability. Int. J. Pharm 2019, 570, 118688. [DOI] [PubMed] [Google Scholar]
  • (25).Seah I; Loh XJ; Su X A Topical Gel for Extended Ocular Drug Release. Nature Biomedical Engineering 2020, 4 (11), 1024–1025. [DOI] [PubMed] [Google Scholar]
  • (26).Kotreka UK; Davis VL; Adeyeye MC Development of Topical Ophthalmic in Situ Gel-Forming Estradiol Delivery System Intended for the Prevention of Age-Related Cataracts. PloS one 2017, 12 (2), No. e0172306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (27).Gholizadeh S; Chen X; Yung A; Naderi A; Ghovvati M; Liu Y; Farzad A; Mostafavi A; Dana R; Annabi N Development and Optimization of an Ocular Hydrogel Adhesive Patch Using Definitive Screening Design (DSD). Biomaterials Science 2023, 11, 1318. [DOI] [PubMed] [Google Scholar]
  • (28).Kouchak M; Mahmoodzadeh M; Farrahi F Designing of a PH-Triggered Carbopol®/HPMC In Situ Gel for Ocular Delivery of Dorzolamide HCl: In Vitro, In Vivo, and Ex Vivo Evaluation. AAPS PharmSciTech 2019, 20 (5), 210. [DOI] [PubMed] [Google Scholar]
  • (29).Bhalerao H; Koteshwara KB; Chandran S Levofloxacin Hemihydrate In Situ Gelling Ophthalmic Solution: Formulation Optimization and In Vitro and In Vivo Evaluation. AAPS PharmSciTech 2019, 20 (7), 272. [DOI] [PubMed] [Google Scholar]
  • (30).Deepthi S; Jose J Novel Hydrogel-Based Ocular Drug Delivery System for the Treatment of Conjunctivitis. International Ophthalmology 2019, 39 (6), 1355–1366. [DOI] [PubMed] [Google Scholar]
  • (31).Keller L-A; Merkel O; Popp A Intranasal Drug Delivery: Opportunities and Toxicologic Challenges during Drug Development. Drug Delivery and Translational Research 2022, 12 (4), 735–757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (32).Lobaina Mato Y Nasal Route for Vaccine and Drug Delivery: Features and Current Opportunities. Int. J. Pharm 2019, 572, 118813. [DOI] [PubMed] [Google Scholar]
  • (33).Rogers TD; Button B; Kelada SNP; Ostrowski LE; Livraghi-Butrico A; Gutay MI; Esther CR; Grubb BR Regional Differences in Mucociliary Clearance in the Upper and Lower Airways. Frontiers in Physiology 2022, 13, 842592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (34).Su Y; Sun B; Gao X; Liu S; Hao R; Han B Chitosan Hydrogel Doped with PEG-PLA Nanoparticles for the Local Delivery of MiRNA-146a to Treat Allergic Rhinitis. Pharmaceutics 2020, 12 (10), 907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (35).Li X; Du L; Chen X; Ge P; Wang Y; Fu Y; Sun H; Jiang Q; Jin Y Nasal Delivery of Analgesic Ketorolac Tromethamine Thermo- and Ion-Sensitive in Situ Hydrogels. Int. J. Pharm 2015, 489 (1), 252–260. [DOI] [PubMed] [Google Scholar]
  • (36).Wu J; Wei W; Wang L-Y; Su Z-G; Ma G-H A Thermosensitive Hydrogel Based on Quaternized Chitosan and Poly(Ethylene Glycol) for Nasal Drug Delivery System. Biomaterials 2007, 28 (13), 2220–2232. [DOI] [PubMed] [Google Scholar]
  • (37).Wang JT-W; Rodrigo AC; Patterson AK; Hawkins K; Aly MMS; Sun J; Al Jamal KT; Smith DK Enhanced Delivery of Neuroactive Drugs via Nasal Delivery with a Self-Healing Supramolecular Gel. Advanced Science 2021, 8 (14), 2101058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (38).Agrawal M; Saraf S; Saraf S; Dubey SK; Puri A; Gupta U; Kesharwani P; Ravichandiran V; Kumar P; Naidu VGM; Murty US; Ajazuddin; Alexander, A. Stimuli-Responsive In Situ Gelling System for Nose-to-Brain Drug Delivery. J. Controlled Release 2020, 327, 235–265. [DOI] [PubMed] [Google Scholar]
  • (39).Chen X; Zhi F; Jia X; Zhang X; Ambardekar R; Meng Z; Paradkar AR; Hu Y; Yang Y Enhanced Brain Targeting of Curcumin by Intranasal Administration of a Thermosensitive Poloxamer Hydrogel. J. Pharm. Pharmacol 2013, 65 (6), 807–816. [DOI] [PubMed] [Google Scholar]
  • (40).Zhou S; Zheng X; Chen J; Xu Y; Du X; Wang C; Cui P; Qiu L; Jiang P; Ni X; et al. Liposome Loaded Ion/Temperature Dual Responsive Gellan Gum Hydrogel as Potential Nasal-To-Brain Delivery System. Journal of Biomedical Nanotechnology 2022, 18 (2), 571–580. [DOI] [PubMed] [Google Scholar]
  • (41).Al Harthi S; Alavi SE; Radwan MA; El Khatib MM; AlSarra IA Nasal Delivery of Donepezil HCl-Loaded Hydrogels for the Treatment of Alzheimer’s Disease. Sci. Rep 2019, 9 (1), 9563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (42).Holmgren J; Czerkinsky C Mucosal Immunity and Vaccines. Nature Medicine 2005, 11 (4), S45–S53. [DOI] [PubMed] [Google Scholar]
  • (43).Zhong Y; Su C; Wu S; Miao C; Wang B Nasal Delivery of an Immunotherapeutic Vaccine in Thermosensitive Hydrogel against Allergic Asthma. International Immunopharmacology 2023, 116, 109718. [DOI] [PubMed] [Google Scholar]
  • (44).Pastor Y; Ting I; Martínez AL; Irache JM; Gamazo C Intranasal Delivery System of Bacterial Antigen Using Thermosensitive Hydrogels Based on a Pluronic-Gantrez Conjugate. Int. J. Pharm 2020, 579, 119154. [DOI] [PubMed] [Google Scholar]
  • (45).Bedford JG; Caminschi I; Wakim LM Intranasal Delivery of a Chitosan-Hydrogel Vaccine Generates Nasal Tissue Resident Memory CD8+ T Cells That Are Protective against Influenza Virus Infection. Vaccines 2020, 8 (4), 572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (46).Squier CA; Kremer MJ Biology of Oral Mucosa and Esophagus. JNCI Monographs 2001, 2001 (29), 7–15. [DOI] [PubMed] [Google Scholar]
  • (47).Sudhakar Y; Kuotsu K; Bandyopadhyay AK Buccal Bioadhesive Drug Delivery — A Promising Option for Orally Less Efficient Drugs. J. Controlled Release 2006, 114 (1), 15–40. [DOI] [PubMed] [Google Scholar]
  • (48).Derrien M; van Passel MWJ; van de Bovenkamp JHB; Schipper R; de Vos W; Dekker J Mucin-Bacterial Interactions in the Human Oral Cavity and Digestive Tract. Gut Microbes 2010, 1 (4), 254–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (49).Collins LMC; Dawes C The Surface Area of the Adult Human Mouth and Thickness of the Salivary Film Covering the Teeth and Oral Mucosa. J. Dent Res 1987, 66 (8), 1300–1302. [DOI] [PubMed] [Google Scholar]
  • (50).Tabak LA In Defense of the Oral Cavity: Structure, Biosynthesis, and Function of Salivary Mucins. Annual review of physiology 1995, 57 (1), 547–564. [DOI] [PubMed] [Google Scholar]
  • (51).Iorgulescu G Saliva between Normal and Pathological. Important Factors in Determining Systemic and Oral Health. Journal of medicine and life 2009, 2 (3), 303–307. [PMC free article] [PubMed] [Google Scholar]
  • (52).Mystkowska J; Niemirowicz-Laskowska K; Łysik D; Tokajuk G; Dąbrowski JR; Bucki R The Role of Oral Cavity Biofilm on Metallic Biomaterial Surface Destruction-Corrosion and Friction Aspects. International Journal of Molecular Sciences 2018, 19 (3), 743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (53).Farthing PM; Speight PM Problems and Pitfalls in Oral Mucosal Pathology. Current Diagnostic Pathology 2006, 12 (1), 66–74. [Google Scholar]
  • (54).Rezazadeh M; Jafari N; Akbari V; Amirian M; Tabbakhian M; Minaiyan M; Rostami M A Mucoadhesive Thermosensitive Hydrogel Containing Erythropoietin as a Potential Treatment in Oral Mucositis: In Vitro and in Vivo Studies. Drug Delivery and Translational Research 2018, 8 (5), 1226–1237. [DOI] [PubMed] [Google Scholar]
  • (55).Su C-Y; Ho H-O; Chen Y-C; Yu Y-T; Liu D-Z; Chao F-C; Sheu M-T Complex Hydrogels Composed of Chitosan with Ring-Opened Polyvinyl Pyrrolidone as a Gastroretentive Drug Dosage Form to Enhance the Bioavailability of Bisphosphonates. Sci. Rep 2018, 8 (1), 8092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (56).Sharpe LA; Daily AM; Horava SD; Peppas NA Therapeutic Applications of Hydrogels in Oral Drug Delivery. Expert opinion on drug delivery 2014, 11 (6), 901–915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (57).Dixon J; Strugala V; Griffin SM; Welfare MR; Dettmar PW; Allen A; Pearson JP Esophageal Mucin: An Adherent Mucus Gel Barrier Is Absent in the Normal Esophagus but Present in Columnar-Lined Barrett’s Esophagus. Am. J. Gastroenterol 2001, 96 (9), 2575–2583. [DOI] [PubMed] [Google Scholar]
  • (58).Naranjo JD; Saldin LT; Sobieski E; Quijano LM; Hill RC; Chan PG; Torres C; Dziki JL; Cramer MC; Lee YC; et al. Esophageal Extracellular Matrix Hydrogel Mitigates Metaplastic Change in a Dog Model of Barrett’s Esophagus. Science advances 2020, 6 (27), eaba4526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (59).Piantanida E; Boškoski I; Quero G; Gallo C; Zhang Y; Fiorillo C; Arena V; Costamagna G; Perretta S; De Cola L Nanocomposite Hyaluronic Acid-Based Hydrogel for the Treatment of Esophageal Fistulas. Materials Today Bio 2021, 10, 100109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (60).Ensign LM; Cone R; Hanes J Oral Drug Delivery with Polymeric Nanoparticles: The Gastrointestinal Mucus Barriers. Adv. Drug Deliv Rev 2012, 64 (6), 557–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (61).Johansson MEV; Sjövall H; Hansson GC The Gastrointestinal Mucus System in Health and Disease. Nat. Rev. Gastroenterol Hepatol 2013, 10 (6), 352–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (62).Bajpai SK; Tankhiwale R Preparation, Characterization and Preliminary Calcium Release Study of Floating Sodium Alginate/Dextran-based Hydrogel Beads: Part I. Polymer international 2008, 57 (1), 57–65. [Google Scholar]
  • (63).Chavda H; Patel C Chitosan Superporous Hydrogel Composite-Based Floating Drug Delivery System: A Newer Formulation Approach. Journal of Pharmacy And Bioallied Sciences 2010, 2 (2), 124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (64).Sheikh FA; Hussain MA; Ashraf MU; Haseeb MT; Farid-ul-Haq M Linseed Hydrogel Based Floating Drug Delivery System for Fluoroquinolone Antibiotics: Design, in Vitro Drug Release and in Vivo Real-Time Floating Detection. Saudi Pharmaceutical Journal 2020, 28 (5), 538–549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (65).Kwang GD; Sampathkumar K; Loo SCJ Ultralong Floating Hydrogel Raft for Prolonged Gastric Retention. MRS Bull. 2022, DOI: 10.1557/s43577-022-00406-2. [DOI] [Google Scholar]
  • (66).Greenway FL; Aronne LJ; Raben A; Astrup A; Apovian CM; Hill JO; Kaplan LM; Fujioka K; Matejkova E; Svacina S; et al. A Randomized, Double-blind, Placebo-controlled Study of Gelesis100: A Novel Nonsystemic Oral Hydrogel for Weight Loss. Obesity 2019, 27 (2), 205–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (67).Jin X; Wei C; Wu C; Zhang W Gastric Fluid-Induced Double Network Hydrogel with High Swelling Ratio and Long-Term Mechanical Stability. Composites Part B: Engineering 2022, 236, 109816. [Google Scholar]
  • (68).Liu J; Pang Y; Zhang S; Cleveland C; Yin X; Booth L; Lin J; Lucy Lee Y-A; Mazdiyasni H; Saxton S; Kirtane AR; von Erlach T; Rogner J; Langer R; Traverso G Triggerable Tough Hydrogels for Gastric Resident Dosage Forms. Nat. Commun 2017, 8 (1), 124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (69).Liu X; Steiger C; Lin S; Parada GA; Liu J; Chan HF; Yuk H; Phan NV; Collins J; Tamang S; Traverso G; Zhao X Ingestible Hydrogel Device. Nat. Commun 2019, 10 (1), 493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (70).Xu X; Xia X; Zhang K; Rai A; Li Z; Zhao P; Wei K; Zou L; Yang B; Wong W-K; Chiu PW-Y; Bian L Bioadhesive Hydrogels Demonstrating PH-Independent and Ultrafast Gelation Promote Gastric Ulcer Healing in Pigs. Science Translational Medicine 2020, 12 (558), eaba8014. [DOI] [PubMed] [Google Scholar]
  • (71).Xue W; Yang R; Liu S; Pu Y; Wang P; Zhang W; Tan X; Chi B Ascidian-Inspired Aciduric Hydrogels with High Stretchability and Adhesiveness Promote Gastric Hemostasis and Wound Healing. Biomaterials Science 2022, 10 (9), 2417–2427. [DOI] [PubMed] [Google Scholar]
  • (72).Chen J; Caserto JS; Ang I; Shariati K; Webb J; Wang B; Wang X; Bouklas N; Ma M An Adhesive and Resilient Hydrogel for the Sealing and Treatment of Gastric Perforation. Bioactive Materials 2022, 14, 52–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (73).Chen X; Zhang J; Chen G; Xue Y; Zhang J; Liang X; Lei IM; Lin J; Xu BB; Liu J Hydrogel Bioadhesives with Extreme Acid-Tolerance for Gastric Perforation Repairing. Adv. Funct. Mater 2022, 32 (29), 2202285. [Google Scholar]
  • (74).Johansson MEV; Larsson JMH; Hansson GC The Two Mucus Layers of Colon Are Organized by the MUC2Mucin, Whereas the Outer Layer Is a Legislator of Host-Microbial Interactions. Proc. Natl. Acad. Sci. U. S. A 2011, 108, 4659–4665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (75).Atuma C; Strugala V; Allen A; Holm L The Adherent Gastrointestinal Mucus Gel Layer: Thickness and Physical State in Vivo. American Journal of Physiology-Gastrointestinal and Liver Physiology 2001, 280 (5), G922–G929. [DOI] [PubMed] [Google Scholar]
  • (76).Hansson GC Role of Mucus Layers in Gut Infection and Inflammation. Curr. Opin Microbiol 2012, 15 (1), 57–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (77).Swidsinski A; Loening-Baucke V; Theissig F; Engelhardt H; Bengmark S; Koch S; Lochs H; Dörffel Y Comparative Study of the Intestinal Mucus Barrier in Normal and Inflamed Colon. Gut 2007, 56 (3), 343–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (78).Van der Sluis M; De Koning BAE; De Bruijn ACJM; Velcich A; Meijerink JPP; Van Goudoever JB; Büller HA; Dekker J; Van Seuningen I; Renes IB; Einerhand AWC Muc2-Deficient Mice Spontaneously Develop Colitis, Indicating That MUC2 Is Critical for Colonic Protection. Gastroenterology 2006, 131 (1), 117–129. [DOI] [PubMed] [Google Scholar]
  • (79).Hosseinifar T; Sheybani S; Abdouss M; Hassani Najafabadi SA; Shafiee Ardestani M Pressure Responsive Nanogel Base on Alginate-cyclodextrin with Enhanced Apoptosis Mechanism for Colon Cancer Delivery. J. Biomed. Mater. Res., Part A 2018, 106 (2), 349–359. [DOI] [PubMed] [Google Scholar]
  • (80).Tang R-C; Chen T-C; Lin F-H Design Strategy for a Hydroxide-Triggered PH-Responsive Hydrogel as a Mucoadhesive Barrier to Prevent Metabolism Disorders. ACS Appl. Mater. Interfaces 2021, 13 (49), 58340–58351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (81).Fajardo AR; Silva MB; Lopes LC; Piai JF; Rubira AF; Muniz EC Hydrogel Based on an Alginate-Ca 2+/Chondroitin Sulfate Matrix as a Potential Colon-Specific Drug Delivery System. Rsc Advances 2012, 2 (29), 11095–11103. [Google Scholar]
  • (82).Hoang HT; Jo S-H; Phan Q-T; Park H; Park S-H; Oh C-W; Lim KT Dual PH-/Thermo-Responsive Chitosan-Based Hydrogels Prepared Using “Click” Chemistry for Colon-Targeted Drug Delivery Applications. Carbohydr. Polym 2021, 260, 117812. [DOI] [PubMed] [Google Scholar]
  • (83).Brøndsted H; Kopeček J Hydrogels for Site-Specific Drug Delivery to the Colon: In Vitro and in Vivo Degradation. Pharm. Res 1992, 9 (12), 1540–1545. [DOI] [PubMed] [Google Scholar]
  • (84).Shantha KL; Ravichandran P; Rao KP Azo Polymeric Hydrogels for Colon Targeted Drug Delivery. Biomaterials 1995, 16 (17), 1313–1318. [DOI] [PubMed] [Google Scholar]
  • (85).Koev TT; Harris HC; Kiamehr S; Khimyak YZ; Warren FJ Starch Hydrogels as Targeted Colonic Drug Delivery Vehicles. Carbohydr. Polym 2022, 289, 119413. [DOI] [PubMed] [Google Scholar]
  • (86).Hua S Physiological and Pharmaceutical Considerations for Rectal Drug Formulations. Frontiers in pharmacology 2019, 10, 1196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (87).Cranston RD; Lama JR; Richardson BA; Carballo-Diéguez A; Kunjara Na Ayudhya RP; Liu K; Patterson KB; Leu C-S; Galaska B; Jacobson CE; Parikh UM; Marzinke MA; Hendrix CW; Johnson S; Piper JM; Grossman C; Ho KS; Lucas J; Pickett J; Bekker L-G; Chariyalertsak S; Chitwarakorn A; Gonzales P; Holtz TH; Liu AY; Mayer KH; Zorrilla C; Schwartz JL; Rooney J; McGowan I MTN-017: A Rectal Phase 2 Extended Safety and Acceptability Study of Tenofovir Reduced-Glycerin 1% Gel. Clinical Infectious Diseases 2016, 64 (5), 614–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (88).Ilomuanya MO; Hameedat AT; Akang EN; Ekama SO; Silva BO; Akanmu AS Development and Evaluation of Mucoadhesive Bigel Containing Tenofovir and Maraviroc for HIV Prophylaxis. Future Journal of Pharmaceutical Sciences 2020, 6 (1), 81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (89).Xu J; Tam M; Samaei S; Lerouge S; Barralet J; Stevenson MM; Cerruti M Mucoadhesive Chitosan Hydrogels as Rectal Drug Delivery Vessels to Treat Ulcerative Colitis. Acta Biomaterialia 2017, 48, 247–257. [DOI] [PubMed] [Google Scholar]
  • (90).Ramadan AA; Elbakry AM; Esmaeil AH; Khaleel SA Pharmaceutical and Pharmacokinetic Evaluation of Novel Rectal Mucoadhesive Hydrogels Containing Tolmetin Sodium. Journal of Pharmaceutical Investigation 2018, 48 (6), 673–683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (91).Saltzman WM; Radomsky ML; Whaley KJ; Cone RA Antibody Diffusion in Human Cervical Mucus. Biophys. J 1994, 66 (2), 508–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (92).Huh CH; Bhutani MS; Farfán EB; Bolch WE Individual Variations in Mucosa and Total Wall Thickness in the Stomach and Rectum Assessed via Endoscopic Ultrasound. Physiological Measurement 2003, 24 (4), N15. [DOI] [PubMed] [Google Scholar]
  • (93).Vermani K; Garg S The Scope and Potential of Vaginal Drug Delivery. Pharmaceutical Science & Technology Today 2000, 3 (10), 359–364. [DOI] [PubMed] [Google Scholar]
  • (94).Cicinelli E; de Ziegler D; Morgese S; Bulletti C; Luisi D; Schonauer LM First Uterine Pass Effect” Is Observed When Estradiol Is Placed in the Upper but Not Lower Third of the Vagina. Fertility and Sterility 2004, 81 (5), 1414–1416. [DOI] [PubMed] [Google Scholar]
  • (95).Bulletti C; de Ziegler D; Flamigni C; Giacomucci E; Polli V; Bolelli G; Franceschetti F Targeted Drug Delivery in Gynaecology: The First Uterine Pass Effect. Hum. Reprod 1997, 12 (5), 1073–1079. [DOI] [PubMed] [Google Scholar]
  • (96).Ferguson LM; Rohan LC The Importance of the Vaginal Delivery Route for Antiretrovirals in HIV Prevention. Therapeutic Delivery 2011, 2 (12), 1535–1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (97).Major I; McConville C Vaginal Drug Delivery for the Localised Treatment of Cervical Cancer. Drug Delivery and Transl. Res 2017, 7 (6), 817–828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (98).Ordikhani F; Erdem Arslan M; Marcelo R; Sahin I; Grigsby P; Schwarz J; Azab A Drug Delivery Approaches for the Treatment of Cervical Cancer. Pharmaceutics 2016, 8 (3), 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (99).Wang X; Liu S; Guan Y; Ding J; Ma C; Xie Z Vaginal Drug Delivery Approaches for Localized Management of Cervical Cancer. Adv. Drug Delivery Rev 2021, 174, 114–126. [DOI] [PubMed] [Google Scholar]
  • (100).Fahrbach KM; Malykhina O; Stieh DJ; Hope TJ Differential Binding of IgG and IgA to Mucus of the Female Reproductive Tract. PLoS One 2013, 8 (10), No. e76176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (101).Lacroix G; Gouyer V; Gottrand F; Desseyn J-L The Cervicovaginal Mucus Barrier. IJMS 2020, 21 (21), 8266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (102).Wang Y-Y; Lai SK; Ensign LM; Zhong W; Cone R; Hanes J The Microstructure and Bulk Rheology of Human Cervicovaginal Mucus Are Remarkably Resistant to Changes in PH. Biomacromolecules 2013, 14 (12), 4429–4435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (103).FOX CA; MELDRUM SJ; WATSON BW Continuous Measurement by Radio-Telemetry of Vaginal pH during Human Coitus. Reproduction 1973, 33 (1), 69–75. [DOI] [PubMed] [Google Scholar]
  • (104).Roberton AM; Wiggins R; Horner PJ; Greenwood R; Crowley T; Fernandes A; Berry M; Corfield AP A Novel Bacterial Mucinase, Glycosulfatase, Is Associated with Bacterial Vaginosis. J. Clin Microbiol 2005, 43 (11), 5504–5508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (105).Borgdorff H; Gautam R; Armstrong SD; Xia D; Ndayisaba GF; van Teijlingen NH; Geijtenbeek TBH; Wastling JM; van de Wijgert JHHM Cervicovaginal Microbiome Dysbiosis Is Associated with Proteome Changes Related to Alterations of the Cervicovaginal Mucosal Barrier. Mucosal Immunol 2016, 9 (3), 621–633. [DOI] [PubMed] [Google Scholar]
  • (106).Hoang T; Toler E; DeLong K; Mafunda NA; Bloom SM; Zierden HC; Moench TR; Coleman JS; Hanes J; Kwon DS; Lai SK; Cone RA; Ensign LM The Cervicovaginal Mucus Barrier to HIV-1 Is Diminished in Bacterial Vaginosis. PLoS Pathog 2020, 16 (1), No. e1008236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (107).Musara P; Milford C; Shapley-Quinn MK; Weinrib R; Mutero P; Odoom E; Mgodi NM; Chirenje ZM; Hanif H; Clark MR; et al. Preferences and Acceptability of Vaginal Delivery Forms for HIV Prevention Among Women, Male Partners and Key Informants in South Africa and Zimbabwe: Qualitative Findings. AIDS and Behavior 2021, 25, 124–138. [DOI] [PubMed] [Google Scholar]
  • (108).Coggins C; Elias CJ; Atisook R; Bassett MT; Ettiègne-Traoré V; Ghys PD; Jenkins-Woelk L; Thongkrajai E; VanDevanter NL Women’s Preferences Regarding the Formulation of Over-the-Counter Vaginal Spermicides: AIDS 1998, 12 (11), 1389–1391. [DOI] [PubMed] [Google Scholar]
  • (109).Fry CH; Vahabi B The Role of the Mucosa in Normal and Abnormal Bladder Function. Basic & clinical pharmacology & toxicology 2016, 119, 57–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (110).Jafari NV; Rohn JL The Urothelium: A Multi-Faceted Barrier against a Harsh Environment. Mucosal Immunology 2022, 15 (6), 1127–1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (111).Cornish J; Lecamwasam J; Harrison G; Vanderwee M; Miller T Host Defence Mechanisms in the Bladder. II. Disruption of the Layer of Mucus. British journal of experimental pathology 1988, 69 (6), 759–770. [PMC free article] [PubMed] [Google Scholar]
  • (112).GuhaSarkar S; Banerjee R Intravesical Drug Delivery: Challenges, Current Status, Opportunities and Novel Strategies. J. Controlled Release 2010, 148 (2), 147–159. [DOI] [PubMed] [Google Scholar]
  • (113).Kolawole OM; Lau WM; Khutoryanskiy VV Chitosan/β-Glycerophosphate in Situ Gelling Mucoadhesive Systems for Intravesical Delivery of Mitomycin-C. International Journal of Pharmaceutics: X 2019, 1, 100007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (114).Lin T; Zhao X; Zhang Y; Lian H; Zhuang J; Zhang Q; Chen W; Wang W; Liu G; Guo S; et al. Floating Hydrogel with Self-Generating Micro-Bubbles for Intravesical Instillation. Materials 2016, 9 (12), 1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (115).Sun P; Wang Z; Wu T; Zuo S; Huang X; Cui Z; Zhang D Injectable, Adhesive, and Self-Healing Composite Hydrogels Loaded With Oxybutynin Hydrochloride for the Treatment of Overactive Bladder in Rats. Frontiers in Bioengineering and Biotechnology 2022, 10, 906835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (116).Goo YT; Yang HM; Kim CH; Kim MS; Kim HK; Chang IH; Choi YW Optimization of a Floating Poloxamer 407-Based Hydrogel Using the Box-Behnken Design: In Vitro Characterization and in Vivo Buoyancy Evaluation for Intravesical Instillation. European Journal of Pharmaceutical Sciences 2021, 163, 105885. [DOI] [PubMed] [Google Scholar]
  • (117).Ramseyer P; Meagher-Villemure K; Burki M; Frey P (Poly)Acrylonitrile-Based Hydrogel as a Therapeutic Bulking Agent in Urology. Biomaterials 2007, 28 (6), 1185–1190. [DOI] [PubMed] [Google Scholar]
  • (118).Qiu H; Li J; Huang Y; Shen C; Dai L; Su Q; Zhi Y; Fang Q; Shi C; Li W Sulfhydryl Functionalized Hyaluronic Acid Hydrogels Attenuate Cyclophosphamide-Induced Bladder Injury. Biomed. Mater 2023, 18 (1), 015026. [DOI] [PubMed] [Google Scholar]
  • (119).Ludwig A The Use of Mucoadhesive Polymers in Ocular Drug Delivery. Adv. Drug Deliv Rev 2005, 57 (11), 1595–1639. [DOI] [PubMed] [Google Scholar]
  • (120).Ugwoke MI; Agu RU; Verbeke N; Kinget R Nasal Mucoadhesive Drug Delivery: Background, Applications, Trends and Future Perspectives. Adv. Drug Deliv Rev 2005, 57 (11), 1640–1665. [DOI] [PubMed] [Google Scholar]
  • (121).Bernkop-Schnuürch A Mucoadhesive Systems in Oral Drug Delivery. Drug Discov Today Technol. 2005, 2 (1), 83–87. [DOI] [PubMed] [Google Scholar]
  • (122).Smart JD The Basics and Underlying Mechanisms of Mucoadhesion. Adv. Drug Deliv Rev 2005, 57 (11), 1556–1568. [DOI] [PubMed] [Google Scholar]
  • (123).Serra L; Doménech J; Peppas NA Engineering Design and Molecular Dynamics of Mucoadhesive Drug Delivery Systems as Targeting Agents. Eur. J. Pharm. Biopharm 2009, 71 (3), 519–528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (124).Bernkop-Schnuürch A Thiomers: A New Generation of Mucoadhesive Polymers. Adv. Drug Delivery Rev 2005, 57 (11), 1569–1582. [DOI] [PubMed] [Google Scholar]
  • (125).Andrews GP; Laverty TP; Jones DS Mucoadhesive Polymeric Platforms for Controlled Drug Delivery. Eur. J. Pharm. Biopharm 2009, 71 (3), 505–518. [DOI] [PubMed] [Google Scholar]
  • (126).Peppas NA; Huang Y Nanoscale Technology of Mucoadhesive Interactions. Adv. Drug Deliv Rev 2004, 56 (11), 1675–1687. [DOI] [PubMed] [Google Scholar]
  • (127).Grabovac V; Guggi D; Bernkop-Schnuürch A Comparison of the Mucoadhesive Properties of Various Polymers. Adv. Drug Deliv Rev 2005, 57 (11), 1713–1723. [DOI] [PubMed] [Google Scholar]
  • (128).Solomonidou D; Cremer K; Krumme M; Kreuter J Effect of Carbomer Concentration and Degree of Neutralization on the Mucoadhesive Properties of Polymer Films. Journal of Biomaterials Science, Polymer Edition 2001, 12 (11), 1191–1205. [DOI] [PubMed] [Google Scholar]
  • (129).Sogias IA; Williams AC; Khutoryanskiy VV Why Is Chitosan Mucoadhesive? Biomacromolecules 2008, 9 (7), 1837–1842. [DOI] [PubMed] [Google Scholar]
  • (130).Lehr C-M; Bouwstra JA; Schacht EH; Junginger HE In Vitro Evaluation of Mucoadhesive Properties of Chitosan and Some Other Natural Polymers. Int. J. Pharm 1992, 78 (1), 43–48. [Google Scholar]
  • (131).Bernkop-Schnuürch A; Kast CE; Richter MF Improvement in the Mucoadhesive Properties of Alginate by the Covalent Attachment of Cysteine. J. Controlled Release 2001, 71 (3), 277–285. [DOI] [PubMed] [Google Scholar]
  • (132).Laffleur F Comparative Mucoadhesive Study of Hyaluronic Acid-Based Conjugates on Different Mucosae. J. Appl. Polym. Sci 2018, 135 (14), 46071. [Google Scholar]
  • (133).Kim K; Kim K; Ryu JH; Lee H Chitosan-Catechol: A Polymer with Long-Lasting Mucoadhesive Properties. Biomaterials 2015, 52, 161–170. [DOI] [PubMed] [Google Scholar]
  • (134).Brannigan RP; Khutoryanskiy VV Progress and Current Trends in the Synthesis of Novel Polymers with Enhanced Mucoadhesive Properties. Macromol. Biosci 2019, 19 (10), 1900194. [DOI] [PubMed] [Google Scholar]
  • (135).Shtenberg Y; Goldfeder M; Schroeder A; Bianco-Peled H Alginate Modified with Maleimide-Terminated PEG as Drug Carriers with Enhanced Mucoadhesion. Carbohydr. Polym 2017, 175, 337–346. [DOI] [PubMed] [Google Scholar]
  • (136).Kaldybekov DB; Filippov SK; Radulescu A; Khutoryanskiy VV Maleimide-Functionalised PLGA-PEG Nanoparticles as Mucoadhesive Carriers for Intravesical Drug Delivery. Eur. J. Pharm. Biopharm 2019, 143, 24–34. [DOI] [PubMed] [Google Scholar]
  • (137).Shitrit Y; Bianco-Peled H Acrylated Chitosan for Mucoadhesive Drug Delivery Systems. Int. J. Pharm 2017, 517 (1), 247–255. [DOI] [PubMed] [Google Scholar]
  • (138).Davidovich-Pinhas M; Bianco-Peled H Novel Mucoadhesive System Based on Sulfhydryl-Acrylate Interactions. J. Mater. Sci.: Mater. Med 2010, 21 (7), 2027–2034. [DOI] [PubMed] [Google Scholar]
  • (139).Shan X; Aspinall S; Kaldybekov DB; Buang F; Williams AC; Khutoryanskiy VV Synthesis and Evaluation of Methacrylated Poly(2-Ethyl-2-Oxazoline) as a Mucoadhesive Polymer for Nasal Drug Delivery. ACS Appl. Polym. Mater 2021, 3 (11), 5882–5892. [Google Scholar]
  • (140).Kolawole OM; Lau WM; Khutoryanskiy VV Methacrylated Chitosan as a Polymer with Enhanced Mucoadhesive Properties for Transmucosal Drug Delivery. Int. J. Pharm 2018, 550 (1), 123–129. [DOI] [PubMed] [Google Scholar]
  • (141).Khutoryanskaya OV; Mayeva ZA; Mun GA; Khutoryanskiy VV Designing Temperature-Responsive Biocompatible Copolymers and Hydrogels Based on 2-Hydroxyethyl(Meth)-Acrylates. Biomacromolecules 2008, 9 (12), 3353–3361. [DOI] [PubMed] [Google Scholar]
  • (142).da Silva JB; Khutoryanskiy VV; Bruschi ML; Cook MT A Mucosa-Mimetic Material for the Mucoadhesion Testing of Thermogelling Semi-Solids. Int. J. Pharm 2017, 528 (1), 586–594. [DOI] [PubMed] [Google Scholar]
  • (143).Zhao L; Wang F; Cai Z; Zhou Q; Chen B; Zhang C; Liu H; Hong L; Zhang T; Zhong J; Cui W; Wang Z Improving Drug Utilization Platform with Injectable Mucoadhesive Hydrogel for Treating Ulcerative Colitis. Chem. Eng. J 2021, 424, 130464. [Google Scholar]
  • (144).Brotherton EE; Neal TJ; Kaldybekov DB; Smallridge MJ; Khutoryanskiy VV; Armes SP Aldehyde-Functional Thermoresponsive Diblock Copolymer Worm Gels Exhibit Strong Mucoadhesion. Chemical Science 2022, 13 (23), 6888–6898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (145).Fahy JV; Dickey BF Airway Mucus Function and Dysfunction. New England Journal of Medicine 2010, 363 (23), 2233–2247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (146).Kirchhof S; Goepferich AM; Brandl FP Hydrogels in Ophthalmic Applications. Eur. J. Pharm. Biopharm 2015, 95, 227–238. [DOI] [PubMed] [Google Scholar]
  • (147).Zignani M; Tabatabay C; Gurny R Topical Semi-Solid Drug Delivery: Kinetics and Tolerance of Ophthalmic Hydrogels. Adv. Drug Delivery Rev 1995, 16 (1), 51–60. [Google Scholar]
  • (148).Liu Z; Li J; Nie S; Liu H; Ding P; Pan W Study of an Alginate/HPMC-Based in Situ Gelling Ophthalmic Delivery System for Gatifloxacin. Int. J. Pharm 2006, 315 (1), 12–17. [DOI] [PubMed] [Google Scholar]
  • (149).Zheng W; Hao Y; Wang D; Huang H; Guo F; Sun Z; Shen P; Sui K; Yuan C; Zhou Q Preparation of Triamcinolone Acetonide-Loaded Chitosan/Fucoidan Hydrogel and Its Potential Application as an Oral Mucosa Patch. Carbohydr. Polym 2021, 272, 118493. [DOI] [PubMed] [Google Scholar]
  • (150).Bertsch P; Schneider L; Bovone G; Tibbitt MW; Fischer P; Gstöhl S Injectable Biocompatible Hydrogels from Cellulose Nanocrystals for Locally Targeted Sustained Drug Release. ACS Appl. Mater. Interfaces 2019, 11 (42), 38578–38585. [DOI] [PubMed] [Google Scholar]
  • (151).Hattori H; Tsujimoto H; Hase K; Ishihara M Characterization of a Water-Soluble Chitosan Derivative and Its Potential for Submucosal Injection in Endoscopic Techniques. Carbohydr. Polym 2017, 175, 592–600. [DOI] [PubMed] [Google Scholar]
  • (152).Wang H; Wang L; Guo S; Liu Z; Zhao L; Qiao R; Li C Rutin-Loaded Stimuli-Responsive Hydrogel for Anti-Inflammation. ACS Appl. Mater. Interfaces 2022, 14 (23), 26327–26337. [DOI] [PubMed] [Google Scholar]
  • (153).Yeruva T; Lee CH Enzyme Responsive Delivery of Anti-Retroviral Peptide via Smart Hydrogel. AAPS PharmSciTech 2022, 23 (7), 234. [DOI] [PubMed] [Google Scholar]
  • (154).Yeruva TK Smart Hydrogel for Enzyme Responsive Vaginal Delivery of Anti-HIV Peptide Therapeutics; University of Missouri—Kansas City: 2021. [Google Scholar]
  • (155).Chen L; Tian Z; Du Y Synthesis and PH Sensitivity of Carboxymethyl Chitosan-Based Polyampholyte Hydrogels for Protein Carrier Matrices. Biomaterials 2004, 25 (17), 3725–3732. [DOI] [PubMed] [Google Scholar]
  • (156).Basan H; Guümuüşdereliogğlu M; Tevfik Orbey M Release Characteristics of Salmon Calcitonin from Dextran Hydrogels for Colon-Specific Delivery. Eur. J. Pharm. Biopharm 2007, 65 (1), 39–46. [DOI] [PubMed] [Google Scholar]
  • (157).Van Tomme SR; Storm G; Hennink WE In Situ Gelling Hydrogels for Pharmaceutical and Biomedical Applications. Int. J. Pharm 2008, 355 (1), 1–18. [DOI] [PubMed] [Google Scholar]
  • (158).Liu Y; Yang F; Feng L; Yang L; Chen L; Wei G; Lu W In Vivo Retention of Poloxamer-Based in Situ Hydrogels for Vaginal Application in Mouse and Rat Models. Acta Pharmaceutica Sinica B 2017, 7 (4), 502–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (159).Li C; Li C; Liu Z; Li Q; Yan X; Liu Y; Lu W Enhancement in Bioavailability of Ketorolac Tromethamine via Intranasal in Situ Hydrogel Based on Poloxamer 407 and Carrageenan. Int. J. Pharm 2014, 474 (1), 123–133. [DOI] [PubMed] [Google Scholar]
  • (160).Zhu M; Wang J; Li N A Novel Thermo-Sensitive Hydrogel-Based on Poly(N-Isopropylacrylamide)/Hyaluronic Acid of Ketoconazole for Ophthalmic Delivery. Artificial Cells, Nanomedicine, and Biotechnology 2018, 46 (6), 1282–1287. [DOI] [PubMed] [Google Scholar]
  • (161).Liu Y; Zhu Y; Wei G; Lu W Effect of Carrageenan on Poloxamer-Based in Situ Gel for Vaginal Use: Improved in Vitro and in Vivo Sustained-Release Properties. European Journal of Pharmaceutical Sciences 2009, 37 (3), 306–312. [DOI] [PubMed] [Google Scholar]
  • (162).Asfour MH; Abd El-Alim SH; Awad GEA; Kassem AA Chitosan/β-Glycerophosphate in Situ Forming Thermo-Sensitive Hydrogel for Improved Ocular Delivery of Moxifloxacin Hydrochloride. European Journal of Pharmaceutical Sciences 2021, 167, 106041. [DOI] [PubMed] [Google Scholar]
  • (163).Srividya B; Cardoza RM; Amin PD Sustained Ophthalmic Delivery of Ofloxacin from a PH Triggered in Situ Gelling System. J. Controlled Release 2001, 73 (2), 205–211. [DOI] [PubMed] [Google Scholar]
  • (164).Rupenthal ID; Green CR; Alany RG Comparison of Ion-Activated in Situ Gelling Systems for Ocular Drug Delivery. Part 2: Precorneal Retention and in Vivo Pharmacodynamic Study. Int. J. Pharm 2011, 411 (1), 78–85. [DOI] [PubMed] [Google Scholar]
  • (165).Janga KY; Tatke A; Balguri SP; Lamichanne SP; Ibrahim MM; Maria DN; Jablonski MM; Majumdar S Ion-Sensitive in Situ Hydrogels of Natamycin Bilosomes for Enhanced and Prolonged Ocular Pharmacotherapy: In Vitro Permeability, Cytotoxicity and in Vivo Evaluation. Artificial Cells, Nanomedicine, and Biotechnology 2018, 46 (sup1), 1039–1050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (166).Li P; Wang S; Chen H; Zhang S; Yu S; Li Y; Cui M; Pan W; Yang X A Novel Ion-Activated in Situ Gelling Ophthalmic Delivery System Based on κ-Carrageenan for Acyclovir. Drug Dev. Ind. Pharm 2018, 44 (5), 829–836. [DOI] [PubMed] [Google Scholar]
  • (167).Sagar P; Singh V; Gupta R; Kaul S; Sharma S; Kaur S; Bhunia RK; Kondepudi KK; Singhal NK PH-Triggered, Synbiotic Hydrogel Beads for In Vivo Therapy of Iron Deficiency Anemia and Reduced Inflammatory Response. ACS Appl. Bio Mater 2021, 4 (10), 7467–7484. [DOI] [PubMed] [Google Scholar]
  • (168).Jiang H; Yu W; Oscai M; Ziaie B A Smart Capsule With a Hydrogel-Based PH-Triggered Release Switch for GI-Tract Site-Specific Drug Delivery. IEEE Transactions on Biomedical Engineering 2018, 65 (12), 2808–2813. [DOI] [PubMed] [Google Scholar]
  • (169).Jiang H; Yu W; Zhou J; Ziaie B A pH-Sensitive Hydrogel-Based Smart Switch for GI-Tract Payload Release. In 2017 IEEE 30th International Conference on Micro Electro Mechanical Systems (MEMS); IEEE: 2017; pp 510–513; DOI: 10.1109/MEMSYS.2017.7863455. [DOI] [Google Scholar]
  • (170).Eljarrat-Binstock E; Raiskup F; Frucht-Pery J; Domb AJ Transcorneal and Transscleral Iontophoresis of Dexamethasone Phosphate Using Drug Loaded Hydrogel. J. Controlled Release 2005, 106 (3), 386–390. [DOI] [PubMed] [Google Scholar]
  • (171).Ayehunie S; Wang Y-Y; Landry T; Bogojevic S; Cone RA Hyperosmolal Vaginal Lubricants Markedly Reduce Epithelial Barrier Properties in a Three-Dimensional Vaginal Epithelium Model. Toxicology Reports 2018, 5, 134–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (172).Fuchs EJ; Lee LA; Torbenson MS; Parsons TL; Bakshi RP; Guidos AM; Wahl RL; Hendrix CW Hyperosmolar Sexual Lubricant Causes Epithelial Damage in the Distal Colon: Potential Implication for HIV Transmission. J. INFECT DIS 2007, 195 (5), 703–710. [DOI] [PubMed] [Google Scholar]
  • (173).Ensign LM; Hoen TE; Maisel K; Cone RA; Hanes JS Enhanced Vaginal Drug Delivery through the Use of Hypotonic Formulations That Induce Fluid Uptake. Biomaterials 2013, 34 (28), 6922–6929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (174).Maisel K; Chattopadhyay S; Moench T; Hendrix C; Cone R; Ensign LM; Hanes J Enema Ion Compositions for Enhancing Colorectal Drug Delivery. J. Controlled Release 2015, 209, 280–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (175).Kim YC; Shin MD; Hackett SF; Hsueh HT; Lima e Silva R; Date A; Han H; Kim B-J; Xiao A; Kim Y; Ogunnaike L; Anders NM; Hemingway A; He P; Jun AS; McDonnell PJ; Eberhart C; Pitha I; Zack DJ; Campochiaro PA; Hanes J; Ensign LM Gelling Hypotonic Polymer Solution for Extended Topical Drug Delivery to the Eye. Nature Biomedical Engineering 2020, 4 (11), 1053–1062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (176).Lin T; Wu J; Zhao X; Lian H; Yuan A; Tang X; Zhao S; Guo H; Hu Y In Situ Floating Hydrogel for Intravesical Delivery of Adriamycin without Blocking Urinary Tract. Journal of pharmaceutical sciences 2014, 103 (3), 927–936. [DOI] [PubMed] [Google Scholar]
  • (177).Zhu G; Zhang Y; Wang K; Zhao X; Lian H; Wang W; Wang H; Wu J; Hu Y; Guo H Visualized Intravesical Floating Hydrogel Encapsulating Vaporized Perfluoropentane for Controlled Drug Release. Drug Delivery 2016, 23 (8), 2820–2826. [DOI] [PubMed] [Google Scholar]
  • (178).Yom-Tov O; Seliktar D; Bianco-Peled H A Modified Emulsion Gelation Technique to Improve Buoyancy of Hydrogel Tablets for Floating Drug Delivery Systems. Materials Science and Engineering: C 2015, 55, 335–342. [DOI] [PubMed] [Google Scholar]
  • (179).Murata Y; Sasaki N; Miyamoto E; Kawashima S Use of Floating Alginate Gel Beads for Stomach-Specific Drug Delivery. Eur. J. Pharm. Biopharm 2000, 50 (2), 221–226. [DOI] [PubMed] [Google Scholar]
  • (180).Budianto E; Al-Shidqi M; Cahyana A Effects of Pore Forming Agents on Chitosan-Graft-Poly (N-Vinylpyrrolidone) Hydrogel Properties for Use as a Matrix for Floating Drug Delivery; IOP Publishing: 2017; Vol. 223, p 012038. [Google Scholar]
  • (181).Huang X; Liu L; Wang J-L; Bi M; Liu C; Liao W; Wang P; Liu J; Hou Z; Xu Z; Ye F Designing Self-Floating Anisotropic Macroporous Hydrogel by Step Emulsification and Buoyancy-Assisted Microfluidics. Chem. Eng. J 2022, 446, 137348. [Google Scholar]
  • (182).Oliveira NM; Zhang YS; Ju J; Chen A-Z; Chen Y; Sonkusale SR; Dokmeci MR; Reis RL; Mano JF; Khademhosseini A Hydrophobic Hydrogels: Toward Construction of Floating (Bio)Microdevices. Chem. Mater 2016, 28 (11), 3641–3648. [Google Scholar]
  • (183).van der Post S; Jabbar KS; Birchenough G; Arike L; Akhtar N; Sjovall H; Johansson MEV; Hansson GC Structural Weakening of the Colonic Mucus Barrier Is an Early Event in Ulcerative Colitis Pathogenesis. Gut 2019, 68 (12), 2142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (184).Monajemi H; Meenan J; Lamping R; Obradov D; Radema S; Trown P; Tytgat G; Van Deventer S Inflammatory Bowel Disease Is Associated with Increased Mucosal Levels of Bactericidal/Permeability-Increasing Protein. Gastroenterology 1996, 110 (3), 733–739. [DOI] [PubMed] [Google Scholar]
  • (185).Zhang S; Ermann J; Succi MD; Zhou A; Hamilton MJ; Cao B; Korzenik JR; Glickman JN; Vemula PK; Glimcher LH; Traverso G; Langer R; Karp JM An Inflammation-Targeting Hydrogel for Local Drug Delivery in Inflammatory Bowel Disease. Sci. Transl. Med 2015, 7 (300), 300ra128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (186).Diaz-Salmeron R; Toussaint B; Huang N; Bourgeois Ducournau E; Alviset G; Goulay Dufaÿ S; Hillaireau H; Dufaÿ Wojcicki A; Boudy V Mucoadhesive Poloxamer-Based Hydrogels for the Release of HP-β-CD-Complexed Dexamethasone in the Treatment of Buccal Diseases. Pharmaceutics 2021, 13 (1), 117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (187).Schilling AL; Kulahci Y; Moore J; Wang EW; Lee SE; Little SR A Thermoresponsive Hydrogel System for Long-Acting Corticosteroid Delivery into the Paranasal Sinuses. J. Controlled Release 2021, 330, 889–897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (188).Chen Z; Cai Z; Zhu C; Song X; Qin Y; Zhu M; Zhang T; Cui W; Tang H; Zheng H Injectable and Self-Healing Hydrogel with Anti-Bacterial and Anti-Inflammatory Properties for Acute Bacterial Rhinosinusitis with Micro Invasive Treatment. Adv. Healthcare Mater 2020, 9 (20), 2001032. [DOI] [PubMed] [Google Scholar]
  • (189).Bansal M; Mittal N; Yadav SK; Khan G; Gupta P; Mishra B; Nath G Periodontal Thermoresponsive, Mucoadhesive Dual Antimicrobial Loaded in-Situ Gel for the Treatment of Periodontal Disease: Preparation, in-Vitro Characterization and Antimicrobial Study. Journal of Oral Biology and Craniofacial Research 2018, 8 (2), 126–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (190).Hamed R; AbuRezeq A; Tarawneh O Development of Hydrogels, Oleogels, and Bigels as Local Drug Delivery Systems for Periodontitis. Drug Dev. Ind. Pharm 2018, 44 (9), 1488–1497. [DOI] [PubMed] [Google Scholar]
  • (191).Ho HN; Le HH; Le TG; Duong THA; Ngo VQT; Dang CT; Nguyen VM; Tran TH; Nguyen CN Formulation and Characterization of Hydroxyethyl Cellulose-Based Gel Containing Metronidazole-Loaded Solid Lipid Nanoparticles for Buccal Mucosal Drug Delivery. Int. J. Biol. Macromol 2022, 194, 1010–1018. [DOI] [PubMed] [Google Scholar]
  • (192).Wróblewska M; Szymańska E; Szekalska M; Winnicka K Different Types of Gel Carriers as Metronidazole Delivery Systems to the Oral Mucosa. Polymers 2020, 12 (3), 680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (193).Şenel S; İkinci G; Kaş S; Yousefi-Rad A; Sargon MF; Hıncal AA Chitosan Films and Hydrogels of Chlorhexidine Gluconate for Oral Mucosal Delivery. Int. J. Pharm 2000, 193 (2), 197–203. [DOI] [PubMed] [Google Scholar]
  • (194).Mirza MA; Panda AK; Asif S; Verma D; Talegaonkar S; Manzoor N; Khan A; Ahmed FJ; Dudeja M; Iqbal Z A Vaginal Drug Delivery Model. Drug Delivery 2016, 23 (8), 3123–3134. [DOI] [PubMed] [Google Scholar]
  • (195).Hong L; Chen G; Cai Z; Liu H; Zhang C; Wang F; Xiao Z; Zhong J; Wang L; Wang Z; Cui W Balancing Microthrombosis and Inflammation via Injectable Protein Hydrogel for Inflammatory Bowel Disease. Advanced Science 2022, 9 (20), 2200281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (196).Varma DM; Batty CJ; Stiepel RT; Graham-Gurysh EG; Roque JAI; Pena ES; Hasan Zahid MS; Qiu K; Anselmo A; Hill DB; Ross TM; Bachelder EM; Ainslie KM Development of an Intranasal Gel for the Delivery of a Broadly Acting Subunit Influenza Vaccine. ACS Biomater. Sci. Eng 2022, 8 (4), 1573–1582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (197).Rupenthal ID; Alany RG; Green CR Ion-Activated In Situ Gelling Systems for Antisense Oligodeoxynucleotide Delivery to the Ocular Surface. Mol. Pharmaceutics 2011, 8 (6), 2282–2290. [DOI] [PubMed] [Google Scholar]
  • (198).Duran-Mota JA; Yani JQ; Almquist BD; Borrós S; Oliva N Polyplex-Loaded Hydrogels for Local Gene Delivery to Human Dermal Fibroblasts. ACS Biomater. Sci. Eng 2021, 7 (9), 4347–4361. [DOI] [PubMed] [Google Scholar]
  • (199).Burdick JA; Mauck RL; Gerecht S To Serve and Protect: Hydrogels to Improve Stem Cell-Based Therapies. Cell Stem Cell 2016, 18 (1), 13–15. [DOI] [PubMed] [Google Scholar]
  • (200).Murali VP; Holmes CA Biomaterial-Based Extracellular Vesicle Delivery for Therapeutic Applications. Acta Biomaterialia 2021, 124, 88–107. [DOI] [PubMed] [Google Scholar]
  • (201).Yu S; Sun H; Li Y; Wei S; Xu J; Liu J Hydrogels as Promising Platforms for Engineered Living Bacteria-Mediated Therapeutic Systems. Materials Today Bio 2022, 16, 100435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (202).Chung L; Maestas DR; Housseau F; Elisseeff JH Key Players in the Immune Response to Biomaterial Scaffolds for Regenerative Medicine. Adv. Drug Delivery Rev 2017, 114, 184–192. [DOI] [PubMed] [Google Scholar]
  • (203).Keane TJ; Dziki J; Sobieski E; Smoulder A; Castleton A; Turner N; White LJ; Badylak SF Restoring Mucosal Barrier Function and Modifying Macrophage Phenotype with an Extracellular Matrix Hydrogel: Potential Therapy for Ulcerative Colitis. J. Crohn’s Colitis 2016, 11 (3), 360–368. [DOI] [PubMed] [Google Scholar]

RESOURCES