Dual-targeted pills synergistic treatment of UC | DDT

2021-12-06 17:43:02 By : Ms. Coral Chen

Javascript is currently disabled in your browser. When javascript is disabled, some functions of this website will not work.

Open access for scientific and medical research

From submission to the first editing decision.

From editor acceptance to publication.

The above percentage of manuscripts have been rejected in the past 12 months.

Open access to peer-reviewed scientific and medical journals.

Dove Medical Press is a member of OAI.

Batch reprints for the pharmaceutical industry.

We provide real benefits for authors, including fast processing of papers.

Register your specific details and specific drugs of interest, and we will match the information you provide with articles in our extensive database and send you a PDF copy via email in a timely manner.

Back to Journal »Drug Design, Development and Treatment» Volume 15

Oral dual-targeted pill for synergistic treatment of ulcerative colitis

Authors: Tang X, Yang Ming, Gu Yan, Jiang Li, Du Yan, Liu Jie

Published on September 29, 2021, the 2021 volume: 15 pages 4105-4123

DOI https://doi.org/10.2147/DDDT.S322702

Single anonymous peer review

Editor who approved for publication: Dr. Huang Tianhui

Xiaomeng Tang,1–3,* Meng Yang,4,* Yongwei Gu,1,2 Liangdi Jiang,1,2,5悦都,1,2,5 Jiyong Liu1–3 1 Department of Pharmacy, Shanghai Cancer Center, Fudan University, Fudan University, Shanghai, 200032; 2 Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032; 3 Department of Pharmacy, Changhai Hospital, Naval Medical University, Shanghai 200433; 4 Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University, Shanghai 200011; 5 Shandong Zhong School of Pharmacy, University of Medicine and Pharmacy, Jinan, Shandong, 250355 *The above authors have contributed equally to this article. Tel/Fax, People’s Republic of China +86-21-64175590 Email [email protected] Purpose: The effective treatment of ulcerative colitis (UC) is facing a huge challenge. The pathogenesis of UC is closely related to infection, immunity and environmental factors. At present, there is a great need to develop oral bioavailable dosage forms that can effectively deliver therapeutic drugs to local gastrointestinal diseases. Method: Berberine (BBR) and Atractylodes macrocephala (AM) volatile oils are derived from Chinese herbal medicines Rhizoma Coptidis and Atractylodes macrocephala, which have anti-inflammatory and immunomodulatory activities. In this study, we prepared BBR-loaded colon-targeted pellets and AM volatile oil-loaded stomach-targeted pellets for the synergistic treatment of UC. The Box-Behnken design and β-cyclodextrin inclusion technology were used to optimize the enteric coating formula and prepare the volatile oil inclusion compound. Results: Both particles are spherical and have satisfactory physical properties. The pharmacokinetic results showed that the AUC and MRT values ​​of the dual targeting (DPs) pellets were higher than those of the control pellets. In addition, in vivo animal imaging confirmed that DPs can effectively deliver BBR to the colon. In addition, compared with sulfasalazine and monotherapy, DPs inhibits the expression of inflammatory factors such as IL-1β, IL-4, IL-6, TNF-α and MPO in serum and tissues, enhances immunity, and exerts better performance. Significant anti-inflammatory effect. By reducing the production of IgA and IgG. Conclusion: DPs exerts a synergistic anti-UC effect by exerting systemic and local anti-inflammatory effects, and provides an effective oral targeted preparation for the treatment of UC. Keywords: ulcerative colitis, oral dual-targeted pellets, synergistic treatment, berberine, AM volatile oil

Ulcerative colitis (UC), known as chronic non-specific ulcerative colitis, is a chronic, debilitating inflammatory disease caused by a complex interaction between infection, immune, environmental, and microbial factors. 1 The main clinical manifestations of UC are digestive system symptoms and difficulty in healing, which can last from several months to decades. 2,3 Depending on the duration and degree of inflammation, patients with long-term UC (~18%) have a much higher risk of colitis-related colorectal cancer (CAC). 4-6 At present, adrenal cortex hormones (prednisone) and aminosalicylic acid (mesalazine) are commonly used drugs for the treatment of UC, but these drugs can only relieve symptoms and often have more side effects. In addition, the effectiveness and safety of other reported drugs, such as immunosuppressants (azathioprine), vary greatly from individual to individual. 7 Therefore, there is a great need to develop expedient measures and practical preparations that can effectively treat UC.

Atractylodes macrocephala (AM) has long been used for the treatment of gastrointestinal hypofunction. 8 Volatile oil contains volatile components extracted from AM, which can improve gastrointestinal function, enhance immunity and exert anti-inflammatory effects. 9 However, the composition of AM volatile oil has poor stability and is easily decomposed and deteriorated under the action of oxygen, light and heat. According to reports, the cyclodextrin inclusion compound technology can prevent the oxidation and decomposition of the volatile oil, and at the same time make the liquid medicine into a powder, which can improve the stability of the medicine during the preparation and storage process. 10 Further granulation of the inclusion compound can facilitate patient administration and mask the unpleasant odor of volatile oil.

Berberine (BBR) is the active ingredient of Coptis (CC), which can reduce the symptoms of colitis, reduce mucosal barrier damage, 11 and restore barrier function. 12 However, BBR is a BCS II compound with low bioavailability when taken orally. 13 Therefore, it is necessary to improve the solubility and bioavailability of BBR to promote targeted delivery of BBR to diseased colon tissues to enhance local anti-inflammatory effects.

The dosage form plays an important role in the absorption and transport of oral preparations. However, clinically, the treatment of UC is always absorbed or degraded before it reaches the colon, resulting in less accumulation in the ulcer lesion, which affects the efficacy. Oral Colon Targeted Drug Delivery System (OCDDS) adopts appropriate preparation technology to prevent the release of drugs in the stomach, duodenum, jejunum and ileum after oral administration. 14,15 Therefore, it can deliver drugs to 16 Oral colon-targeted drug delivery systems. There are many types, including pH type, enzyme contact type, time-dependent type, pressure control type, bioadhesive type, prodrug type, combination Types, etc.17,18 At present, most of the varieties that have been marketed or entered clinical research are coated with pH-dependent materials. In addition, pellets are a multi-unit drug delivery system that can increase the contact area between the drug and the gastrointestinal tract, and promote absorption and bioavailability. 19 Therefore, the development of oral colon-targeted pellets may improve the oral bioavailability of BBR and colon-targeting.

At present, most of the commercially available OCDDS are pH-sensitive coating types based on the pH of the gastrointestinal tract (0.9-1.5 for stomach, 6.5-7 for small intestine, 6.8-7.5 for colon). 20 Directly sold polymer materials, such as EUDRAGIT® L 30D-55 and EUDRAGIT® FS 30D, have inherent pH sensitivity and are not suitable for targeted drug delivery to the colon. 21 Therefore, we optimized the mixed polymer materials to enable the coating formulation to release a large amount of drug after administration. Reach the colon.

In this study, BBR and AM volatile oil were loaded into colon-targeted pellets and stomach-targeted pellets, respectively, to prepare DPs for synergistic treatment of UC. After administration, BBR is released in the colon and exerts a local targeted anti-inflammatory effect, while AM ​​volatile oil is absorbed in the stomach and exerts a systemic immunomodulatory effect. In this study, we optimized the particle preparation process, characterized the physical and chemical properties of the particles, and evaluated colon targeting. In addition, the pharmacokinetic characteristics of DPs in rats were also tested. Construct a DSS-induced rat UC model to study the pharmacodynamic properties of DP. These results may have a significant contribution to the management of UC and may provide pharmaceutical strategies for the treatment of gastrointestinal diseases.

Atractylodes macrocephala volatile oil and Coptis chinensis extract were purchased from Xi’an Xiaocao Plant Technology Co., Ltd. (Xi’an, Shanxi, China). Atractylodes lactone I (AT-1) and berberine hydrochloride standard products were purchased from the State Food and Drug Administration (Beijing, China). β-Cyclodextrin (β-CD) was purchased from Tianli Pharmaceutical Excipients Co., Ltd. (Qufu, Shandong, China). Microcrystalline cellulose (MCC) was purchased from Huzhou Linghu Xinwang Chemical Co., Ltd. (Huzhou, Zhejiang, China). Lactose was purchased from DMV-Fonterra Excipients GmbH & Co. KG (Goch, Germany). Cross-linked polyvinylpyrrolidone XL was purchased from Shanghai Yuanye Biotechnology Co., Ltd. (Shanghai, China). Tween 80 was purchased from Shanghai Lingfeng Chemical Reagent Co., Ltd. (Shanghai, China). EUDRAGIT® FS 30 D and EUDRAGIT® L 30D-55 were donated by Evonik Rohm GmbH. (Darmstadt, Germany). Hypromellose (HPMC), triethyl citrate (TEC) and sulfasalazine (SASP) were purchased from Aladdin Reagent Co., Ltd. (Shanghai, China). Glyceryl monostearate (GMS) was purchased from Alfa Aesar Chemical Co., Ltd. (Ward Hill, US). Dextran sulfate sodium salt (DSS, MV: 36000~5000) was purchased from Shanghai Yisheng Biotechnology Co., Ltd. (Shanghai, China). Sulfo CY7 NHS ester was purchased from Xi'an Ruixi Biotechnology Co., Ltd. (Xi'an, Shanxi, China). HPLC grade acetonitrile and methanol were purchased from Tedia Co., Ltd. (Shanghai, China). Other chemicals and solvents used in the study are of analytical grade.

Sprague-Dawley (SD) rats (male, 180 ± 20 g, Experimental Animal Center, Shanghai Naval Medical University, China; certificate number: SCXK (Hu) 2017–0002) and BALC/c mice (male, 18 ± 2 g) ) Used in all in vivo studies. The rats were acclimated for at least 7 days. Animal experiments were carried out in accordance with the protocol evaluated by the ethics committee of the Second Military Medical University (Shanghai, China), and were in compliance with the National Institutes of Health guidelines for the care and use of laboratory animals.

AM volatile oil/β-CD inclusion compound was first prepared to improve oxidative deterioration and at the same time mask the taste. At the same time, the solidification of the liquid drug also laid the foundation for the subsequent preparation of gastric-coated pellets. Use differential scanning calorimetry (DSC) and X-ray powder diffraction (XRD) to analyze the resulting product to confirm whether the inclusions are successful. The cores of GPs and EPs are prepared by extrusion spheronization. EPs cores coated with 4% HPMC as a sub-coating (Sub-Ps) were also prepared for further coating. The enteric coating dispersion is prepared by mixing EUDRAGIT® FS 30D, EUDRAGIT® L 30D-55, GMS, TEC, Tween-80 and purified water. The coating process parameters are summarized in Table S1. For a more detailed description of the preparation process and characterization parameters, please refer to the Supplementary Material Methods.

Polymer ratio, plasticizer concentration and coating weight gain are the three key factors that regulate the release rate of film-controlled pellets. Therefore, in previous studies, the value range of each factor was determined through single-factor experiments. Since then, the Box-Behnken design (BBD) has EUDRAGIT®FS 30D and EUDRAGIT®L 30D-55 (g/g, X1), plasticizer concentration (%, X2) and coating weight gain (%, X3) for Optimize the enteric coating formula. This technique is suitable for the study of the secondary response surface, so that the optimization process can be run through a small number of experiments. The design consists of a replicated center point and a set of points located at the midpoint of each edge of a multidimensional cube that defines the area of ​​interest. 19

Response surface modeling (RSM) was performed using Design Expert 8.0 (StateEase Inc., Minneapolis, MN). Table 1 summarizes the factors and levels of the independent variables. The selected dependent variable is the cumulative release percentage value of BBR dissolved at a certain time (after 2, 6, and 12 hours). Table 1 Factors and levels in Box-Behnken design and response

Table 1 Factors and levels in Box-Behnken design and response

The particle size is represented by the average value of the length and width determined using a digital vernier caliper (Shanghai Minnet Industrial Co., Ltd., Shanghai, China). For each particle type, randomly characterize 20 particles and calculate the average result. The particle size distribution of 22 GPs and EPs is determined by the sieving method, and the diameter is 550-880 μm (30-18 mesh) or 830-1400 μm (20-12 mesh). The result is displayed as a percentage of each size score. The particles coated with a thin gold layer were inspected by a scanning electron microscope (SEM; HITACHI-S3400N, Japan) to analyze the shape and surface morphology. twenty three

Plane Critical Stability (PCS) is used to reflect the roundness of particles. During the measurement, 20 g of pellets are placed flat on the glass plate, and one end of the glass plate is slowly lifted. Measure the critical angle (θ) of the plane formed by the inclined surface and the horizontal plane before the ball starts rolling. The smaller the value of θ, the better the roundness of the pellets. 19

A multi-purpose tablet tester (78X-3C, Shanghai Huanghai Pharmaceutical Testing Instrument Co., Ltd., Shanghai, China) was used to determine the friability (Fr) of the pills. Weigh approximately 50 grams of both particle types and add them to the wear drum along with 200 glass beads (2 mm in diameter). The wear drum rotates at a speed of 25 r/min for 10 minutes. The content of the abrasive drum is sieved with 550μm (30 mesh) or 830μm (20 mesh), and the part below 550μm or 830μm is weighed. The friability is measured in triplicate and calculated as follows. 19

Weigh 50 g of particles (M), place them in a 100 mL glass measuring cylinder, and then drop them 5 cm away from the table to measure the bulk density. 24 Accurately read the volume (V) occupied by the particles, and the volume density (d) is obtained by the formula d = M/V. Three batches of particles are measured in parallel.

Drug loading is defined as the percentage of drug embedded in the pellet per unit weight. Grind 50 mg of granules and dissolve in 50 ml of methanol. Then, the mixture was sonicated and filtered through a 0.45 μm membrane filter. The samples are stored at 4 °C for testing. 25

For EP, HPLC (Agilent 1200 series, USA) was used to analyze the BBR concentration. The mobile phase consists of acetonitrile, water and 0.05 mol/L potassium dihydrogen phosphate (adjusted to pH=3, 30/70, v/v with phosphoric acid). The flow rate and wavelength were set to 1 mL/min and 345 nm, respectively. For GP, HPLC-MS/MS (Agilent Technologies, USA) was used to quantitatively analyze AT-1 (the active ingredient in AM volatile oil). The mobile phase consists of methanol, water and 0.1% (v/v) acetic acid (75/25, v/v). Other parameters are as follows: column temperature, 35°C; flow rate, 0.3 mL/min; dry gas (N2) temperature, 350°C; atomizer gas (N2) pressure, 20 psi; fragmentation voltage, 165 eV; collision energy, 35 eV; positive ion monitoring mode; and m/z = 231.1→128.1.

The pH dissolution method was used to evaluate the drug release of the pellets. 26 GPs quickly and completely disintegrated in artificial gastric juice, with a high release rate (pH 1.2, cumulative release of more than 98% within 5 minutes). The release of BBR from EPs was determined by using ChP (2020 version) dissolution apparatus II (basket method) at a speed of 100 r/min in 750 mL of dissolution medium under sink conditions at 37 ± 0.5 °C. The dissolution test was performed as follows: pH 1.2 for 2 hours, then pH 6.8 phosphate buffer for 4 hours, and pH 7.6 phosphate buffer for 18 hours. 27 After filtration, the concentration of BBR (0.45 μm) in different samples was measured by HPLC. The dissolution test of Sub-Ps and EPs was performed 6 times.

In order to study the transport and release characteristics of EPs in the gastrointestinal tract of mice, mice were administered EPs containing fluorescein (during the preparation of pellet cores, CY7 aqueous solution was used instead of water as a wetting agent) or uncoated pellet cores. Then, the mice were anesthetized with isoflurane and fixed on an imager (Quick View 3000, Bio-Real Sciences Technology Co., Ltd.) at predetermined time points (0 h, 1 h, 2 h, 4 h, 6 h). , 8 h, 10 h) after administration. The excitation light wavelength is 655 nm, the emission light wavelength is 716 nm, and the exposure time is 3 s.28

The rats were fasted for 12 hours and drank freely. They were randomly divided into two groups with 3 rats in each group. Both groups were orally administered DPs or APIs (a mixture of BPC, BBR and AM volatile oil) at a dose of 200 mg/kg BBR and 50 mg/kg AT-1. Pass through the retro-orbital vein for 0, 0.5, 1, 2, 4, 6, 8, 10, 12, 18, 21, 24, 27, 30, 33, 36, 42, 48, and 54 hours. Immediately centrifuge the blood sample at 3000 rpm (Eppendorf (China) Co., Ltd., Shanghai, China) for 15 minutes. Then store the supernatant in a refrigerator at -20 °C for further analysis. Prior to analysis, plasma samples were subjected to protein precipitation with methanol and acetonitrile, and dried with nitrogen at 40 °C. The sample was reconstituted in 100 μL methanol and centrifuged, and then analyzed by HPLC-MS/MS.

Standard methods were used to calculate the pharmacokinetic parameters AUC, half-life (t1/2) and MRT using Kinetica 5.0 software (Thermo Fisher Scientific Inc. Waltham, MA, USA). Cmax and Tmax are calculated directly from the curve of plasma concentration versus time. In addition, the relative bioavailability (Frel) of the reference development DPs group compared with the BPC group AUC is calculated using the formula given below. 29

Where Frel is the relative bioavailability, AUCA and AUCB are the area under the drug concentration-time curve of DP (test) and BPC (reference), respectively.

The concentrations of BBR and AT-1 in plasma were also determined using HPLC-MS/MS. The mobile phase consists of acetonitrile (phase A) and water (phase B) containing 0.1% (v/v) acetic acid. For BBR, m/z = 336.2→320.2, the operating parameters are set as follows: atomizing gas (N2), 20 psi; fragmentation voltage, 155 eV; collision energy, 30 eV; and dry gas (N2) temperature, 320 °C . For AT-1, the flow comparison is A:B=75:25, and the remaining parameters are the same as before.

According to the previously described procedure, UC was induced by substituting a 5% (w/v) DSS solution for the rat’s drinking water for 7 days. 30 80 rats were randomly divided into eight groups: group A (normal control group) received tap water for 14 days; group B (model control group) received DSS (5% w/v) in drinking water for 6 days, after which the animals received Ordinary tap water for 7 days; group C received DSS (5% w/v) water treatment for 6 days, and then treated with sulfasalazine (SASP suspended in 0.5% w/v normal saline, 100 mg/kg). The DH group received DSS similar to the C group (5% w/v). Group D received GP (equivalent to 50 mg/kg AT-1), group E received EP for 7 days after DSS (equivalent to 200 mg/kg BBR). After DSS administration, group FH received DPs (equivalent to 50 mg/kg BBR). kg AT-1 and 50, 100, 200 mg/kg BBR) for 7 days. The rats were taken orally once a day for 7 days (groups A and B were treated with saline instead of therapeutic drugs), and their body weight was recorded every day. The start of UC model induction is considered to be the first day. The body weight of the rats is recorded every day. Blood samples collected from rats through the retro-orbital vein are used to determine the concentration of IL-1β, IL-4, IL-6, TNF-α, MPO, IgA and IgG. The rats were sacrificed by cervical dislocation on the 14th day. Measure the length of the colon from the ileocecal junction to the edge of the anus. After that, colon tissue was collected for scoring, gross morphology, histopathological analysis, and determination of the expression levels of various inflammatory factors. In addition, the spleen and thymus were separated to calculate the spleen and thymus index.

The dissected colon tissue was cut longitudinally along the edge of the mesentery, washed with ice-cold normal saline to remove the contents of the lumen, and then weighed. The colon/body weight ratio (C/B ratio) is an index used to quantify inflammation and is calculated as the quotient of the wet weight of the 10 cm colon and the total weight of each rat. 31

Assess the clinical activity of colitis, including weight loss, stool consistency, and rectal bleeding to confirm the disease activity index (DAI). DAI is determined by calculating the average of the above three parameters in the range of 0-4 according to the standards shown in Table S3.32-34

In order to assess the damage visible to the naked eye, the entire distal colon was examined by an independent observer who did not know the study group. Table S4.35, 36 describes the standards and scales used to classify macroscopic injuries

The histological scoring system is used to assess microscopic colon changes. A 1 cm sample of colon tissue was cut from the distal colon and prepared as a "Swiss roll" for full-length histopathological evaluation. The "Swiss rolls" of each group were immersed in 10% neutral formaldehyde and then embedded in paraffin wax. Sections with a thickness of 5 μm were stained with hematoxylin and eosin (H&E) to assess the degree of inflammation. The slides were then unknowingly examined by a pathologist based on a scale that grades the presence of edema, erosion-ulcers, crypt lesions, and the percentage of inflammatory areas using the standard optical microscope previously described. The basis of inflammation score based on microscopic (100×) histological changes assessment is shown in Table S5.37,38

Cytokines are considered to be key signals in the intestinal immune system. Immune cells, such as T cells, dendritic cells, macrophages and intestinal epithelial cells, participate in the secretion of various cytokines that regulate the inflammatory response of UC. 39 Therefore, IL-1β (Multi Sciences Rat IL-1β ELISA Kit, Hangzhou, China), IL-4 (Multi Sciences Rat IL-4 ELISA Kit, Hangzhou, China), IL-6 (Multi Sciences Rat IL-6 ELISA Kit, Hangzhou, China) Kit) levels, Hangzhou, China) and TNF-α (Thermo Fisher Rat TNF-α ELISA kit, Vienna, Austria) were measured using an appropriate ELISA kit according to the manufacturer’s instructions. The levels of MPO (Cusabio Rat MPO ELISA kit, Wuhan, China), IgA (Multi Sciences Rat IgA ELISA kit, Hangzhou, China) and IgG (Multi Sciences Rat IgG ELISA kit, Hangzhou, China) in serum samples were also Use commercial measurements to provide usable ELISA kits according to the manufacturer's instructions. The MPO activity unit is defined as the amount of enzyme that converts 1 μmol of hydrogen peroxide into water in 1 minute at room temperature. The results are expressed in U/g organization.

All data are expressed as mean±standard deviation, and SPSS 20.0 statistical software is used for statistical analysis. The comparison between parametric data sets used one-way analysis of variance (ANOVA) followed by Bonferroni test. Kruskal-Wallis test and Mann-Whitney test were used for nonparametric statistical analysis to assess differences between groups. A threshold of P <0.05 was defined as statistically significant.

The experimental runs using independent variables and the observed responses of the 17 formulations are shown in Table 2. Based on the Box-Behnken model, factor combinations lead to different drug release rates of EP. The cumulative release rate within 6 hours is very low (less than 10%), indicating that the main limiting factor is the barrier effect of the enteric coating. However, the cumulative release rate in PBS buffer (pH 7.6, 6-12 hours) is much higher (over 70%). In order to determine the level of factors that produce the best dissolution response, the experimental design software Design-Expert 8.0 was used to generate a mathematical relationship between the dependent variable and the independent variable. The equations derived from the coding factors of all responses are as follows: (2) (3) (4) Table 2 Experimental runs designed by Box-Behnken and the observed response values

Table 2 The experimental runs designed by Box-Behnken and the observed response values

Equations (2)–(4) respectively represent the quantitative effects of the independent variables X1-X3 of the formula on the three related responses Y1-Y3. The independent values ​​of X1-X3 are determined in the equation. (2)-(4) Get the predicted value of Y1-Y3. In addition, the three-dimensional (3D) response surface and two-dimensional (2D) contour plots shown in Figure 1 are used to clarify and analyze the relationship between the independent variable and the dependent variable. Figure 1 Variable (X1-X3) vs. response (Y1-Y3): 3D response surface plot and 2D contour plot. X1: the ratio of EUDRAGIT®FS 30D to EUDRAGIT®L 30D-55; X2: plasticizer concentration (based on polymer dry weight); X3: coating weight gain. Y1-Y3: Cumulative drug release for 2 hours, 6 hours and 12 hours. (A) The influence of X1 and X2 on Y1; (B) The influence of X2 and X3 on Y1; (C) The influence of X1 and X3 on Y1; (D) The influence of X1 and X2 on Y2; (E) X2 and X3 The influence on Y2; (F) the influence of X1 and X3 on Y2; (G) the influence of X1 and X2 on Y3; (H) the influence of X2 and X3 on Y3; (I) the influence of X1 and X3 on Y3.

Figure 1 The influence of variables (X1-X3) on response (Y1-Y3): 3D response surface plot and 2D contour plot. X1: the ratio of EUDRAGIT®FS 30D to EUDRAGIT®L 30D-55; X2: plasticizer concentration (based on polymer dry weight); X3: coating weight gain. Y1-Y3: Cumulative drug release for 2 hours, 6 hours and 12 hours. (A) The influence of X1 and X2 on Y1; (B) The influence of X2 and X3 on Y1; (C) The influence of X1 and X3 on Y1; (D) The influence of X1 and X2 on Y2; (E) X2 and X3 The influence on Y2; (F) the influence of X1 and X3 on Y2; (G) the influence of X1 and X2 on Y3; (H) the influence of X2 and X3 on Y3; (I) the influence of X1 and X3 on Y3.

It can be seen from Figure 1 that when X1, X2, and X3 increase, Y1, Y2, and Y3 also increase. It is found that the ratio of EUDRAGIT®FS 30D to EUDRAGIT®L 30D-55 (X1), plasticizer concentration (X2) and coating weight gain (X3) affect Y1, Y2 and Y3 (2 hours, 6 hours and 12 hours) . When X1, X2, and X3 are 2.0, 11.7, and 20.0, respectively, the Y1, Y2, and Y3 values ​​predicted by the model are 2.50, 6.34, and 70.92. The results of three verification experiments show that the predicted value and the observed value are quite close (Table 3). 40 Table 3 Comparison between the observed and predicted responses of the optimized enteric coating layer

Table 3 Comparison of the observed and predicted response of the optimized enteric coating layer

Figure 2A shows the DSC curves of β-CD, volatile oil/β-CD physical mixture, volatile oil and volatile oil/β-CD inclusion compound. The thermogram of β-CD shows a broad endothermic peak at approximately 145.48 °C, which indicates the release of water. There is another endothermic peak at about 314.24 °C, which is mainly related to the phase transition of β-CD. The volatile oil exhibits an endothermic effect at approximately 307.40 °C, which is caused by the decomposition of the volatile oil. The thermogram of the physical mixture is similar to the superposition of separate thermograms of β-CD and volatile oil. However, the thermogram of the inclusion compound showed a different pattern, and the two characteristic peaks of β-CD disappeared, indicating that the water molecules in the β-CD cavity had shifted. 41 In addition, a new characteristic peak appeared at 277.49 °C. We speculated that this was due to the intermolecular interaction, which produced a new phase instead of volatile oil and β-CD. These results indicate that the volatile oil is encapsulated in the cavity of β-CD. Figure 2 Using DSC (A) and XRD (B) to characterize β-CD inclusion compounds: (a) β-CD; (b) volatile oil/β-CD physical mixture; (c) volatile oil; (d) volatile oil/β- CD inclusion compound.

Figure 2 Using DSC (A) and XRD (B) to characterize β-CD inclusion compounds: (a) β-CD; (b) volatile oil/β-CD physical mixture; (c) volatile oil; (d) volatile oil/β- CD inclusion compound.

X-ray powder diffraction can be used to determine the crystal type of a crystalline compound. As shown in Figure 2B, β-CD shows many crystalline peaks between 5° and 50° (2θ = 8.88, 12.47, 18.76 and 27.05°), indicating that β-CD mainly exists in crystalline form. The volatile oil presents a large broad peak, indicating that it is in an amorphous state. The XRD spectrum of the physical mixture shows an approximate superposition of the individual spectra of β-CD and volatile oil. The inclusion compounds show different main peaks (2θ = 5.86, 8.98, 11.70, 17.44, and 21.06°) and unique peak patterns. Compared with physical mixtures, they have relatively broad bands. 41 The appearance of these new characteristic peaks indicates that the inclusion compound is a new crystal form.

The WG (%), yield (%), RSDw (%) and coating loss (%) of GP, sub-P and EP are listed in Table S2. The results show that the preparation process of GPs, Sub-Ps and EPs is reasonable and reproducible, and the process stability is good.

According to naked eye and SEM observations, GPs are white spheres or ellipsoids, while EPs are yellow and brown spheres with smooth and round surfaces (Figure 3A and C (a and b)). The morphological study of these two particles shows that GPs and EPs have uniform size, ideal narrow size distribution, regular round shape and smooth surface. Figure 3 Physical and chemical properties of particles. (A) The size and shape of the particles observed with the naked eye. (a) GPs and (b) EPs (B) In vitro release curves of Sub-Ps and EPs in different media (pH=1.2, 2 hours, pH=6.8, 4 hours, pH=7.6, 18 hours) (C) A magnified scanning electron micrograph of the particles. (a) Intact GP and (b) 100 times intact EP; (c) GPs cross section and (d) in 1000 times dissolution medium EPs cross section.

Figure 3 Physical and chemical properties of particles. (A) The size and shape of the particles observed with the naked eye. (a) GPs and (b) EPs (B) In vitro release curves of Sub-Ps and EPs in different media (pH=1.2, 2 hours, pH=6.8, 4 hours, pH=7.6, 18 hours) (C) A magnified scanning electron micrograph of the particles. (a) Intact GP and (b) 100 times intact EP; (c) GPs cross section and (d) in 1000 times dissolution medium EPs cross section.

The particle size distribution of the obtained coated particles is shown in Table 4. Most GP and EP are located between 700-830 μm (24-20 mesh) and 880-1180 μm (18-14 mesh). The SEM image of the cross section of the particle showing the structure of the internal area of ​​the particle is shown in Figure 3C (c). The sub-coating layer (Figure 3C (d)) and the polymer layer can be clearly observed in the cross-sectional view, which indicates that the particle core has a compact structure, and the polymer layer is tightly coated on the core. Table 4 Particle size distribution of GPs and EPs (n=3)

Table 4 Particle size distribution of GPs and EPs (n=3)

Roundness, friability and bulk density are three important product quality requirements. These parameters are listed in Table 5. The lower plane critical angle and friability are related to excellent roundness, indicating that both GP and EP have good rigidity to avoid being crushed in the fluidized bed coating process. Table 5 Physical characteristics of GP and EP (n=3, average ± SD)

Table 5 Physical characteristics of GP and EP (n=3, average ± SD)

Established HPLC-QQQ-MS and HPLC assay methods for BBR and AT-1 to calculate drug loading and in vitro drug release. The determination method has good linearity, and the intra-day/inter-day precision RSD is less than 2% at each concentration level, indicating that the determination precision is good. Under this premise, the loadings of BBR and AT-1 in EP and GP are 125.61 ± 0.1 mg/g and 17.11 ± 0.16 mg/g, respectively.

The release curves of Sub-Ps and EPs are shown in Figure 3B. The release of BBR from Sub-Ps without an enteric layer was immediate and almost completely released within 4 hours, indicating that Sub-Ps is not suitable for colonic administration. When Sub-P is coated with the enteric coating layer optimized by BBD-RSM, sufficient acid resistance can be obtained. The cumulative release of BBR in artificial gastric media (pH 1.2) for 2 h is less than 5%, and the cumulative release of BBR in artificial intestinal media (pH 6.8) for 4 h is less than 10%. Excitingly, almost 95% of the BBR release occurred in the artificial colon culture medium (pH 7.6) within the next 10 hours. The increase of the pH value of the dissolving solution (pH 7.6) significantly accelerates the dissolution rate.

The in vivo targeting evaluation results of EPs are shown in Figure 4. Compared with the coated pellets, the uncoated fluorescent pellets showed large spot-like fluorescence 1-2 hours after oral administration, indicating that they began to disintegrate after reaching the stomach. 4-6 hours after oral administration, the entire small intestine treated with uncoated pellets was full of fluorescence, indicating that most of the pellets had disintegrated at this time. After eight to ten hours, the fluorescence of the uncoated pellets in the colon gradually weakened due to complete disintegration, and the fluorescein was quenched by the intestinal contents. However, after oral administration of enteric-coated fluorescent pellets, the intestine from the ileocecal area to the end of the colon was full of fluorescence, and a large number of pellets collapsed to release the drug. Figure 4 Imaging of fluorescent colon targeting particles in mice.

Figure 4 Imaging of fluorescent colon targeting particles in mice.

The pharmacokinetic study was performed on SD rats after oral administration of DPs and BPC, and the constructed average plasma concentration-time curve is shown in Figure 5. The corresponding pharmacokinetic parameter list is shown in Table 6. As shown in Figure 5A, after oral administration of BPC and DPs, AT-1 can be quickly absorbed into the blood. The Tmax values ​​of the two formulations are the same at 1.00 ± 0.00 hours. The Cmax values ​​of BPC and DP were 109.49 ± 5.61 ng/mL and 106.94 ± 11.43 ng/mL (P>0.05). These results indicate that the inclusion of β-CD increases the solubility and solubility of the active ingredient in the volatile oil. However, the Tmax of AT-1 shows a "double peak" phenomenon, indicating that there may be secondary absorption in the blood. Interestingly, the AUC0→t value of the DPs group was slightly higher than that of the BPC group, and the relative bioavailability value was 153.13%. In addition, this model showed consistent AT-1 plasma concentrations, indicating that the DPs group has a higher bioavailability. In addition, the MRT value of the DPs group was higher than that of the BPC group, indicating that the AT-1 released by the pellets showed a certain slow-release effect. Table 6 The pharmacokinetic parameters of AT-1 and BBR after oral administration of DPs and BPC (n=3, mean±SD) Figure 5 AT-1 (A) and BBR (B) after oral administration in SD rats The pharmacokinetic characteristics of BPC and DP are single doses of 50 mg AT-1 and 200 mg BBR. Each value represents the mean ± SD (n = 3).

Table 6 The pharmacokinetic parameters of AT-1 and BBR after oral administration of DPs and BPC (n=3, mean±standard deviation)

Figure 5 The pharmacokinetic characteristics of AT-1 (A) and BBR (B) after oral administration of BPC and DP in SD rats with a single dose of 50 mg AT-1 and 200 mg BBR. Each value represents the mean ± SD (n = 3).

According to Figure 5B, the Tmax values ​​of BBR in the plasma of the BPC group and the DPs group were 2.00 ± 0.00 and 10.00 ± 0.00 h, respectively. In addition, the obvious time lag in the DPs group indicates that the coated pellets have good controlled release and colon-specific effects. Due to the enterohepatic circulation in the process of BBR absorption, the Tmax of both groups showed a "double peak" phenomenon. The Cmax of the 42 DPs group was significantly different from that of the BPC group. The relative bioavailability of the two experimental groups was 121.38%, indicating that the relative bioavailability of DPs was higher. In addition, the plasma drug concentration-time curve is relatively flat, and the MRT value of the DPs group is higher than that of the BPC group, indicating that the DPs group has a significant slow-release effect and can better stabilize the blood drug concentration, thereby increasing the bioavailability.

Figure 6 shows that the body weight of rats in the healthy control group A increased slightly over time. All DSS induction groups had the lowest body weight on the 7th or 8th day. During the experiment on these two days, the symptoms of blood in the stool and diarrhea were the most severe, indicating that the UC model was successfully established. After the 8th day, the body weight of almost all rats in the DSS induction group showed an upward trend, indicating that each administration group had a certain effect on alleviating the weight loss caused by UC. Among them, the G group and the SASP C group have the best effect. Figure 6 Changes in body weight over time. (A) control group, (B) model group, (C) SASP group, (D) GPs group, (E) EPs group, (F) DPs low-dose group, (G) DPs medium-dose group, (H)) DPs high-dose group (n = 10).

Figure 6 Changes in body weight over time. (A) control group, (B) model group, (C) SASP group, (D) GPs group, (E) EPs group, (F) DPs low-dose group, (G) DPs medium-dose group, (H)) DPs high-dose group (n = 10).

As shown in Figure 7, for all DSS induction groups, DAI scores increased rapidly and consistently within 6 days of the induction experiment. Starting from day 7, all drug-receiving groups showed a reduction in the severity of inflammation and a reduction in DAI scores after a lag time of 24 to 48 hours. The largest reduction in the medium and high dose groups of DPs was observed on day 14. This reflects the efficacy of the prepared DPs, and it is inferred that a dose of 50 mg/kg AT-1 + 100 mg/kg BBR in rats is sufficient to achieve a satisfactory therapeutic effect. In the case of group F, it was observed that the DAI index was significantly lower than that of model B group on day 14 (Figure 8A), indicating that inflammation partially reversed mg/kg BBR even at a dose of 50 mg/kg AT-1 + 50 . In addition, it was also found that the DAI scores of the G and H groups were significantly lower than that of the SASP C group (P<0.05), indicating that DPs have a better effect on UC-induced inflammation. However, in groups A and B, treatment was less effective in reducing DAI scores. This clearly shows that DP showed enhanced efficacy compared to GP or EP monotherapy, which resulted in a significant decrease in DAI score. Figure 7 Clinical Activity Score (DAI) throughout the experiment period. (A) control group, (B) model group, (C) SASP group, (D) GPs group, (E) EPs group, (F) DPs low-dose group, (G) DPs medium-dose group, (H)) DPs high-dose group (n = 10). Figure 8 (A) Disease activity index (DAI) scores of each group, (B) colon/weight ratio, (C) colonic mucosal injury index (CMDI) score and (D) microhistological changes on day 14 (, n = 10). Comparison: **P<0.01 vs control group, ΔP<0.05, ΔΔP<0.01 vs model group, ▲P<0.05, ▲▲P<0.01 vs SASP group, □P<0.05, □□P<0.01 vs DPs low- Dose group, ■P<0.05 vs DPs middle dose group, **P<0.01 vs control group.

Figure 7 Clinical Activity Score (DAI) throughout the experiment period. (A) control group, (B) model group, (C) SASP group, (D) GPs group, (E) EPs group, (F) DPs low-dose group, (G) DPs medium-dose group, (H)) DPs high-dose group (n = 10).

Figure 8 (A) Disease activity index (DAI) scores of each group, (B) colon/weight ratio, (C) colonic mucosal injury index (CMDI) score and (D) microhistological changes on day 14 (, n = 10). Comparison: **P<0.01 vs control group, ΔP<0.05, ΔΔP<0.01 vs model group, ▲P<0.05, ▲▲P<0.01 vs SASP group, □P<0.05, □□P<0.01 vs DPs low- Dose group, ■P<0.05 vs DPs middle dose group, **P<0.01 vs control group.

As shown in Figure 8B, the administration of GPs did not have a significant effect; however, in the other administration groups, the C/B ratio was significantly reduced.

The Colonic Mucosal Injury Index (CMDI) is shown in Figure 8C. The results showed that the colorectal morphology of the rats in the control group was normal, and the inner wall of the intestine was smooth and elastic. The color of the intestinal wall in all DSS induction groups was dark red. Diffuse edema and mucosal destruction appeared in various parts of the intestinal wall. Compared with the control group, the CMDI scores of rats in each administration group were significantly different (P<0.01). Except for the EPs group, there were significant differences between the administration group and the model group (P<0.05), indicating that the administration of each treatment has a certain effect on improving the colorectal mucosal injury. The scores of the DPs middle-dose group were lower than those of other administration groups, indicating that the DPs middle-dose group had better curative effect.

H&E stained colon tissue section (Figure 9) showed that the rats in the control group were basically normal. Colonic epithelial cells are intact, crypts are neatly arranged, goblet cells are abundant, and no signs of inflammation or tissue morphological damage are seen. However, in other groups, there was significant inflammatory cell infiltration in the colon. Mucosal epithelial cells fall off to varying degrees, goblet cells are reduced, and crypts are arranged disorderly. The model group showed a large area of ​​deep inflammatory cell infiltration, glandular deformation and loss of goblet cells. The degree of reduction of mucosal epithelial lesions in the colon tissue of rats in each administration group was different from that of rats in the model group. Some small ulcers still exist, but the histopathological score of the colon (Figure 8D) is lower than the model group. The colon tissue inflammation and pathological changes were significantly reduced in the DPs medium and high dose groups. The crypts and glands are arranged in an orderly manner and have excellent anti-inflammatory effects. Figure 9 H&E stained pathological colonic sections (200×) (A) control group, (B) model group, (C) SASP group, (D) GPs group, (E) EPs group, (F) DPs low-dose group , (G) DPs medium dose group, (H) DPs high dose group.

Figure 9 H&E stained pathological colonic sections (200×) (A) control group, (B) model group, (C) SASP group, (D) GPs group, (E) EPs group, (F) DPs low-dose group , (G) DPs medium dose group, (H) DPs high dose group.

As shown in Figure 10, the expression level of IL-1β in the serum and tissues of the DPs high-dose group of rats was not significantly different from that of the control group, indicating that the rats have basically recovered under the high-dose treatment. Dose DP. In serum, except for the GPs group, there were significant differences between the administration groups and the model group (P<0.01), indicating that the drug intervention effect is better than the self-recovery effect. The DPs treatment in the medium/high dose group had a significant effect on increasing the expression of IL-4 in serum and tissues (P<0.01), and the medium dose group had a more obvious promotion effect on the expression of IL-4. obviously. The control group was only significantly different from the model group, EPs group and GPs group (P<0.05), and the expression of IL-6 in the other groups was significantly inhibited. The expression level of IL-6 in the DPs middle/high dose group decreased more significantly (P<0.01). In addition, the expression level of TNF-α in the serum or tissues of the DPs group was not significantly different from that of the control group (P>0.05), indicating that DPs has a great effect on inhibiting the expression of TNF-α. α. In addition, there were significant differences between the DPs middle-dose group, DPs high-dose group, SASP group and the model group (P<0.01), indicating that the MPO activity of the three groups was effectively inhibited. All results show that DPs has a good anti-inflammatory effect, and its activity is even better than that of the positive control drug SASP and EPs alone. Figure 10 The effects of each treatment on the expression of inflammatory factors and immune-related indicators (, n=10). (A) Serum, (B) Colon tissue, (C) Serum. Comparison: *P<0.05, **P<0.01 vs control group, ΔP<0.05, ΔΔP<0.01 vs model group, ▲P<0.05, ▲▲P<0.01 vs SASP group, □□P<0.01 vs DPs low- Dose group, ■■ P<0.05 and DPs medium-dose group.

Figure 10 The effects of each treatment on the expression of inflammatory factors and immune-related indicators (, n=10). (A) Serum, (B) Colon tissue, (C) Serum. Comparison: *P<0.05, **P<0.01 vs control group, ΔP<0.05, ΔΔP<0.01 vs model group, ▲P<0.05, ▲▲P<0.01 vs SASP group, □□P<0.01 vs DPs low- Dose group, ■■ P<0.05 and DPs medium-dose group.

Compared with the control group, the spleen index and thymus index of model rats decreased, and the serum levels of IgA and IgG increased. Compared with the model group, the spleen index and thymus index of rats in each DPs dose group increased in a dose-dependent manner, and the serum IgA and IgG levels also decreased in a dose-dependent manner.

In this study, after treatment with GPs containing AM volatile oil, the expression levels of IgA and IgG were significantly reduced. However, these levels are still significantly different from those of the healthy control group. To our satisfaction, the concentration of IgA and IgG in the DPs group was significantly reduced. In particular, there was no significant difference between the DPs medium/high dose group and the control group (P>0.05), suggesting that DPs can improve the immune function of UC rats.

Ulcerative colitis (UC) is a common autoimmune disease that is difficult to cure and can even lead to colon cancer in severe cases. At present, the clinical treatment of UC is still dominated by drug methods, and commonly used chemotherapeutic drugs can only relieve symptoms, are difficult to cure, and have obvious adverse reactions, which reduce the patient's compliance. Traditional Chinese medicine has unique advantages in the treatment of chronic gastrointestinal diseases caused by many reasons. At the same time of symptomatic treatment, pay attention to the overall balance. In this study, two traditional Chinese medicines, BBR and AM volatile oil, were combined with an oral colon-specific drug delivery system to study the preparation and in vitro-in vivo evaluation of DPs. Our research results show that oral DPs can effectively alleviate the symptoms of UC rat models, and its anti-UC mechanism may not only be related to inhibiting inflammation, but also related to enhancing the body's immunity.

In recent decades, the most advanced small animal imaging modes have provided non-invasive images with a wealth of quantitative anatomical and functional information, which makes longitudinal research possible, allowing precise monitoring of disease progression and response to treatment in different disease models . 43 Our research is significant because it provides the possibility of using in vivo imaging to localize the delivery of oral formulations. Take pictures regularly to observe the transport of the pellets and drug release in the gastrointestinal tract. According to our assumptions, if the coating of the particle is not destroyed, it should emit dot or flake fluorescence. After the coating is dissolved, the punctate fluorescence gradually diffuses throughout the gastrointestinal tract with the release of the drug. Combining the fluorescent signal and the anatomical characteristics of the mouse gastrointestinal tract, the transport position of EPs in the gastrointestinal tract can be judged to evaluate its in vivo targeting properties. To our satisfaction, the results of the in vivo targeting evaluation show that EPs have good colon targeting properties and can be transported to the colon to release drugs after oral administration, thereby promoting drug absorption and improving drug efficacy.

MPO is an indicator of neutrophil recruitment in the mouse UC model. Therefore, compared with cytokine concentration, MPO activity may reflect more specific inflammatory events. In our study, compared with the normal control group, the colonic MPO activity was significantly increased in the DSS group, and the group receiving DPs showed a significant decrease in MPO activity (P <0.01) (Figure 10). Therefore, DPs prevent the infiltration of neutrophils into damaged tissues. Cytokines are considered to be key signals in the intestinal immune system. Immune cells, such as T cells, dendritic cells, macrophages and intestinal epithelial cells, participate in the secretion of various cytokines that regulate the inflammatory response of UC. Previous studies have shown that the levels of cytokines in UC.30, 44, and 45 are elevated, such as TNF-α, IFN-γ, IL-1β, IL-6, IL-17 and IL-21. In our study, we detected an increase in the expression levels of TNF-α, IL-1β, and IL-6 in DSS-induced mice. However, oral administration of DPs caused a decrease in the expression levels of the above-mentioned pro-inflammatory cytokines.

The spleen and thymus are important immune organs of the human body. It has been reported that the spleen index and thymus index of UC rats were significantly reduced, and the organ immunity was reduced46. Immune globulin is involved in humoral immunity, and elevated levels of IgA and IgG indicate that the humoral immune UC of rats is overactive, which may be a compensatory effect on the decreased cellular immune response. Studies have shown that the unbalanced secretion of anti-inflammatory and pro-inflammatory factors is an important part of intestinal inflammation in patients with UC. Increased levels of pro-inflammatory factors participate in cellular immunity and promote intestinal inflammation, which may cause further damage to intestinal tissues. In our study, the expression levels of pro-inflammatory factors TNF-α, IL-1β and IL-6 in the DPs administration group were significantly reduced, while the expression levels of anti-inflammatory factors such as IL-4 were relatively low. Increase. In addition, the concentration of IgA and IgG in the DPs group was significantly reduced. In short, oral DPs has a synergistic therapeutic effect on UC. It can regulate the body's immunity and play an anti-inflammatory effect at the same time. The research on this subject is expected to provide a new drug for the treatment of UC.

Developed a promising dual-targeted pill containing BBR and AM essential oils for the treatment of UC. BBR is loaded in the enteric layer for colon-targeted release, so it has a local effect. The AM volatile oil in gastric targeted pellets is prepared by extrusion spheronization and fluidized bed technology, with HPMC as the gastric soluble layer, which has rapid release and systemic effects. Both EPs and GPs after optimization have satisfactory physical and chemical properties. The bioavailability of BBR and AT-1 is much higher after granulation. In vivo targeting evaluation showed that EPs have strong acid resistance and satisfactory colon targeting ability. GPs decompose in the stomach within 5 minutes, allowing the drug to be completely released. The synergistic effect of DPs improves the clinical symptoms of UC, inhibits the expression of several inflammatory cytokines, and reduces the levels of IgA and IgG. This indicates that the anti-UC mechanism may not only be related to inhibiting inflammation, but also related to enhancing the body's immunity. Therefore, the developed oral dual-targeted drug delivery system can greatly promote the management of UC, and may provide a drug strategy for the coordinated treatment of gastrointestinal diseases.

This work was supported by the National Natural Science Foundation of China (81873011, 82074272), the Shanghai Science and Technology Commission (20S21900300, 21XD1403400), and the Shanghai Municipal Health and Family Planning Commission Excellent Talents Program (2018BR27). The author would like to thank Professor Ren Fuzheng, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, for his help in using the extrusion spheronization granulator and fluidized bed coating machine.

The author declares that there is no conflict of interest in this work.

1. Abraham C, Cho JH. Inflammatory bowel disease. N Engl J Med. 2009;361(21):2066-2078. doi:10.1056/NEJMra0804647

2. Sicilia B, Garcia-Lopez S, Gonzalez-Lama Y, Zabana Y, Hinojosa J, Gomollon F. GETECCU 2020 Guidelines for the treatment of ulcerative colitis developed by the GRADE method. Gastrointestinal heparin. 2020; 43 (Supplement 1): 1-57. doi:10.1016/j.gastrohep.2020.07.001

3. Gajendran M, Loganathan P, Jimenez G, etc. A comprehensive review and update of ulcerative colitis (). Desmond 2019;65(12):100851. doi:10.1016/j.disamonth.2019.02.004

4. Han W, Xie B, Li Y, Shi L, Wang H. Oral nano-therapeutic drugs for synergistic treatment of colitis-related colorectal cancer. Therapeutics. 2019; 9(24): 7458-7473. doi:10.7150/thno.38081

5. Grivennikov SI. Inflammation and colorectal cancer: colitis-related tumors. Semen immunopathology. 2013; 35: 229-244.

6. Danese S, Mantovani A. Inflammatory bowel disease and bowel cancer: a paradigm of Yin-Yang interaction between inflammation and cancer. Oncogene. 2010;29(23):3313–3323. doi:10.1038/onc.2010.109

7. Porter RJ, Kalla R, Ho GT. Ulcerative colitis: understand the latest developments in the pathogenesis of the disease. F1000Res. 2020; 9:294. doi:10.12688/f1000research.20805.1

8. Wang R, Zhou G, Wang M, Peng Y, Li X. Metabolism of Atractylodes macrocephala polysaccharide and its effect on intestinal flora. Compl Alt based on evidence. 2014; 2014: 1-7.

9. Zhang Li, Cao Nan, Wang Y, et al. Using andrographolide to improve oxazolone-induced ulcerative colitis in rats. molecular. 2020; 25(1):76. doi:10.3390/molecules25010076

10. Kringel DH, Antunes MD, Klein B, etc. Production, characterization and stability of orange or eucalyptus essential oil/β-cyclodextrin inclusion compound. J Food Science. 2017;82(11):2598-2605. doi:10.1111/1750-3841.13923

11. Tan S, Yu W, Lin Z, et al. Berberine improves intestinal mucosal barrier damage caused by peritoneal air exposure. Biopharmaceutical bull. 2015;38(1):122–126. doi:10.1248/bpb.b14-00643

12. Li YH, Xiao H, Hu D, et al. Berberine improves chronic recurrent dextran sodium sulfate-induced colitis in C57BL/6 mice by inhibiting Th17 response. Pharmaceutical Research 2016; 110: S1943242085.

13. Liao Zhong, Xie Yi, Zhou Yi, etc. Berberine improves colon damage while regulating the dysfunctional bacteria and function of rats with ulcerative colitis. Apply microbial biotechnology. 2020;104(4):1737-1749. doi:10.1007/s00253-019-10307-1

14. Leopold CS, Eikeler D. Basic coating polymer for colon-specific drug delivery in inflammatory bowel disease. Drug Dev Ind Pharm. 2000;26(12):1239–1246. doi:10.1081/DDC-100102305

15. Yang L, Chu JS, Fix JA. Colon-specific drug delivery: new methods and in vitro/in vivo evaluation. Int J Pharmaceut. 2002;235(1):1-15. doi:10.1016/S0378-5173(02)00004-2

16. Lee SH, Bajracharya R, Min JY, Han JW, Park BJ, Han HK. A strategic approach to colon-targeted drug delivery: an overview of recent developments. pharmaceutics. 2020; 12(1):68.

17. Wang Q, Wang G, Zhou J, Gao L, Cui Y. Colon-targeted oral drug delivery system based on icariin-loaded alginate-chitosan microspheres for the treatment of ulcerative colitis. Int J Pharmaceut. 2016;515(1-2):176-185. doi:10.1016/j.ijpharm.2016.10.002

18. Xiao B, Merlin D. Oral colon-specific treatment for inflammatory bowel disease. Expert Opin Drug Del. 2012;9(11):1393-1407. doi:10.1517/17425247.2012.730517

19. Pandey S, Swamy S, Gupta A, etc. Multi-response optimization of processing and formulation parameters of colon-targeted capecitabine pH-sensitive sustained-release pellets. J Microcapsules. 2018;35(3):259–271. doi:10.1080/02652048.2018.1465138

20. Sun X, Liu C, Omer AM, et al. pH-sensitive ZnO/carboxymethylcellulose/chitosan bio-nanocomposite beads for colon-specific release of 5-fluorouracil. Int J Biol Macromol. 2019; 128: 468-479. doi:10.1016/j.ijbiomac.2019.01.140

21. Moghimipour E, Rezaei M, Kouchak M, etc. The effect of coating layer and release media on the release profile of Eudragit FS 30D coated capsules: an in vitro and in vivo study. Drug Dev Ind Pharm. 2018;44(5):861–867. doi:10.1080/03639045.2017.1415927

22. Wan D, Zhao M, Zhang J, Luan L. Development and in vitro-in vivo evaluation of a new type of double-coated sustained-release loxoprofen pellets. pharmaceutics. 2019;11(6):260. doi:10.3390/pharmaceutics11060260

23. Segale L, Mannina P, Giovannelli L, Muschert S, Pattarino F. Formula and coating of alginate containing ranolazine and alginate-hydroxypropyl cellulose granules. J Pharm Sci United States. 2016;105(11):3351–3358. doi:10.1016/j.xphs.2016.08.001

24. Kim Y, Pradhan R, Paudel BK, Choi JY, Im HT, Kim JO. A fluidized bed coating machine was used to prepare and evaluate duloxetine hydrochloride enteric-coated sustained-release pellets. Arch Pharm Res. 2015;38(12):2163–2171. doi:10.1007/s12272-015-0590-y

25. Melegari C, Bertoni S, Genovesi A, etc. Ethylcellulose film coating of guaifenesin-loaded pellets: a comprehensive evaluation of the manufacturing process to prevent drug migration. Eur J Pharm Biopharm. 2016; 100:15-26. doi:10.1016/j.ejpb.2015.12.001

26. Belew S, Suleman S, Duguma M, etc. Develop the dissolution method of benzofluorenol and artemether in the quick-release fixed-dose artemether/benzfluorene amidine tablets. Maral J. 2020; 19(1):139. doi:10.1186/s12936-020-03209-5

27. Verstraete G, De Jaeghere W, Vercruysse J, etc. Using partially hydrolyzed polyvinyl alcohol to produce high drug loading sustained-release pellets by extrusion spheronization and coating: in vitro and in vivo evaluation. Int J Pharmaceut. 2017;517(1-2):88-95. doi:10.1016/j.ijpharm.2016.11.067

28. Novobilsky A, Hoglund J. In vivo imaging of parasite-infected small animals: a systematic review. Exp Parasitol. 2020;214:107905. doi:10.1016/j.exppara.2020.107905

29. Karrout Y, Dubuquoy L, Piveteau C, etc. In vivo efficacy of microbiota-sensitive coatings for colon targeting: a promising tool for IBD treatment. J control release. 2015;197:121-130. doi:10.1016/j.jconrel.2014.11.006

30. Pandurangan AK, Ismail S, Saadatdoust Z, Esa NM. Allicin can alleviate ulcerative colitis induced by dextran sodium sulfate (DSS-) in BALB/c mice. Oxid Med Cell Longev. 2015; 2015: 605208. doi:10.1155/2015/605208

31. Ferri D, Costero AM, Gavina P, etc. The efficacy of budesonide-loaded mesoporous silica particles in TNBS-induced colitis rats was blocked with a large azo derivative. Int J Pharm. 2019;561:93-101. doi:10.1016/j.ijpharm.2019.02.030

32. Shi C, Liang Y, Yang J, et al. MicroRNA-21 knockout improves the survival rate of DSS-induced fatal colitis by preventing inflammation and tissue damage. ProLogis one. 2013; 8(6): e66814. doi:10.1371/journal.pone.0066814

33. Nunes NS, Chandran P, Sundby M, etc. Therapeutic ultrasound reduces DSS-induced colitis through the cholinergic anti-inflammatory pathway. Biomedical Science. 2019; 45: 495-510. doi:10.1016/j.ebiom.2019.06.033

34. Li X, Xu Y, Zhang C, et al. Protective effect of bezoar on ulcerative colitis induced by sodium dextran sulfate in mice. Compl Alt based on evidence. 2015; 2015: Article ID 469506.doi:10.1155/2015/469506

35. Gao Wei, Wang C, Yu Li, et al. Chlorogenic acid reduces ulcerative colitis in mice induced by sodium dextran sulfate through the MAPK/ERK/JNK pathway. Biomedical Res Int. 2019; 2019: Article ID: 6769789.doi: 10.1155/2019/6769789

36. Ke Jie, Bian X, Liu Hai, etc. Edaravone reduces oxidative stress and intestinal cell apoptosis after burns by up-regulating the expression of miR-320. Moore Medicine. 2019; 25:54. doi:10.1186/s10020-019-0122-1

37. Kverka M, Rossmann P, Tlaskalova-Hogenova H, etc. The safety and effectiveness of the immunosuppressant 6-thioguanine in mouse models of acute and chronic colitis. Bmc gastrointestinal. 2011;11(1):47. doi:10.1186/1471-230X-11-47

38. Banerjee A, Bizzaro D, Burra P, etc. Umbilical cord mesenchymal stem cells modulate dextran sodium sulfate-induced acute colitis in immunodeficient mice. Stem cell spa. 2015; 6:79. doi:10.1186/s13287-015-0073-6

39. Neurath MF. Cytokines in inflammatory bowel disease. Nat Rev Immunology. 2014;14(5):329–342. doi:10.1038/nri3661

40. Kramar A, Turk S, Vrečer F. Statistical optimization of diclofenac sustained-release pellets coated with polymethacrylic acid film. Int J Pharmaceut. 2003;256(1–2):43–52. doi:10.1016/S0378-5173(03)00061-9

41. Wang X, Luo Z, Xiao Z. Preparation, characterization and thermal stability of β-cyclodextrin/soy lecithin inclusion compound. Carbohydrate polymer. 2014; 101: 1027-1032. doi:10.1016/j.carbpol.2013.10.042

42. Kumar A, Ekavali CK, Mukherjee M, Pottabathini R, Dhull DK, Dhull DK. Current knowledge and pharmacological profile of berberine: update. European Journal of Pharmacy. 2015; 761: 288-297. doi:10.1016/j.ejphar.2015.05.068

43. Lauber DT, Fulop A, Kovacs T, Szigeti K, Mathe D, Szijarto A. The latest level of imaging technology in laboratory animals. Laboratory animation. 2017; 51(5): 465–478. doi:10.1177/0023677217695852

44. Araki A, Nara H, Rahman M, etc. The role of interleukin 21 subtype in dextran sodium sulfate (DSS)-induced colitis. Cytokines. 2013;62(2):262–271. doi:10.1016/j.cyto.2013.03.006

45. Pervin M, Hasnat MA, Lim JH, etc. Blueberry (Vaccinium corymbosum) extract can prevent and treat DSS-induced ulcerative colitis by regulating antioxidant and inflammatory mediators. J Nutr Biochem. 2016; 28:103-113. doi:10.1016/j.jnutbio.2015.10.006

46. ​​Gong Yan, Niu Wei, Tang Yan, etc. The mucosal and immune damage in a mouse model of stress ulcerative colitis is aggravated. Exp Ther Med. 2019;17(3):2341–2348.

This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and include the Creative Commons Attribution-Non-commercial (unported, v3.0) license. By accessing the work, you hereby accept the terms. The use of the work for non-commercial purposes is permitted without any further permission from Dove Medical Press Limited, provided that the work has an appropriate attribution. For permission to use this work for commercial purposes, please refer to paragraphs 4.2 and 5 of our terms.

Contact Us• Privacy Policy• Associations and Partners• Testimonials• Terms and Conditions• Recommend this site• Top

Contact Us• Privacy Policy

© Copyright 2021 • Dove Press Ltd • Software development of maffey.com • Web design of Adhesion

The views expressed in all articles published here are those of specific authors and do not necessarily reflect the views of Dove Medical Press Ltd or any of its employees.

Dove Medical Press is part of Taylor & Francis Group, the academic publishing department of Informa PLC. Copyright 2017 Informa PLC. all rights reserved. This website is owned and operated by Informa PLC ("Informa"), and its registered office address is 5 Howick Place, London SW1P 1WG. Registered in England and Wales. Number 3099067. UK VAT group: GB 365 4626 36

In order to provide our website visitors and registered users with services that suit their personal preferences, we use cookies to analyze visitor traffic and personalize content. You can understand our use of cookies by reading our privacy policy. We also retain data about visitors and registered users for internal purposes and to share information with our business partners. By reading our privacy policy, you can understand which of your data we retain, how to process it, with whom to share it, and your right to delete data.

If you agree to our use of cookies and the content of our privacy policy, please click "Accept".