2,095
Views
3
CrossRef citations to date
0
Altmetric
Research Article

Responsive nanosystems for targeted therapy of ulcerative colitis: Current practices and future perspectives

, , &
Article: 2219427 | Received 30 Jan 2023, Accepted 20 May 2023, Published online: 08 Jun 2023

Abstract

The pharmacological approach to treating gastrointestinal diseases is suffering from various challenges. Among such gastrointestinal diseases, ulcerative colitis manifests inflammation at the colon site specifically. Patients suffering from ulcerative colitis notably exhibit thin mucus layers that offer increased permeability for the attacking pathogens. In the majority of ulcerative colitis patients, the conventional treatment options fail in controlling the symptoms of the disease leading to distressing effects on the quality of life. Such a scenario is due to the failure of conventional therapies to target the loaded moiety into specific diseased sites in the colon. Targeted carriers are needed to address this issue and enhance the drug effects. Conventional nanocarriers are mostly readily cleared and have nonspecific targeting. To accumulate the desired concentration of the therapeutic candidates at the inflamed area of the colon, smart nanomaterials with responsive nature have been explored recently that include pH responsive, reactive oxygen species responsive (ROS), enzyme responsive and thermo – responsive smart nanocarrier systems. The formulation of such responsive smart nanocarriers from nanotechnology scaffolds has resulted in the selective release of therapeutic drugs, avoiding systemic absorption and limiting the undesired delivery of targeting drugs into healthy tissues. Recent advancements in the field of responsive nanocarrier systems have resulted in the fabrication of multi-responsive systems i.e. dual responsive nanocarriers and derivitization that has increased the biological tissues and smart nanocarrier’s interaction. In addition, it has also led to efficient targeting and significant cellular uptake of the therapeutic moieties. Herein, we have highlighted the latest status of the responsive nanocarrier drug delivery system, its applications for on–demand delivery of drug candidates for ulcerative colitis, and the prospects are underpinned.

1. Introduction

Ulcerative colitis is a recurrent and chronic inflammatory condition with debilitative effects on patients’ normal physiology (Mandlik et al. Citation2021). It affects the gastrointestinal tract by distributing in the distal portion of the colon. The incidence of the recurrent type ulcerative colitis is relatively high and thus to control the progression of the ailment, long-term and regular treatment is required (Kucharzik et al. Citation2020). Antibiotics, sulfasalazine, Probiotics, immunosuppressive agents, and salicylic acid derivatives are the common medications used for the treatment of UC currently (Ni et al. Citation2017). However, in the existing therapies for ulcerative colitis, there are several limitations and potentially worse side effects that limit their use in clinical settings. Accordingly, the therapeutic entities from natural sources such as polysaccharides, polyphenols, and flavonoids have shown somehow safer profiles with minimal side effects with progression inhibition of ulcerative colitis (Chen Z et al. Citation2021; Pang et al. Citation2022).

To proceed with the therapy outcomes for ulcerative colitis, recently nanotechnology-based approaches have been exploited that have shown bright prospects by boosting drug solubility, bioavailability, and pharmacokinetics (Zhang, Wang, et al. Citation2022). In addition, such nanoparticles were surface modified through the introduction of ligands in order to target specific sites of the colon surface and subsequently avoid off-target drug release, side effects and minimize toxicity (Chen L et al. Citation2021). In this context, hyaluronic acid-modified nanoparticles were evaluated that resulted in the release of loaded cargo following endocytosis and also targeted macrophages with the CD44 mediation during ulcerative colitis treatment (Liu P et al. Citation2021; Hlaing et al. Citation2022). It was also found that degradation, burst drug release, and early GIT uptake of the nanoparticles failed in ulcerative colitis therapy. A colonic intelligent response-based polysaccharide microparticulate system was developed and evaluated for pH sensitivity, colonic microbial activation, and time response and results showed a joint strategy for the smooth release of the drug in the colon (Zu et al. Citation2021). The Food and Drug Administration (FDA) has approved chitosan and alginates as natural and safe biopolymers and therefore are used extensively as delivery carriers for drugs in nanotechnology. The positive charge chitosan and negative charge alginates attract each other in solution and could be used for the fabrication of microparticles that could be targeted to the colon. To circumvent nanoparticles’ limitations of pre-mature clearance, they could be encapsulated in the microparticles for the oral delivery of cargo to target the colon (Ling et al. Citation2019). Such fabrication provides greater area and depot in terms of nanoparticles that could become stimulus-responsive after certain engineering processes. Such chemistry of nanoparticles within the microparticles for oral delivery integrates two particulate mechanisms (Naeem et al. Citation2020). Furthermore, these systems protect the nanoparticles from enzymatic degradation until it safely reaches the absorption site.

The targeting of ulcerative colitis tissue is achieved through various nanotechnology scaffolds that protect the nanosystems as well as loaded cargoes through encapsulation, and instability in the gastric transition is prevented along with eliminating the premature drug release. It also promotes the nanoparticle release profile during the course of therapy. Thus responsive oral delivery system from nanotechnology platform for ulcerative colitis treatment presents a promising approach in terms of specific localization and intestinal delivery (Wang et al. Citation2021). This review article aims to highlight the current status of ulcerative colitis targeted therapies using responsive nanosystems to overcome the main challenges/barriers associated with conventional targeted therapy as well as non-targeted therapies. The future directions for the safe and effective use of responsive nanosystems are also underpinned as a part of this review article.

2. Pathophysiology of ulcerative colitis

The gastrointestinal mucosa comes in contact with millions of antigens usually from the microbiome, the surrounding environment, and the ingested food (Fändriks Citation2017). The gut immune system is protected by a thick layer of mucin that lies above the epithelium being the most exposed layer of the GIT mucosa (Grondin et al. Citation2020). The mucin layer not only provides a partition between immune cells of the GIT and antigens but also exhibits antimicrobial potential (Donaldson et al. Citation2016). In the case of ulcerative colitis, mucin synthesis and secretion is impaired, and the resultant epithelial injury causes the pathogens to penetrate inside due to high permeability of the injured mucosa. This subsequently leads to the gut immune system stimulation by increased uptake of pathogens (antigens) (Cantero-Recasens et al. Citation2022; Wan and Zhang Citation2022). The colonocytes – epithelial cells of the colon are highly implicated in ulcerative colitis pathogenesis that’s why ulcerative colitis is confined mainly to the mucosal and sub-mucosal layers of the colon (Thomas et al. Citation2019). It is believed that PPARg – a nuclear receptor is abnormally expressed in case of ulcerative colitis that leads to the down-regulation of inflammation (Toumi et al. Citation2021). The overall scheme of ulcerative colitis pathophysiology is depicted in .

Figure 1. Pathophysiology of Ulcerative colitis.

Figure 1. Pathophysiology of Ulcerative colitis.

The innate immune response is mainly activated by certain antigens such as T cells and antigen-presenting cells that also activate the adaptive immune system and eventually stimulate the inflammatory cascades (Ordás Citation2012). In ulcerative colitis, apart from pre – mentioned initiators the sensitivity and activation of mature dendritic also take part in the induction of inflammation as well (Tatiya-Aphiradee et al. Citation2018). Furthermore, the expression of these mature dendritic cells leads to the activation of a lot of Toll – like receptors that in turn provide recognition pattern to pathogens for the multiple transcription factor signal activation which finally starts various inflammatory cascades (Lee and Kim Citation2007). In turn, the triggered inflammatory cascades results in proinflammatory cytokine production particularly interleukins and tumor necrosis factor alpha that transduce message across the intracellular proteins notably Janus kinases that in response potentiate the activation of lymphocytes and proliferation (Danese et al. Citation2016; D’Amico et al. Citation2022). These findings suggest that such intracellular proteins and proinflammatory cytokines in the currently available treatments of ulcerative colitis act as suitable targets. In ulcerative colitis, there is also an imbalance between regulatory and effector T – cells that lead to dysregulation of the adaptive immune system (Xu X-R et al. Citation2014). Among the effector cells, the T – helper 2 cells in the colon activate natural killer T cells that upon stimulation trigger the secretion of various cytokines i.e. interleukin − 13, which results in the apoptosis induction within epithelial cells and hence leads to tight junction disturbances (Heller et al. Citation2008). The inflamed mucosa also attracts circulating leukocytes that mimic the inflammatory responses and such effect is mostly carried out by cytokines, which in the vascular endothelium trigger the adhesion molecule expression and thus promote the extravasations and adhesion of leukocytes into the tissues (Claesson-Welsh et al. Citation2021).

3. Current therapeutic approach for UC and limitations

Due to uncertainties about the etiology of the disease, it currently has limited therapeutic success. Therapeutic approaches, therefore, aim to achieve and sustain remission from inflammatory episodes. Anti-inflammatory medications such as 5-aminosalicylates (5-ASAs) e.g. mesalamine and corticosteroids, immuno-suppressants such as 6-mercaptopurine, azathioprine, methotrexate, and tacrolimus, and biologic anti- TNF (tumor necrosis factor) agents i.e. infliximab, golimumab, and adalimumab are some of the commonly prescribed medications (Yang et al. Citation2020). Several recently developed medications are also awaiting marketing approval, such as natalizumab, a leukocyte inhibitor, and several pro-inflammatory cytokines inhibitors such as interleukin 12 and 23 (IL-12 and IL-23) (Neurath Citation2017; Pagnini et al. Citation2019).

The treatment approach changes considerably with the type and progression of UC. There are three main types of UC namely, Mild-Moderate, Moderate-Severe, and Acute severe UC. Clinically, mild-moderate UC is defined as having four to six bowel movements per day with mild-moderate rectal bleeding, without constitutional symptoms like fever and tachycardia, and without laboratory abnormalities like anemia or elevated inflammatory markers (Kayal and Shah Citation2019). Mesalamines are the first-line treatment for mild-to-moderate UC remission induction. Sulfasalazine is a suitable substitute for mesalamine in patients with severe arthritic symptoms, though it is frequently poorly tolerated due to side effects like headache, nausea, diarrhea, and rash (Ko et al. Citation2019). The other moderate-severe type of UC is characterized by daily 4 to 6 bowel movements and moderate to severe rectal bleeding without any constitutional symptoms (Sc and Lj Citation1955). In addition to the small-molecule Janus kinase (JAK) inhibitor tofacitinib, biologics are frequently employed to treat this moderate-sever type of UC. The acute-severe UC requires hospitalization as it is a life-threatening condition. This type of UC is characterized by symptoms tachycardia (>90 bpm), fever (>37.8 °C), decreased hemoglobin (around 10.5 gm/dL), and/or raised erythrocyte sedimentation rate (ESR >30 mm/h) and at least six bloody bowel movements per day. Cyclosporine, Steroids and infliximab are currently accepted medical treatments for people with this type of UC who are being treated in a hospital (Lynch et al. Citation2013). A general schematic diagram for the most commonly used drugs class based on severity of the disease is given in .

Figure 2. Schematic diagram for the commonly used classes of drugs based on severity of the UC.

Figure 2. Schematic diagram for the commonly used classes of drugs based on severity of the UC.

Although these medications can reduce inflammation to some extent, 30 to 50% of patients still experience little to no improvement. In addition, long-term administration increases the likelihood of relapse and multiple adverse effects (), thus limiting further clinical applications (Mohan et al. Citation2019). Therefore, there is an urgent need to develop new treatment strategies to enhance targeting and reduce side effects. Decreasing drug-related side effects, effectively targeting drugs to the colonic region of the GIT, and increasing the drug concentration at the particular site are major challenges in treating UC. The main therapeutic benefits of using nanocarriers based drug delivery for the treatment of colonic diseases include: high drug concentration in the colonic inflamed tissues, which results in better therapeutic effects; a reduction in drug-related side effects due to a decreased systemic absorption of active agents; need of smaller drug dose to achieve the desired therapeutic outcome because higher local concentration is achieved; and a significantly increased patient compliance.

Table 1. Examples of the major drugs used for ulcerative colitis with their limitations.

4. Barriers/challenges in successful therapy of ulcerative colitis

In the successful treatment of ulcerative colitis, there are various barriers that should be considered during the therapy and drug delivery that are mainly due to physiological and pharmaceutical factors. The drug delivery system must overcome all obstacles in the stomach and small intestine to reach the colon in the proper form. Major factors to be considered for successful therapy of UC are listed below:

4.1. Gastrointestinal (GI) motility and pH

Transit time in colon and intestine is one major challenge from physiological point of view. In this regard 21 healthy volunteers were evaluated for transit time and results showed an average transit time of 53 hours. As compared to other parts of gastrointestinal tract the colonic passage time was 40 hours (Sardo et al. Citation2019). Moreover, to hit ileocecal junction, duration of 3 – 4 hours is required for the administered dosage form (Sarvar et al. Citation2016). After an oral drug administration, the passage of bowl and gastric emptying as well pose a profound effect on the drug delivery. Here the matter of issue when the administered drug enters the stomach and before reaching the duodenum the time spent by it in the stomach (Caster et al. Citation2019). Furthermore, after administration of the drug, the process of gastric emptying varies with variation in the stomach phases. Particle size of the drug after reaching the colon plays an important role and decides the drug transit time i.e. small particles traverse colon faster than large particles and time taken ranges from 5/10 minutes up to two hours (Gauri et al. Citation2011). It shows that a drug delivery system targeted to colon should spent sometime in the stomach and so the release of drug at a far place from the colon will manifest an efficient delivery system. A small transit time is shown by diarrhea patients while larger by constipation patients (Deng et al. Citation2019). Drugs with delayed release (enteric coatings) kinetics and its association with the gastric pH is an important factor during drug delivery. A through look into the pH window of GIT shows that, in a healthy individual the stomach pH ranges from 1.5 − 3.5, while this acidic pH converts into basic pH down the duodenum (pH 6) reaches to 7.4 in small intestine and eventually drops to nearly 5.5 in the colon (Gaohua et al. Citation2021). Of note, colonic pH is considered as one major factor during the design of colonic delivery system because it is involved in the release of drug from the dosage form. In this regard, polymers are used to target the drug to a specific location and also to protect it from the harsh gastric environment (Verma et al. Citation2012).

4.2. Gastrointestinal enzymes and microflora

The metabolism process in the body is accelerated by certain intestinal enzymes that are secreted by certain microbial flora in the gastrointestinal tract. Such microbial floras also help in the eradication of several gastrointestinal disorders such as irritable bowel disease and thus have a significant impact on health (Garbern et al. Citation2011). The growth of existing microbial flora is controlled by the gastrointestinal contents and peristaltic movements. The terminal area of ileum is rich in microbial flora and thus it enrich the colon as well (Peng C-L et al. Citation2010). The drug from dosage form is released into the colon through the action of certain enzymes i.e. azoreductase, glycosidase etc. that is released by microbial flora into the colon. Moreover, gut flora have the ability to hydrolyze the certain polysaccharides, thus delivery polymers of this nature can be hydrolyzed by floral enzymes that will deliver the drug cargo efficiently (Nicholls et al. Citation2013). Therefore, selection of the polymers/other components liable to microbial degradation should be considered for proper release of the loaded contents in the right targeted site of the GI.

4.3. Mucus layer of the GI

Highly glycosylated material known as mucin is the mucus layer of the GI that works as a pseudo physical barrier for some large molecular drugs and that must be overcome by orally administered drugs (Xiao and Merlin Citation2012). Mucin is a hydrogel type mixture consisting of lipids, bacteria, proteins (including antibodies), carbohydrates, and inorganic salts. Mucin fibers, which make up the dense network of mucus and contain highly glycosylated, negatively charged segments, are the foundation of mucus’ barrier qualities. Additionally, mucins have regularly distributed hydrophobic domains that have a strong affinity for attaching hydrophobic particles. Whether a medication can enter and be absorbed by epithelial cells depends on how well a carrier interacts with mucus (Yang et al. Citation2014). Orally administered nanosystems could encounter any of the three different fates i.e. remaining bound to chyme, transiting rapidly through the GI tract and being quickly eliminated through the feces; binding to loosely adherent mucus, remaining bound until the mucus is cleared; or penetrating the mucus, remaining bound to firmly adherent mucus or entering the intestinal epithelium. In spite of the fact that mucus does not serve as an obstruction to the oral transport of many small and large molecular drugs, encapsulating these medicines within nanocarriers may increase their oral bioavailability (Lundquist and Artursson Citation2016).

4.4. P-glycoprotein efflux system

P-glycoprotein (P-gp) is an ATP-dependent drug efflux system at the apical surface of cells and works as an efflux membrane carrier (Johnstone et al. Citation2000). Lipophilic substrates that cross the lipid bilayer membrane into the lumen might get removed by P-gp and returned to the external medium before they can enter the cytoplasm. Examples of hydrophobic substrates include anticancer drugs, immuno-suppressants, steroidal drugs, calcium channel blockers, beta receptor blockers and glycosidic caridiac drugs. Small intestinal epithelial cells produce p-gp at significant levels, indicating the significance of this protein in reducing the oral bioavailability of certain drugs (Gavhane and Yadav Citation2012). Lymphocytes, the luminal epithelium of the colon, and other barrier-functioning tissues all show P-gp that must be considered before designing a carrier system for treating UC (Cortada et al. Citation2009).

4.5. Comorbid gastrointestinal conditions

In addition, the disease conditions such as constipation, diarrhea, irritable bowel disease etc. alter the colonic drug delivery system and should be considered during therapy and desing of nanosystem (Kadiyam and Muzib Citation2015). In the context of disorders as barriers, irritable bowel disorders causes thickening of the mucosa that leads to reduction in the surface area and subsequently low absorption of the lipophillic drugs. In addition, diarrhea leads to reduced drug retention time and less absorption (Kotla et al. Citation2014). Therefor, these factors should be kept in mind when selecting a nanosystem for specific patient.

4.6. Pharmaceutical factors

Weakly absorbed substances, like peptides, are more frequently absorbed in colon because the colon provides a longer retention time for them. Drug delivery to the colon can be accomplished using the same drug molecules that are used to treat GIT diseases. In the context of pharmaceutical factors, the colonic stay of drugs affects the drug absorption and thus for the low absorbed drugs colon provide more stay time and hence high absorption (Singh N and Khanna Citation2012). Therefore, such parameters should be considered for the delivery systems targeting colon. Drugs’ nature and intended use can be employed as carrier screening criteria. Other variables at play include the active functional groups in the drug, the chemistry of the medicine, stability factors, and other variables like the partition coefficient of the drug.

5. Conventional nanocarriers targeting ulcerative colitis

Among conventional nanocarriers systems targeting ulcerative colitis, the therapeutic agents or nanosystem could be efficiently protected from the acidic pH environment of the GIT by using nanogel and thus release the payloads when the carrier reaches the intestine (Bertoni et al. Citation2020). An extracellular matrix based hydrogel was fabricated and targeted to ulcerative colitis to avoid surgical procedure. Results showed a marked change in the phenotype of macrophage leading to a rapid replacement of barriers in the way of colonic mucosa. The prepared nanosystem was adhesive to the colonic tissue that resulted in an accelerated healing outcome (Keane et al. Citation2017). Similarly, another study showed the promising potential against ulcerative colitis by decreasing the expression of proinflammatory cytokines (Sun et al. Citation2021). To overcome barriers in the oral route of administration, siRNA loaded metal – organic – framework encapsulated in the sodium alginate hydrogel as a hybrid delivery system was targeted to ulcerative colitis. Results showed efficient delivery of the loaded cargo into the colon without degradation in the stomach and ensured targeted colon delivery (Gao et al. Citation2022).

Liposome is a nanoscafold that encapsulate both hydrophobic and water soluble drug candidates and provide the opportunity of surface modification as well that helps in targeting the inflamed colon (Singh AK et al. Citation2019). Solid – lipid nanoparticles provide a stable platform that offer more extended drug release and better protection due to slow disruption of the used matrix in these dosage units (Lee et al. Citation2020). Furthermore, self-micro emulsifying delivery system is used for hydrophobic drugs targeting into gastrointestinal disease conditions (Alsaad et al. Citation2020). In term of surface modification, curcumin loaded folate modified self micro emulsifying liposome were designed and targeted to ulcerative colitis that showed effective therapeutic outcome for the natural curcumin from the nanocarrier system (Zhang et al. Citation2012). Taking benefit of the anti-inflammatory properties of Oxymatrine – a Chinese medicinal herb, it was loaded into liposome for targeted delivery to ulcerative colitis. Due to the anti-inflammatory potential of Nitric oxide, it was also loaded to the same liposomal system and biodistribution studies investigated higher accumulation and retention time for the loaded drug entities. Results from ulcerative colitis mice model showed significant down – regulation of proinflammatory cytokines and decrease in macrophages’ infiltration, suggesting the anti – colitis potential of oxymatrine and nitric oxide loaded liposomes (Tang et al. Citation2020).

The role of polymeric nanoparticles for drug delivery is widely explored in recent years; however it bears certain serious hurdles such as synthesis scale up and toxicity. Such issues are somehow resolved by using polymers from natural sources (Yang Mei et al. 2020). For instance, nanoparticles from ginger and Lyceum barbarum have shown excellent anti-inflammatory effects by delivering siRNA (Zhang et al. Citation2017; Zu et al. Citation2020). Moreover, mesalamine loaded apple polysaccharide silver nanoparticles were developed for the effective treatment of ulcerative colitis. Results from acetic acid induced ulcerative colitis rat model showed effective therapeutic outcomes at higher doses of the drug loaded nanocarrier system as compared to low doses showing the efficacy of the afore mentioned nanosystem (Kaur et al. Citation2020). Similarly, Phragmites rhizoma polysaccharide selenium nanoparticles were developed and loaded with Azathioprine for targeting ulcerative colitis lesions. In-vivo results showed significant down regulation of the inflammatory cytokines at colonic and serum level that resulted in the clinical symptoms alleviations of the ulcerative colitis as well as healing of the lesions (Cui et al. Citation2022).

Still, these traditional nanocarriers are incapable of transporting and releasing medicines at the proper concentration at the desired site in response to internal or external stimulation. Consequently, conventional nanocarriers are not intelligent and they must be modified or functionalized to become intelligent and smart. The intelligent nanosystems should avoid the immune system’s clearance, accumulate in designated locations and release the loaded cargo at the desired concentration at the target site in response to internal or external stimulation. Moreover, they should be able to co-deliver drugs and other substances, like imaging agents, genetic components etc. Depending on the types and uses of nanocarriers, there are measures to convert conventional nanocarriers into intelligent ones (Hossen et al. Citation2019). Various features of the conventional and smart nanosystems are compared and contrasted in .

Table 2. Salient features of the conventional and smart nanosystems.

6. Smart nanosystem targeting ulcerative colitis

Strategies to convert traditional nanocarriers to smart ones rely on their intended use. The nanocarriers must pass through various physiological barriers, including reticulo-endothelial system (RES) of the liver and spleen. PEGylation- first reported by Davies and Abuchowsky (Abuchowski et al. Citation1977), is one way to avoid such clearance of the nanocarriers. However, PEGylation also decreases drug uptake by cells (Moghimi et al. Citation2001; Moghimi and Szebeni Citation2003). Additionally, nanocarriers can be decorated with certain ligands that could specifically distinguish diseased cells from healthy ones. The physiochemical variations of the diseased tissues and overexpression of certain components can be exploited for such purposes. Smart nanocarriers target such overexpressed receptors or proteins and ligands attach to their receptors making the system able to deliver the loaded drug to desired sites. Stimulating the nanocarrier system to release the drug in the desired target site is also a problem. Grafting chemical bonds or functional groups to nanocarriers makes them sensitive to internal and external stimuli. For synergistic and theranostic effects, co-delivery of drugs and other agents can also be exploited using smart delivery systems (Yan et al. Citation2022).

In nanocarriers based drug delivery systems, smart polymers based nanosystems have shown extensive applications particularly in colonic drug delivery (Singh D et al. Citation2015). Among these smart polymers, two types are widely explored including cationic and anionic pH sensitive smart polymers. Example of anionic pH sensitive smart polymer includes; poly(N,N-dimethylaminoethyl methacrylamide and the later one includes; chitosan, poly(lysine) and poly ethylenemie (Zhou and Chen Citation2015). Many studies have shown the use of smart nanosystem for the colon specific drug delivery. The use of such smart nanosystems have shown efficacy in various GIT disorders including ulcerative colitis (Salvi and Pawar Citation2019). It is one of the versatile advantages of the drug delivery from smart nanosystem that it offers reduced particle size, which targets a specified location and thus boosts bioavaibility. Reduction in the particle size also minimizes the delivery dose for the drugs and subsequently leads to reduction in the systemic side effects (Cherukuri et al. Citation2012). In addition, the conventional mood of drug delivery lacks the potential to absolutely deliver the desired drug concentration to the target site with accuracy; however, smart nanosystems ensure the target specific delivery of the payloads with minimal toxicity and better therapeutic outcomes (E Leucuta Citation2012). The delivery of drugs from different nano-platforms for treating UC is diagrammatically illustrated in .

Figure 3. Illustration of smart nanosystems and their potential benefits for colon specific drug delivery.

Figure 3. Illustration of smart nanosystems and their potential benefits for colon specific drug delivery.

6.1. Microbially triggered smart nanosystems for UC

Microorganisms known as the gut microbiota live in harmony with the host in the gut and secrete a number of enzymes that are involved in the metabolism of carbohydrates and reductive compounds (Peng et al. Citation2021). In the colon – stimuli – responsive systems, microbially triggered system are those that release the loaded contents when the system is degraded by the microflora causing release of the drug in to the intestinal environment and thus deliver at specific site (Dev et al. Citation2011). Of note, the smart polymers are not degraded at the gastric environment and early intestinal levels since less quantity of the flora is present at these areas. In this regard, metronidazole loaded azo aromatase based capsule were targeted to colon for treating colitis and the desired effect was achieved through degradation of polymer by floral enzymes at colon level (Hita et al. Citation1997). The enzymes from microbial flora involved in such mechanisms include; nitroreductase, arabinosidase, galactosidase, dehydroxylase, xylosidase (Sharif et al. Citation2017). In another study, dextran sulfate sodium induced colitis mice was evaluated by administration of a tripeptide loaded hydrogel. Results showed a lowest delivery concentration for the tripeptide as compared to free solution, however provided similar therapeutic efficacy with minimal side effects (Soni et al. Citation2015). A guar gum based 5 fluorouracil loaded system was delivered to the colon and resulted in an efficient release of the drug after exposure to the microbial flora indicating the enzyme responsive potential and subsequent use of the guar gum for UC targeting (Kumar et al. Citation2017).

Similarly, in vitro results from chitosan modified lipid nanoparticles showed esterase responsive property along with significant release of the drug that showed colon targeting effects. The invivo distribution results showed high retention of the drug in colitis bearing mice model as compared to free dexamethasone. This enzyme responsive smart nano system showed prominent anti ulcerative colitis effect (Chen S-q et al. Citation2020). To specifically deliver the payloads, curcumin loaded nanomicelles were fabricated in order to evaluate the enhanced mucoadhesion of the fabricated system and the azoreductase based drug release. The prepared nanosystem significantly released the drug based on azoreductase sensitive particles dissociation. The designed smart nano system based complex also mitigated ulcerative colitis symptoms through acceleration of colitis repair (Zhang, Li, et al. Citation2022b). In the field of oral gene therapy, nucleic acid drugs exhibit anti inflammatory potential by healing ulcers and thus manifest suitable opportunity for the ulcerative colitis. In a study, miR − 320 loaded alginate based polymeric nanocapsules were prepared and targeted to colon. Results from invivo imaging showed that the designed nanosystem reached the colon after 2 hours and released miR − 320 at colonic mucosa after degradation of alginates by enzymes (Li et al. Citation2022). To overcome the anatomical and physiological barrier in the colonic drug delivery, Li et, al. designed curcumin – cyclodextrin complex loaded chitosan and alginate based nanoparticles and targeted to ulcerative colitis bearing mice. Invitro studies’ results showed that the drug release exhibited enzyme responsive characteristics i.e. α – amylase responsive release while invivo results showed reshaping of the microbial flora, rapid uptake of macrophages, strong biodistribution at colonic level and therapeutic efficacy that all resulted in healing of ulcerative colitis (Li S et al. Citation2021).

6.2. Osmotically controlled smart nanosystems for UC

Osmotic controlled drug delivery system is another scaffold that targets colonic disorders including ulcerative colitis with efficient systemic absorption and consist of five to six push – pull unit within a single osmotic unit enclosed in a hard gelatin capsule (Arévalo-Pérez et al. Citation2020). The system consist of a drug layer and an osmotic push layer that collectively form a bilayer that is covered by a semi permeable membrane consist of an orifice in drug layer (Akbarzadeh et al. Citation2019). The osmotic push compartment is swelled after the absorption of water into the system and form a gel. The drug gel mass is expelled outside through orifice (Ogueri and Shamblin Citation2022). In the context of ulcerative colitis therapy, the delivery system is designed in such a way that a few hours of post gastric delay the drug delivery is avoided in the small intestine (Aslan et al. Citation2013). Furthermore, when the delivery system reaches the colon, a constant drug release rate is achieved after delivering the drug cargo for almost 24 hours within the colitis site (Awad et al. Citation2022). Calcium phosphate (CaP), a naturally pH-sensitive substance, was used to construct siRNA-loaded CaP/PLGA nanoparticles (Frede et al. Citation2016), which had a high affinity for nucleic acids but dissolved in the low pH environment inside the lysosome after endocytosis. The release of nucleic acids into the cytosol was aided by an increase in osmotic pressure. CaP/PLGA NPs loaded with TNF-, keratinocyte chemoattractant, or IP-10 siRNA were efficiently taken up by gut epithelial cells, resulting in a significant decrease in target gene expression in colonic biopsies and mesenteric lymph nodes of colonic inflammation mice model. The system resulted in significant relief of the colitis symptoms in mice model.

6.3. pH responsive smart nanosystems for UC

The pH of empty stomach gets rise after the ingestion of meal and rises in the descending parts of the GIT eventually reaches to a pH 7 at the colon level (Vinarov et al. Citation2021). Smart nanosystems sense these changes in the pH and accordingly release the loaded cargo at the colon pH. Because of their susceptibility to digestion while passing through the GI system, liposomes are not preferred for oral drug delivery. Coating liposomes with polymers is one way to protect them during transit, but there has been very little research into specifically targeting the colonic area (Barea et al. Citation2012). Methacrylic acid (Eudragit L − 100) and ethyl acrylate (Eudragit S − 100) are the two pH responsive polymers used in colonic drug delivery (Hua Citation2014). During the treatment of ulcerative colitis, such polymers get protonated or deprotonated depending on pH changes and thus behave accordingly to swell or contract resulting in the retention or release of the loaded contents (Nagpal et al. Citation2010). In a study, liposome-in-microsphere (LIM) containing drug-loaded liposomes within pH responsive Eudragit S100 microspheres were developed. The liposomes were loaded with model drug 5-ASA and coated with chitosan to protect them from clumping in subsequent encapsulation in microspheres and to enhance tailored release features. LIMs inhibited drug release in simulated stomach and small intestine conditions, with subsequent drug release happening in large intestine conditions, according to in vitro drug release studies. As a result, the nanosystem showed the ability for oral colonic drug administration (Barea et al. Citation2012). In another research study, drug release from liposomes based on Eudragit S100 was lower in solution that simulated the stomach pH conditions i.e. 1.4 and 6.3 of small intestine thus showed pH responsive drug release (Barea et al. Citation2010).

Polymers used in the pH responsive nanoparticles includes anionic polymers and acrylate that at high pH gets swell and at low pH pose insolubility property (Liu L et al. Citation2017). Such pH sensitivity behavior enables them to deliver the loaded cargoes at higher pH of the colon and protect the payloads from degradation by harsh acidic environment (Gugulothu et al. Citation2014). In this regard, a PLGA and Eudragit S − 100 co – polymer was used as a pH sensitive system loaded with budesonide and was fabricated into nanoparticles. The resultant system showed pH dependent release and at a pH of 7.4, sustained release kinetics was shown by the prepared system (Makhlof et al. Citation2009). In another study, prednisolone was loaded into mesoporous silica nano particles based on ε – polylysine. Invitro results from the study showed pH responsive drug release from the smart nanosystem such as; no drug release at pH 5 of small intestine, 1.9 of stomach and high drug release at pH 7 of colon (Nguyen et al. Citation2017).

To explore the core shell nano particles in ulcerative colitis, curcumin nanocrystals were prepared and loaded to chitosan – alginate based nanoparticles that were targeted to ulcerative colitis. Biodistribution and drug release studies showed that the resultant nanoparticles showed higher distribution at colon as compared to stomach and small intestine, wherease the drug release was high at higher pH and lower at reduced pH thus presenting it a suitable ulcerative colitis therapy (Oshi et al. Citation2020). Similarly, 5 amino salicylic acid core shell nanoparticulate system was fabricated and targeted to ulcerative colitis. Results from invitro studies showed enhanced stability of the fabricated core shell system, avoided the drug release at upper gastro intestinal tract and sustained drug release at colon (Wang N et al. Citation2022). Cyclosporine A is an immunosuppressant agent that has limited use in ulcerative colitis due to systemic side effects. To overcome this issue and increase the therapeutic efficacy, cyclosporine A loaded alginate micro particles coated with Eudragit S100 was prepared and targeted to colon for the treatment of ulcerative colitis (). The resultant complex smart polymer based delivery system upon exposure to simulated gastric condition showed no release, no absorption and thus avoided side effects while upon exposure to simulated colon condition showed disintegration of the microparticles with sustained release behaviors for 24 hours (Oshi et al. Citation2021). In a recent study, sulfasalazine loaded pectin – PEG – methacrylic acid grafted hydrogel was prepared and evaluated in ulcerative colitis model. Free radical polymerization was used for the grafting of smart polymers. Invitro studies confirmed the degradation of used smart polymer by microbial flora and the drug release was maximal at pH 7.4 of the colon (Abbasi et al. Citation2019). Over all, despite of the marked results observed from pH responsive delivery systems targeting ulcerative colitis, the intra – individual as well as inter – individual differences in pH is a major concern here and the luminal pH changes due to disease conditions should also be considered.

Table 3. Examples of major studies conducted for treating UC with smart nanosystems.

6.3.1. Reactive oxygen species (ROS) and redox- responsive smart nanosystems for UC

In the treatment of ulcerative colitis, reactive oxygen species responsive delivery systems have shown promising potentials (Talaei et al. Citation2013). The imbalance in the production of reactive oxygen species and decrease response of the immune system is referred as oxidative stress that is mainly observed with inflammatory conditions and an over production of reactive oxygen species is observed in ulcerative colitis (Birben et al. Citation2012; Piechota-Polanczyk and Fichna Citation2014). The redox – responsive nanosystem takes the advantage of the pathological condition of ulcerative colitis and thus exhibit a profound potential for the therapy of ulcerative colitis. In this regard, TNF – α siRNA loaded thioketal nanoparticles were fabricated for oral administration and targeting of ulcerative colitis. The aim of using thioketal was as a carrier and degradation in response to the reactive oxygen species. Results from murine ulcerative colitis model showed the degradation of the polymer at ulcerative colitis site due to high contents of the reactive oxygen species and efficient delivery of siRNA to protect the mice from ulcerative colitis (Wilson et al. Citation2010). Similarly, nanoparticles loaded with nitroxide radicals having an amphiphilic block co – polymer was targeted to ulcerative colitis bearing mice and results showed high accumulation of the nitroxide radicals at inflammation site. Mechanistically, the used radicals significantly scavenged reactive oxygen species concluding that nitroxide radicals could be effective therapeutic alternative for ulcerative colitis (Vong et al. Citation2012). In relationship between reactive oxygen species and ulcerative colitis, luteolin being anti – Inflammatory and ROS scavenging natural flavonoid, was loaded in tocopherol polyethylene glycol succinate – b – poly(β – thioester) copolymer based nanoparticles and targeted to inflamed colon of murine model. Results showed healing of the inflammatory mucosa and resolving inflammation through the suppression of reactive oxygen species and upregulation of anti – inflammatory factors (Tan et al. Citation2022).

Budesonide encapsulated nanoparticles based on self – assembled polyether micelles were designed to effectively target ulcerative colitis. The purpose of using smart polymer was its oxidation potential at ulcerative colitis site due to increased ROS concentration. Results of this study showed that the fabricated delivery system significantly repaired the intestinal barriers and the colonic damaged tissues were healed effectively (Zhao et al. Citation2022). Similarly, Haiting et, al. fabricated pH and ROS dual responsive curcumin loaded chitosan – alginate based hydrogel for targeting ulcerative colitis. Their results showed significant suppression of the high reactive oxygen species level at colon level and also protected the degradation of complex delivery system from the harsh gastric acidic environment. The mechanism behind the anti – inflammatory effect of the drug loaded smart drug delivery system in animal model was targeting TLR4-MAPK/NF-κB signaling pathway (Xu H et al. Citation2022). In the context of dual smart delivery system i.e. both pH and ROS responsive delivery system another study investigated acid resistant – ROS responsive hyper branched self assembling polythioether micelle targeting ulcerative colitis. Findings of the study showed that the thioester bonds were significantly oxidized into sulfone groups that in response to accumulated high reactive oxygen species enhanced the hydrophilicity and ultimately mimicked the encapsulated drug release. Furthermore, the self assembling micellar structure helped in bypassing the acidic gastric environment without leaking the drug candidate (Shi et al. Citation2020). Prominent research studies conducted for treating UC via the application of smart nanosystems are summarized in .

6.3.2. Thermo- responsive smart nanosystems for UC

The thermo- responsive drug delivery platform has been frequently applied for liposomes, causing drug release at 42 °C and resulting in maximum drug concentration at the desired site. Certain lipids and polymers have abrupt and discontinuous shifts in physical properties with temperature fluctuations (Lee and Thompson Citation2017). This drug delivery method is also applicable to hydrogels. In this regard mesalazine was delivered to the ulcerative colitis by loading it into the pectin cross linked chitosan based thermo – responsive hydrogel. Results of this study showed sustained release kinetics for the loaded drug due to cross linkage between chitosan and pectin. The release pattern of drug at targeted site and drug loading to the smart nanocarrier was in desirable range (Ding et al. Citation2017). In another research study thermo responsive smart carrier was used for targeting ulcerative colitis and to delivery mesalamine or budesonide. The nanocarrier system was administered through rectal route to the mice that was initially in liquid form and upon exposure to body temperature it was converted into a viscous gel (Sinha et al. Citation2015). The hypothetical mechanism is depicted in . All these findings suggest that the stimuli responsive smart drug delivery system is an efficient way for targeting the ulcerative colitis and it give an opportunity for the therapeutic candidates to be used for other gastro intestinal disorders using such stimulus responsive delivery system.

Figure 4. Ulcerative colitis targeted by thermo responsive nanocarrier system via rectal route.

Figure 4. Ulcerative colitis targeted by thermo responsive nanocarrier system via rectal route.

7. Conclusion and future perspectives

The current therapeutic agents used for the treatment of ulcerative colitis are suffering from chronic systemic exposure and immunosuppression. In such circumstances, nanotechnology platform offers an effective and safe approach targeting ulcerative colitis. Up till now, various conventional nanocarriers have been investigated however; the use of smart nanocarrier system has opened new avenues in the field of bowel diseases through providing more retention time at inflamed site, responsiveness toward various physiological conditions and reduction in frequency of administration. Furthermore, such smart nanocarrier system has resulted in the efficient accumulation of therapeutic candidates at inflamed site and protection of healthy tissues from concerned side effects. In addition, responsive smart nanocarrier delivery system results in delivery of loaded cargoes at specified site of the colon at desired pH, high contents of the reactive oxygen species and presence of specific enzymes. All these factors enables the drug delivery system to release the payloads at target site without disrupted by the harsh gastric environment of the stomach.

Although such responsive smart nano delivery system could improved patient’s quality of life by boosting the therapeutic efficacy, but so far no conclusion and factual data regarding the level of nano carrier adverse effects have been brought up that needs nanotoxicology studies in the human gastrointestinal tract and animal models. The responsive nanocarrier system have been explored for surface interactions, multiple materials and different sizes however; with enhanced understanding of the human gastrointestinal tract, the applications of such responsive nano system needs further expansion. Moreover, ulcerative colitis is an inflammatory condition and in such cases the immune cells and functions dramatically alters, so macrophages being major immune system regulators are polarized to pro – inflammatory state leading to dysfunctioning of the system. Thus, using responsive smart nanocarriers to switch the phenotypes of the immune cells need further investigations in ulcerative colitis. Among stimuli responsive nanocarrier system, excluding pH responsive system, there are certain obstacles in the way of other responsive systems that include the enzyme rich and harsh acidic environment of the upper gastro intestinal tract where the drug undergoes instability issues, so it should be focused in the future for successful ulcerative colitis therapy. Also the premature release of drugs leads to less delivery of therapeutic agents to the target colon site and also causes side effects needs future investigations. The use of dual-responsive nanosystem have shown effective outcomes in the treatment of ulcerative colitis however it needs clinical translation to ensure and confirm its status of pre – metabolism and long term safety within the systemic circulation. In summary, to mitigate the drugs concern problems in the future, work should be carried out to design multifunctional delivery systems that respond to various stimuli at a time and overcome the multiple barriers in a single unit so then it could be easily transformed from bench top to the clinical settings for the effective therapy of ulcerative colitis.

Authors’ contributions

Min Chen: writing initial draft and revision of the manuscript; Huanrong Lan: initial draft and revised the manuscript; Ketao Jin: Conception and design of the study; Yun Chen: revision and finalization of the manuscript.

Disclosure statement

The authors report there are no competing interests to declare.

Additional information

Funding

This work was supported by Zhejiang Provincial Science and Technology Projects (grants no. LGF22H160017 to MC, LGD19H160001 to KTJ, and LGF22H160046 to HRL), National Natural Science Foundation of China (grant no. 82104445 to HRL), and Jinhua Municipal Science and Technology Projects (grants no.2021-3-040 to KTJ, and 2021-3-046 to HRL).

References

  • Abbasi M , Sohail M , Minhas MU , et al. ( 2019). Novel biodegradable pH-sensitive hydrogels: An efficient controlled release system to manage ulcerative colitis. Int J Biol Macromol 136: 1–14.
  • Abuchowski A , McCoy JR , Palczuk NC , et al. ( 1977). Effect of covalent attachment of polyethylene glycol on immunogenicity and circulating life of bovine liver catalase. J Biol Chem 252: 3582–6.
  • Aib S , Iqbal K , Khan N , et al. ( 2022). pH-sensitive liposomes for colonic co-delivery of mesalazine and curcumin for the treatment of ulcerative colitis. J Drug Delivery Sci Technol 72: 103335.
  • Akbarzadeh A , Samiei M , Joo SW , et al. ( 2019). Synthesis, characterization and in vitro studies of doxorubicin-loaded magnetic nanoparticles grafted to smart copolymers on A549 lung cancer cell line (Retraction of Vol 10, art no 46. BMC CAMPUS, 4 CRINAN ST, LONDON N1 9XW, ENGLAND.
  • Alsaad A , Hussien AA , Gareeb MM. ( 2020). Solid lipid nanoparticles (SLN) as a novel drug delivery system: A theoretical review. Syst Rev Pharm 11: 259–73.
  • Arévalo-Pérez R , Maderuelo C , Lanao JM. ( 2020). Recent advances in colon drug delivery systems. J Control Release 327: 703–24.
  • Aslan B , Ozpolat B , Sood AK , Lopez-Berestein G. ( 2013). Nanotechnology in cancer therapy. J Drug Target 21: 904–13.
  • Awad A , Madla CM , McCoubrey LE , et al. ( 2022). Clinical translation of advanced colonic drug delivery technologies. Adv Drug Delivery Rev 181: 114076.
  • Barea M , Jenkins M , Gaber M , Bridson R. ( 2010). Evaluation of liposomes coated with a pH responsive polymer. Int J Pharm 402: 89–94.
  • Barea M , Jenkins M , Lee Y , et al. ( 2012). Encapsulation of liposomes within pH responsive microspheres for oral colonic drug delivery. Int J Biomater 2012: 1–8.
  • Beloqui A , Coco R , Memvanga PB , et al. ( 2014). pH-sensitive nanoparticles for colonic delivery of curcumin in inflammatory bowel disease. Int J Pharm 473: 203–12.
  • Bertoni S , Machness A , Tiboni M , et al. ( 2020). Reactive oxygen species responsive nanoplatforms as smart drug delivery systems for gastrointestinal tract targeting. Biopolymers 111: e23336.
  • Birben E , Sahiner UM , Sackesen C , et al. ( 2012). Oxidative stress and antioxidant defense. World Allergy Organ J 5: 9–19.
  • Cantero-Recasens G , Burballa C , Ohkawa Y , et al. ( 2022). The ulcerative colitis-associated gene FUT8 regulates the quantity and quality of secreted mucins. Proc Natl Acad Sci U S A 119: e2205277119.
  • Caster JM , Callaghan C , Seyedin SN , et al. ( 2019). Optimizing advances in nanoparticle delivery for cancer immunotherapy. Adv Drug Deliv Rev 144: 3–15.
  • Chen L , Hong W , Ren W , et al. ( 2021). Recent progress in targeted delivery vectors based on biomimetic nanoparticles. Sig Transduct Target Ther 6: 1–25.
  • Chen R , Lin X , Wang Q , et al. ( 2023). Dual-targeting celecoxib nanoparticles protect intestinal epithelium and regulate macrophage polarization for ulcerative colitis treatment. Chemical Engineering Journal 452: 139445.
  • Chen S-q , Song Y-q , Wang C , et al. ( 2020). Chitosan-modified lipid nanodrug delivery system for the targeted and responsive treatment of ulcerative colitis. Carbohydr Polym 230: 115613.
  • Chen Z , Farag MA , Zhong Z , et al. ( 2021). Multifaceted role of phyto-derived polyphenols in nanodrug delivery systems. Adv Drug Deliv Rev 176: 113870.
  • Cherukuri S , Neelabonia V , Reddipalli S , Komaragiri K. ( 2012). A review on pharamceutical approaches on current trends of colon specific drug delivery system. Int Res J Pharm 3: 45–55.
  • Claesson-Welsh L , Dejana E , McDonald DM. ( 2021). Permeability of the endothelial barrier: identifying and reconciling controversies. Trends Mol Med 27: 314–31.
  • Cortada CM , Gil A , Goncalves S , et al. ( 2009). P-glycoprotein functional activity in peripheral blood lymphocytes in ulcerative colitis. Medicina 69: 437–41.
  • Cui M , Fang Z , Song M , et al. ( 2022). Phragmites rhizoma polysaccharide-based nanocarriers for synergistic treatment of ulcerative colitis. Int J Biol Macromol 220: 22–32.
  • D’Amico F , Magro F , Peyrin-Biroulet L , Danese S. ( 2022). Positioning filgotinib in the treatment algorithm of moderate to severe ulcerative colitis. J Crohns Colitis 16: 835–44.
  • Danese S , Grisham M , Hodge J , Telliez J-B. ( 2016). JAK inhibition using tofacitinib for inflammatory bowel disease treatment: a hub for multiple inflammatory cytokines. Am J Physiol Gastrointest Liver Physiol 310: G155–G162.
  • Deng X-Q , Zhang H-B , Wang G-F , et al. ( 2019). Colon-specific microspheres loaded with puerarin reduce tumorigenesis and metastasis in colitis-associated colorectal cancer. Int J Pharm 570: 118644.
  • Dev RK , Bali V , Pathak K. ( 2011). Novel microbially triggered colon specific delivery system of 5-Fluorouracil: Statistical optimization, in vitro, in vivo, cytotoxic and stability assessment. Int J Pharm 411: 142–51.
  • Ding C , Sun Y , Wang Y , et al. ( 2017). Adsorbent for resorcinol removal based on cellulose functionalized with magnetic poly (dopamine). Int J Biol Macromol 99: 578–85.
  • Donaldson GP , Lee SM , Mazmanian SK. ( 2016). Gut biogeography of the bacterial microbiota. Nat Rev Microbiol 14: 20–32.
  • E, Leucuta ( 2012). Drug delivery systems with modified release for systemic and biophase bioavailability. Current Clinical Pharmacology 7: 282–317.
  • Ordás I , Eckmann L , Talamini M , et al. ( 2012). Ulcerative colitis. Lancet 380: 1606–19.
  • Fändriks L. ( 2017). Roles of the gut in the metabolic syndrome: an overview. J Intern Med 281: 319–36.
  • Feagan BG , MacDonald JK. ( 2012). Oral 5-aminosalicylic acid for induction of remission in ulcerative colitis. Cochrane database of systematic reviews.(10).
  • Feagan BG , Rutgeerts P , Sands BE , et al. ( 2013). Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med 369: 699–710.
  • Frede A , Neuhaus B , Klopfleisch R , et al. ( 2016). Colonic gene silencing using siRNA-loaded calcium phosphate/PLGA nanoparticles ameliorates intestinal inflammation in vivo. J Control Release 222: 86–96.
  • Gao M , Yang C , Wu C , et al. ( 2022). Hydrogel–metal-organic-framework hybrids mediated efficient oral delivery of siRNA for the treatment of ulcerative colitis. J Nanobiotechnol 20: 1–12.
  • Gaohua L , Miao X , Dou L. ( 2021). Crosstalk of physiological pH and chemical pKa under the umbrella of physiologically based pharmacokinetic modeling of drug absorption, distribution, metabolism, excretion, and toxicity. Expert Opin Drug Metab Toxicol 17: 1103–24.
  • Garbern JC , Minami E , Stayton PS , Murry CE. ( 2011). Delivery of basic fibroblast growth factor with a pH-responsive, injectable hydrogel to improve angiogenesis in infarcted myocardium. Biomaterials 32: 2407–16.
  • Gauri B , Singh SK , Mishra D. ( 2011). Formulation and evaluation of colon targeted oral drug delivery systems for metronidazole in treatment of amoebiasis. International Journal of Drug Delivery 3: 503.
  • Gavhane YN , Yadav AV. ( 2012). Loss of orally administered drugs in GI tract. Saudi Pharm J 20: 331–44.
  • Grondin JA , Kwon YH , Far PM , et al. ( 2020). Mucins in intestinal mucosal defense and inflammation: learning from clinical and experimental studies. Front Immunol 11: 2054.
  • Gugulothu D , Kulkarni A , Patravale V , Dandekar P. ( 2014). pH-sensitive nanoparticles of curcumin–celecoxib combination: evaluating drug synergy in ulcerative colitis model. J Pharm Sci 103: 687–96.
  • Günter EA , Markov PA , Melekhin AK , et al. ( 2018). Preparation and release characteristics of mesalazine loaded calcium pectin-silica gel beads based on callus cultures pectins for colon-targeted drug delivery. Int J Biol Macromol 120: 2225–33.
  • Heller F , Fromm A , Gitter A , et al. ( 2008). Epithelial apoptosis is a prominent feature of the epithelial barrier disturbance in intestinal inflammation: effect of pro-inflammatory interleukin-13 on epithelial cell function. Mucosal Immunology 1: S58–S61.
  • Helmy AM , Elsabahy M , Abd-Elkareem M , et al. ( 2020). High-Payload chitosan microparticles for the colonic delivery of quercetin: Development and in-vivo evaluation in a rabbit colitis model. J Drug Delivery Sci Technol 58: 101832.
  • Hita V , Singh R , Jain SK. ( 1997). Colonic targeting of metronidazole using azo aromatic polymers: development and characterization. Drug Delivery 4: 19–22.
  • Hlaing SP , Cao J , Lee J , et al. ( 2022). Hyaluronic Acid-Conjugated PLGA Nanoparticles Alleviate Ulcerative Colitis via CD44-Mediated Dual Targeting to Inflamed Colitis Tissue and Macrophages. Pharmaceutics 14: 2118.
  • Hossen S , Hossain MK , Basher M , et al. ( 2019). Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity studies: A review. J Adv Res 15: 1–18.
  • mHua S , s. ( 2014). Orally administered liposomal formulationfor colon targeted drug delivery. Frontiers Media SA. 138.
  • Johnstone RW , Ruefli AA , Smyth MJ. ( 2000). Multiple physiological functions for multidrug transporter P-glycoprotein? Trends Biochem Sci 25: 1–6.
  • Kadiyam R , Muzib YI. ( 2015). Colon specific drug delivery of tramadol HCl for chronotherapeutics of arthritis. Int J Pharm Investig 5: 43–9.
  • Kaur G , Singh SK , Kumar R , et al. ( 2020). Development of modified apple polysaccharide capped silver nanoparticles loaded with mesalamine for effective treatment of ulcerative colitis. J Drug Delivery Sci Technol 60: 101980.
  • Kayal M , Shah S. ( 2019). Ulcerative colitis: current and emerging treatment strategies. JCM 9: 94.
  • Keane TJ , Dziki J , Sobieski E , et al. ( 2017). Restoring mucosal barrier function and modifying macrophage phenotype with an extracellular matrix hydrogel: potential therapy for ulcerative colitis. Journal of Crohn’s and Colitis 11: 360–8.
  • Ko CW , Singh S , Feuerstein JD , et al. ( 2019). AGA clinical practice guidelines on the management of mild-to-moderate ulcerative colitis. Gastroenterology 156: 748–64.
  • Kotla NG , Gulati M , Singh SK , Shivapooja A. ( 2014). Facts, fallacies and future of dissolution testing of polysaccharide based colon-specific drug delivery. J Control Release 178: 55–62.
  • Kucharzik T , Koletzko S , Kannengiesser K , Dignass A. ( 2020). Ulcerative colitis—diagnostic and therapeutic algorithms. Deutsches Ärzteblatt International 117: 564.
  • Kumar B , Kulanthaivel S , Mondal A , et al. ( 2017). Mesoporous silica nanoparticle based enzyme responsive system for colon specific drug delivery through guar gum capping. Colloids Surf B Biointerfaces 150: 352–61.
  • Lamprecht A , Yamamoto H , Takeuchi H , Kawashima Y. ( 2004). Design of pH-sensitive microspheres for the colonic delivery of the immunosuppressive drug tacrolimus. Eur J Pharm Biopharm 58: 37–43.
  • Lee C-M , Kim D-W , Lee H-C , Lee K-Y. ( 2004). Pectin microspheres for oral colon delivery: Preparation using spray drying method and in vitro release of indomethacin. Biotechnol Bioprocess Eng 9: 191–5.
  • Lee MS , Kim Y-J. ( 2007). Signaling pathways downstream of pattern-recognition receptors and their cross talk. Annu Rev Biochem 76: 447–80.
  • Lee Y , Sugihara K , Gillilland MG , et al. ( 2020). Hyaluronic acid–bilirubin nanomedicine for targeted modulation of dysregulated intestinal barrier, microbiome and immune responses in colitis. Nat Mater 19: 118–26.
  • Lee Y , Thompson D. ( 2017). Stimuli-responsive liposomes for drug delivery. Wiley Interdiscip Rev Nanomed Nanobiotechnol 9: e1450.
  • Li S , Jin M , Wu Y , et al. ( 2021). An efficient enzyme-triggered controlled release system for colon-targeted oral delivery to combat dextran sodium sulfate (DSS)-induced colitis in mice. Drug Deliv 28: 1120–31.
  • Li X , Yang Y , Wang Z , et al. ( 2022). Multistage-responsive nanocomplexes attenuate ulcerative colitis by improving the accumulation and distribution of oral nucleic acid drugs in the colon. ACS Appl Mater Interfaces 14: 2058–70.
  • Lichtiger S , Present DH , Kornbluth A , et al. ( 1994). Cyclosporine in severe ulcerative colitis refractory to steroid therapy. N Engl J Med 330: 1841–5.
  • Ling K , Wu H , Neish AS , Champion JA. ( 2019). Alginate/chitosan microparticles for gastric passage and intestinal release of therapeutic protein nanoparticles. J Control Release 295: 174–86.
  • Liu L , Yao W , Rao Y , et al. ( 2017). pH-Responsive carriers for oral drug delivery: challenges and opportunities of current platforms. Drug Deliv 24: 569–81.
  • Liu P , Gao C , Chen H , et al. ( 2021). Receptor-mediated targeted drug delivery systems for treatment of inflammatory bowel disease: opportunities and emerging strategies. Acta Pharm Sin B 11: 2798–818.
  • Lundquist P , Artursson P. ( 2016). Oral absorption of peptides and nanoparticles across the human intestine: Opportunities, limitations and studies in human tissues. Adv Drug Deliv Rev 106: 256–76.
  • Lynch R , Lowe D , Protheroe A , et al. ( 2013). Outcomes of rescue therapy in acute severe ulcerative colitis: data from the United Kingdom inflammatory bowel disease audit. Aliment Pharmacol Ther 38: 935–45.
  • Makhlof A , Tozuka Y , Takeuchi H. ( 2009). pH-Sensitive nanospheres for colon-specific drug delivery in experimentally induced colitis rat model. Eur J Pharm Biopharm 72: 1–8.
  • Mandlik DS , Mandlik SK , Patel S. ( 2021). Protective effect of sarsasapogenin in TNBS induced ulcerative colitis in rats associated with downregulation of pro-inflammatory mediators and oxidative stress. Immunopharmacol Immunotoxicol 43: 571–83.
  • Moghimi SM , Hunter AC , Murray JC. ( 2001). Long-circulating and target-specific nanoparticles: theory to practice. Pharmacol Rev 53: 283–318.
  • Moghimi SM , Szebeni J. ( 2003). Stealth liposomes and long circulating nanoparticles: critical issues in pharmacokinetics, opsonization and protein-binding properties. Prog Lipid Res 42: 463–78.
  • Mohan LJ , Daly JS , Ryan BM , Ramtoola Z. ( 2019). The future of nanomedicine in optimising the treatment of inflammatory bowel disease. Scand J Gastroenterol 54: 18–26.
  • Naeem M , Bae J , Oshi MA , et al. ( 2018). Colon-targeted delivery of cyclosporine A using dual-functional Eudragit® FS30D/PLGA nanoparticles ameliorates murine experimental colitis. Int J Nanomedicine 13: 1225–40.
  • Naeem M , Lee J , Oshi MA , et al. ( 2020). Colitis-targeted hybrid nanoparticles-in-microparticles system for the treatment of ulcerative colitis. Acta Biomater 116: 368–82.
  • Nagpal K , Singh SK , Mishra DN. ( 2010). Chitosan nanoparticles: a promising system in novel drug delivery. Chem Pharm Bull (Tokyo) 58: 1423–30.
  • Naserifar M , Hosseinzadeh H , Abnous K , et al. ( 2020). Oral delivery of folate-targeted resveratrol-loaded nanoparticles for inflammatory bowel disease therapy in rats. Life Sci 262: 118555.
  • Neurath MF. ( 2017). Current and emerging therapeutic targets for IBD. Nat Rev Gastroenterol Hepatol 14: 269–78.
  • Nguyen CT , Webb RI , Lambert LK , et al. ( 2017). Bifunctional succinylated ε-polylysine-coated mesoporous silica nanoparticles for pH-responsive and intracellular drug delivery targeting the colon. ACS Appl Mater Interfaces 9: 9470–83.
  • Ni J , Wu GD , Albenberg L , Tomov VT. ( 2017). Gut microbiota and IBD: causation or correlation? Nat Rev Gastroenterol Hepatol 14: 573–84.
  • Nicholls A , Harris-Collazo R , Huang M , et al. ( 2013). Bioavailability profile of Uceris MMX extended-release tablets compared with Entocort EC capsules in healthy volunteers. J Int Med Res 41: 386–94.
  • Niebel W , Walkenbach K , Béduneau A , et al. ( 2012). Nanoparticle-based clodronate delivery mitigates murine experimental colitis. J Control Release 160: 659–65.
  • Ogueri KS , Shamblin SL. ( 2022). Osmotic-controlled release oral tablets: Technology and functional insights. Trends Biotechnol 40: 606–19.
  • Oshi MA , Lee J , Kim J , et al. ( 2021). pH-Responsive Alginate-Based Microparticles for Colon-Targeted Delivery of Pure Cyclosporine A Crystals to Treat Ulcerative Colitis. Pharmaceutics 13: 1412.
  • Oshi MA , Lee J , Naeem M , et al. ( 2020). Curcumin nanocrystal/pH-responsive polyelectrolyte multilayer core–shell nanoparticles for inflammation-targeted alleviation of ulcerative colitis. Biomacromolecules 21: 3571–81.
  • Oshi MA , Naeem M , Bae J , et al. ( 2018). Colon-targeted dexamethasone microcrystals with pH-sensitive chitosan/alginate/Eudragit S multilayers for the treatment of inflammatory bowel disease. Carbohydr Polym 198: 434–42.
  • Pagnini C , Pizarro TT , Cominelli F. ( 2019). Novel pharmacological therapy in inflammatory bowel diseases: beyond anti-tumor necrosis factor. Front Pharmacol 10: 671.
  • Pang L , Wang T , Liao Q , et al. ( 2022). Protective role of ergothioneine isolated from Pleurotus ostreatus against dextran sulfate sodium-induced ulcerative colitis in rat model. J Food Sci 87: 415–26.
  • Peng C-L , Yang L-Y , Luo T-Y , et al. ( 2010). Development of pH sensitive 2-(diisopropylamino) ethyl methacrylate based nanoparticles for photodynamic therapy. Nanotechnology 21: 155103.
  • Peng X , Wilken SE , Lankiewicz TS , et al. ( 2021). Genomic and functional analyses of fungal and bacterial consortia that enable lignocellulose breakdown in goat gut microbiomes. Nat Microbiol 6: 499–511.
  • Piechota-Polanczyk A , Fichna J. ( 2014). The role of oxidative stress in pathogenesis and treatment of inflammatory bowel diseases. Naunyn Schmiedebergs Arch Pharmacol 387: 605–20.
  • Qi S , Luo R , Han X , et al. ( 2022). pH/ROS Dual-Sensitive Natural Polysaccharide Nanoparticles Enhance “One Stone Four Birds” Effect of Rhein on Ulcerative Colitis. ACS Appl Mater Interfaces 14: 50692–709.
  • Ribeiro LN , Alcântara AC , Darder M , et al. ( 2014). Pectin-coated chitosan–LDH bionanocomposite beads as potential systems for colon-targeted drug delivery. Int J Pharm 463: 1–9.
  • Rutgeerts P , Sandborn WJ , Feagan BG , et al. ( 2005). Infliximab for induction and maintenance therapy for ulcerative colitis. N Engl J Med 353: 2462–76.
  • Salvi VR , Pawar P. ( 2019). Nanostructured lipid carriers (NLC) system: A novel drug targeting carrier. J Drug Delivery Sci Technol 51: 255–67.
  • Sandborn WJ , Feagan BG , Marano C , et al. ( 2014a). Subcutaneous golimumab induces clinical response and remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology 146: 85–95.
  • Sandborn WJ , Feagan BG , Marano C , et al. ( 2014b). Subcutaneous golimumab maintains clinical response in patients with moderate-to-severe ulcerative colitis. Gastroenterology 146: 96–109.e1. e101.
  • Sandborn WJ , Su C , Sands BE , et al. ( 2017). Tofacitinib as induction and maintenance therapy for ulcerative colitis. N Engl J Med 376: 1723–36.
  • Sandborn WJ , Van Assche G , Reinisch W , et al. ( 2012). Adalimumab induces and maintains clinical remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology 142: 257–65. e253.
  • Sardo HS , Saremnejad F , Bagheri S , et al. ( 2019). A review on 5-aminosalicylic acid colon-targeted oral drug delivery systems. Int J Pharm 558: 367–79.
  • Sarvar DP , Shamsasenjan K , Akbarzadehlaleh P. ( 2016). Mesenchymal stem cell-derived exosomes: new opportunity in cell-free therapy. Adv Pharm Bull 6: 293–9.
  • Sc T , Lj W. ( 1955). Cortisone in ulcerative colitis; final report on a therapeutic trial. British Medical Journal 2: 1041–8.
  • Sharif MK , Butt MS , Sharif HR. ( 2017). Role of nanotechnology in enhancing bioavailability and delivery of dietary factors. Nutrient delivery. London, United Kingdom: Elsevier; 587–618.
  • Shi H , Zhao X , Gao J , et al. ( 2020). Acid-resistant ROS-responsive hyperbranched polythioether micelles for ulcerative colitis therapy. Chin Chem Lett 31: 3102–6.
  • Singh AK , Yadav TP , Pandey B , et al. ( 2019). Engineering nanomaterials for smart drug release: Recent advances and challenges. Applications of targeted nano drugs and delivery systems.411–49.
  • Singh D , Srivastava S , Pradhan M , et al. ( 2015). Inflammatory bowel disease: pathogenesis, causative factors, issues, drug treatment strategies, and delivery approaches. Critical Reviews™ in Therapeutic Drug Carrier Systems 32:181–214.
  • Singh N , Khanna R. ( 2012). Colon targeted drug delivery systems-A Potential Approach. The Pharma Innovation 1:40–47.
  • Sinha SR , Nguyen LP , Inayathullah M , et al. ( 2015). A thermo-sensitive delivery platform for topical administration of inflammatory bowel disease therapies. Gastroenterology 149: 52–5.e2. e52.
  • Soni K , Kukereja BK , Kapur M , Kohli K. ( 2015). Lipid nanoparticles: future of oral drug delivery and their current trends and regulatory issues. Int J Curr Pharm Rew Res 7: 1–8.
  • Sun J , Xue P , Liu J , et al. ( 2021). Self-Cross-Linked Hydrogel of Cysteamine-Grafted γ-Polyglutamic Acid Stabilized Tripeptide KPV for Alleviating TNBS-Induced Ulcerative Colitis in Rats. ACS Biomater Sci Eng 7: 4859–69.
  • Sutherland LR , MacDonald JK. ( 2006). Oral 5-aminosalicylic acid for maintenance of remission in ulcerative colitis. Cochrane Database of Systematic Reviews.(2).
  • Talaei F , Atyabi F , Azhdarzadeh M , et al. ( 2013). Overcoming therapeutic obstacles in inflammatory bowel diseases: a comprehensive review on novel drug delivery strategies. Eur J Pharm Sci 49: 712–22.
  • Tan C , Fan H , Ding J , et al. ( 2022). ROS-responsive nanoparticles for oral delivery of luteolin and targeted therapy of ulcerative colitis by regulating pathological microenvironment. Mater Today Bio 14: 100246.
  • Tang Q , Zhang W , Zhang C , et al. ( 2020). Oxymatrine loaded nitric oxide-releasing liposomes for the treatment of ulcerative colitis. Int J Pharm 586: 119617.
  • Tao Y , Zhao X , Liu X , et al. ( 2022). Oral delivery of chitosan-coated PLGA nanoemulsion loaded with artesunate alleviates ulcerative colitis in mice. Colloids Surf B Biointerfaces 219: 112824.
  • Tatiya-Aphiradee N , Chatuphonprasert W , Jarukamjorn K. ( 2018). Immune response and inflammatory pathway of ulcerative colitis. J Basic Clin Physiol Pharmacol 30: 1–10.
  • Thomas S , Hoxha K , Alexander W , et al. ( 2019). Intestinal barrier tightening by a cell-penetrating antibody to Bin1, a candidate target for immunotherapy of ulcerative colitis. J Cell Biochem 120: 4225–37.
  • Timmer A , Patton PH , Chande N , et al. ( 2016). Azathioprine and 6-mercaptopurine for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev 2016: CD000478.
  • Toumi R , Samer A , Soufli I , et al. ( 2021). Role of probiotics and their metabolites in inflammatory bowel diseases (IBDs). Gastroenterology Insights 12: 56–66.
  • Verma S , Kumar V , Mishra D , Singh S. ( 2012). Colon targeted drug delivery: current and novel perspectives. Int J Pharm Sci Res 3: 1274.
  • Vinarov Z , Abdallah M , Agundez JA , et al. ( 2021). Impact of gastrointestinal tract variability on oral drug absorption and pharmacokinetics: An UNGAP review. Eur J Pharm Sci 162: 105812.
  • Vong LB , Tomita T , Yoshitomi T , et al. ( 2012). An orally administered redox nanoparticle that accumulates in the colonic mucosa and reduces colitis in mice. Gastroenterology 143: 1027–36.e3. e1023.
  • Wan Y , Zhang B. ( 2022). The impact of zinc and zinc homeostasis on the intestinal mucosal barrier and intestinal diseases. Biomolecules 12: 900.
  • Wang B , Zhuang X , Deng Z-B , et al. ( 2014). Targeted drug delivery to intestinal macrophages by bioactive nanovesicles released from grapefruit. Mol Ther 22: 522–34.
  • Wang N , Shao L , Lu W , et al. ( 2022). 5-aminosalicylic acid pH sensitive core-shell nanoparticles targeting ulcerative colitis. J Drug Delivery Sci Technol 74: 103578.
  • Wang X , Gu H , Zhang H , et al. ( 2021). Oral Core–Shell Nanoparticles Embedded in Hydrogel Microspheres for the Efficient Site-Specific Delivery of Magnolol and Enhanced Antiulcerative Colitis Therapy. ACS Appl Mater Interfaces 13: 33948–61.
  • Wilson DS , Dalmasso G , Wang L , et al. ( 2010). Orally delivered thioketal nanoparticles loaded with TNF-α–siRNA target inflammation and inhibit gene expression in the intestines. Nat Mater 9: 923–8.
  • Xiao B , Merlin D. ( 2012). Oral colon-specific therapeutic approaches toward treatment of inflammatory bowel disease. Expert Opin Drug Deliv 9: 1393–407.
  • Xu H , Luo R , Dong L , et al. ( 2022). pH/ROS dual-sensitive and chondroitin sulfate wrapped poly (β-amino ester)-SA-PAPE copolymer nanoparticles for macrophage-targeted oral therapy for ulcerative colitis. Nanomedicine 39: 102461.
  • Xu Q , Zhang N , Qin W , et al. ( 2013). Preparation, in vitro and in vivo evaluation of budesonide loaded core/shell nanofibers as oral colonic drug delivery system. J Nanosci Nanotechnol 13: 149–56.
  • Xu X-R , Liu C-Q , Feng B-S , Liu Z-J. ( 2014). Dysregulation of mucosal immune response in pathogenesis of inflammatory bowel disease. World J Gastroenterol 20: 3255–64.
  • Yan X , Yang C , Yang M , et al. ( 2022). All-in-one theranostic nano-platform based on polymer nanoparticles for BRET/FRET-initiated bioluminescence imaging and synergistically anti-inflammatory therapy for ulcerative colitis. J Nanobiotechnol 20: 1–18.
  • Yang M , Lai SK , Yu T , et al. ( 2014). Nanoparticle penetration of human cervicovaginal mucus: The effect of polyvinyl alcohol. J Control Release 192: 202–8.
  • Yang M , Zhang Y , Ma Y , et al. ( 2020). Nanoparticle-based therapeutics of inflammatory bowel diseases: a narrative review of the current state and prospects. Journal of Bio-X Research 3: 157–73.
  • Zhang C , Li J , Xiao M , et al. ( 2022). Oral colon-targeted mucoadhesive micelles with enzyme-responsive controlled release of curcumin for ulcerative colitis therapy. Chin Chem Lett 33: 4924–9.
  • Zhang C , Wang X , Xiao M , et al. ( 2022). Nano-in-micro alginate/chitosan hydrogel via electrospray technology for orally curcumin delivery to effectively alleviate ulcerative colitis. Materials & Design 221: 110894.
  • Zhang L , Zhu W , Yang C , et al. ( 2012). A novel folate-modified self-microemulsifying drug delivery system of curcumin for colon targeting. Int J Nanomedicine 7: 151–62.
  • Zhang M , Viennois E , Prasad M , et al. ( 2016). Edible ginger-derived nanoparticles: A novel therapeutic approach for the prevention and treatment of inflammatory bowel disease and colitis-associated cancer. Biomaterials 101: 321–40.
  • Zhang M , Wang X , Han MK , et al. ( 2017). Oral administration of ginger-derived nanolipids loaded with siRNA as a novel approach for efficient siRNA drug delivery to treat ulcerative colitis. Nanomedicine (Lond) 12: 1927–43.
  • Zhao X-X , Ma S-B , Wen J-P , et al. ( 2022). Reactive Oxygen Species-Responsive Polyether Micelle Nanomaterials for Targeted Treatment of Ulcerative Colitis. J Biomed Nanotechnol 18: 120–31.
  • Zhou X , Chen Z. ( 2015). Preparation and performance evaluation of emulsomes as a drug delivery system for silybin. Arch Pharm Res 38: 2193–200.
  • Zu M , Ma Y , Cannup B , et al. ( 2021). Oral delivery of natural active small molecules by polymeric nanoparticles for the treatment of inflammatory bowel diseases. Adv Drug Deliv Rev 176: 113887.
  • Zu M , Song H , Zhang J , et al. ( 2020). Lycium barbarum lipid-based edible nanoparticles protect against experimental colitis. Colloids Surf B Biointerfaces 187: 110747.