Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease of the colon with abdominal pain, diarrhea, and mucopurulent stools as the main symptoms. Its incidence is increasing worldwide, and traditional treatments have problems such as immunosuppression and metabolic disorders. In this article, the etiology and pathogenesis of ulcerative colitis are reviewed to clarify the targeted drugs of UC in the latest research. Our aim is to provide more ideas for the clinical treatment and new drug development of UC, mainly by analyzing and sorting out the relevant literature on PubMed, summarizing and finding that it is related to the main genetic, environmental, immune and other factors, and explaining its pathogenesis from the NF-κB pathway, PI3K/Akt signaling pathway, and JAK/STAT signaling pathway, and obtaining anti-TNF-α monoclonal antibodies, integrin antagonists, IL-12/IL-23 antagonists, novel UC-targeted drugs such as JAK inhibitors and SIP receptor agonists. We believe that rational selection of targeted drugs and formulation of the best dosing strategy under the comprehensive consideration of clinical evaluation is the best way to treat UC.
Keywords: pathogenesis, targeted drugs, UC
1. Introduction
Ulcerative colitis (UC) is a chronic idiopathic intestinal disease of unknown etiology that involves mainly the distal colon and rectum, often resulting in abdominal pain, diarrhea, and mucopurulent stools.[1] UC is characterized by recurrent and remitting mucosal inflammation, and therefore, if not treated properly, leads to recurrent UC symptoms, ongoing intestinal damage, and increased risk of cancer.[2] Nowadays, UC has become a global disease, and according to statistics, more than 1.5 and 2 million people in North America and Europe have suffered from UC in the past decade, and the incidence continues to increase, which means that the treatment of UC will pose a great challenge to health systems around the world.[3,4] However, the etiology of UC is not fully understood and is now mostly thought to be related to various factors such as genetics,[5] environment,[6] infection,[7] and immune regulation disorders,[8] while damage to the intestinal mucosal barrier is central to the pathogenesis of UC. The intestinal mucosal barrier includes a mechanical barrier, an immune barrier, a biological barrier, and a chemical barrier, and the destabilization of any of the intestinal barrier functions will lead to the destruction of the intestinal mucosal tissue, which will cause inflammatory lesions.[9,10]
Current UC treatment still aims to induce and maintain remission of symptoms, reduce the risk of complications, and improve quality of life as the main goals.[11] Individualized treatment plans are selected according to the severity of UC patients,[12] and 3 major classes of drugs, namely aminosalicylic acid agents, glucocorticoids, and immunosuppressive agents, are mostly used clinically.[2,6] Aminosalicylic acid agents can suppress the acute attack of UC and prolong the clinical remission period of UC patients, but in the subsequent treatment, aminosalicylic acid agents can cause gastrointestinal side effects such as loss of appetite, nausea, and vomiting, and have the possibility of autoimmune hemolysis, granulocytopenia, and nephrotoxicity.[13] Glucocorticoids can regulate immune function and reduce inflammatory cell infiltration, thus suppressing the inflammatory response and inducing remission of UC disease, but glucocorticoids do not prevent the recurrence of UC and can lead to an early recurrence of UC when the dose is rapidly reduced, and serious adverse effects such as osteoporosis, muscle weakness, Cushing’s syndrome, growth inhibition, and peptic ulcers can occur with long-term use, so long-term use is not recommended.[14–16] Immunosuppressants are mainly used in patients who are ineffective in aminosalicylate preparations or glucocorticoid therapy and toxic reactions to glucocorticoid therapy and long-term dependence on glucocorticoid use, but because immunosuppressants are cytotoxic, long-term use can cause serious adverse effects such as liver and kidney function impairment and bone marrow hematopoietic dysfunction, so they should also be selected with caution in clinical practice.[17,18] With the application of monoclonal antibodies and recombinant proteins targeting cytokines and cytokines in the clinical treatment of UC, it marks a milestone in progress in the targeted treatment of UC. In this paper, we review the pathogenesis of UC and the research progress of UC-related targeted drugs.
2. Pathogenesis of UC
2.1. Genetic factors
UC patients have a certain genetic susceptibility, about 12% of UC patients have a family history,[19] and there are racial differences, with the incidence rate of Caucasians being 3 times higher than that of blacks, and predominantly patients of Western and Northern European origin.[20,21] UC can be induced by deletion, mutation, or overlap of genes on chromosomes.[22] In addition, Luke Jostins[23] analyzed all genome-wide association studies and immunoassay data from 75,000 Europeans and found that 163 loci were associated with confirmed inflammatory bowel disease (IBD), with 23 specific loci strongly associated with UC, but the study was only obtained in a cohort of European ancestry, which may be ethnically biased. Subsequently, a cross-descent association study of IBD was conducted with data from 86,640 Europeans in genome-wide association studies and immunoassay data and 9846 non-Europeans in immunoassay data, which identified 38 new risk loci, most of which were shared across ethnic groups and explained 8.2% of the risk of UC disease, and differences in allele frequencies (NOD2) or effect sizes (TNFSF15 and ATG16L1) or combinations of these factors (IL23R and IRGM) can lead to genetic heterogeneity in different populations.[24,25] And by deeply resequencing 108 UC-associated candidate genes in the Korean population in search of novel variants, it was found that the genes BTNL2 and C5orf55 were associated with UC susceptibility or could be considered as susceptibility genes for the development of UC in Asian populations.[26] Frauke Degenhardt[27] identified through genetic association studies highly variable human Leukocyte antigen regions have been identified as important in UC susceptibility in different ethnic groups, while DRB1*01:03 is mainly found in Western European descendants and rare in non-Caucasians.
2.2. Environmental factors
Traditionally, UC is regarded as a Western disease, and the incidence of UC is concentrated in Western developed countries, but according to epidemiological studies, the prevalence of UC has increased nearly 20-fold in some Asian regions in the past 4 decades.[28,29] This also suggests that environmental factors play a crucial role in the development of UC.[30] The current study found that the environmental factors affecting the development of UC include: diet,[31] smoking,[32] and the use of antibiotics.[33] With the improvement of people’s living conditions, the intake of high oil and fat, protein, and dairy products has increased, and the intake of dietary fiber has decreased,[34] in which a large amount of intake of trans-unsaturated fat makes the risk of UC disease increased.[35] Dietary fiber itself induces insulin resistance, regulates fatty acid imbalance caused by intestinal epithelial inflammation, alters intestinal permeability, promotes sulfate production, and alters the intestinal microbiota by promoting the growth environment of sulfate-reducing bacteria.[36] Meanwhile, dietary fiber is metabolized into short-chain fatty acids by fermentation, inhibits the transcription of pro-inflammatory factors,[37] and scavenges oxygen free radicals,[38] so a high dietary fiber diet plays a preventive role in UC.
Smoking reduces the recurrence rate of UC by a mechanism related to the mediation of CO,[39] and long-term exposure to CO can induce the production of heme oxidase-1, which represents an endogenous host defense mechanism with endogenous upregulation and effective cytoprotection against various pro-inflammatory stimuli and has powerful anti-inflammatory and antioxidant capacities.[40,41] Studies have shown that the use of antibiotics in childhood can prevent the onset of UC,[42] and patients with UC have immune dysfunction and have been attacked by bacteria, and the application of antibiotics has a certain therapeutic effect on UC-related intestinal infections,[43] but the abuse of antibiotics can lead to changes in the intestinal microflora and cause immune system disorders to induce the onset of UC, so the rational use of antibiotics should be advocated in clinical practice.
2.3. Immune factors
The immune system plays an important role in the development of UC, and disruption of normal immune regulation in the gut or abnormal immune responses are important aspects of UC pathogenesis,[44] which can be caused by pathological interactions between the microflora in the gut and mucosal immunity in genetically susceptible individuals.[45] Neutrophils, macrophages, mast cells, T and B lymphocytes, and natural killer cells are involved in a continuous chronic immune process, and the loss of immune tolerance leads to inflammation due to the increased release of antibodies, cytokines, and pro-inflammatory mediators from these effector cells, which stimulate the proliferation of antigen-specific effectors, thereby triggering the adaptive immune system and leading to local and systemic inflammation.[8] Th cells are also important effector cells in the intestinal immune response process, and in the normal state of the organism, Th1 and Th2 are in dynamic balance,[46] while when external antigens invade the organism, Th1 cells are activated and Th2 cell function decreases accordingly, while Th2 cells expand in large numbers to stimulate increased secretion of cytokines and suppress Th1, which in turn suppresses the series of immune The Th2 cell function will be reduced, while the Th2 cell expansion will stimulate an increase in cytokine secretion and suppress Th1, which in turn will suppress a series of immune effects mediated by this cell.[47] In patients with active UC, there is an imbalance between the constraints of Th1/Th2 cells in their colonic tissues, and inflammatory cells are heavily and continuously activated, aggregating and infiltrating the lining of the colon, where IL-1, IL-1 receptors, IL-6, IL-8, and TNF-γ are upregulated in the patients’ colonic tissues, with the most significant increase in TNF-γ and the IL-1/IL-1 receptor ratio.[48] In contrast, for the exudative inflammatory response and epithelial ulcer-associated intestinal wall edema seen in UC patients, which is associated with an early cytokine IL-4 response, the IL-4 response decreases and is replaced by the Th2 cytokine IL-13 when the patient has a long duration of inflammation.[49] It has also been suggested that innate lymphoid-like cells (ILCs), which are central to innate immunity, are at the surface of the intestinal mucosa, maintain intestinal homeostasis and play a role in fighting intestinal inflammation.[50]
2.4. Pathway mechanism
2.4.1. NF-κB pathway.
NF-κB is an important intracellular nuclear transcription factor involved in the inflammatory response, immune response, regulation of apoptosis, and stress response of the body. In unstimulated cells, most NF-κB dimers are inactivated and retained in the cytoplasm by the binding of small inhibitory molecules from the family of NF-κB inhibitory proteins (inhibition of NF-κB).[51] The NF-κB pathway is regulated by tumor necrosis factor (TNF)-α, interleukin (IL), etc. IKK (IKK) activation phosphorylates inhibition of NF-κB, degrades the proteasome, activates and releases NF-κB-related factor complexes into the nucleus, and activates the expression of target genes involved in cell proliferation and apoptosis, initiating downstream signaling pathways.[52] It was found that the expression of intestinal mucosal inflammation was increased in both mouse models of ulcerative colitis and NF-κB P-p65 was abundant in colonic tissues, and the loss of regulatory factors on the NF-κB signaling pathway leads to pathological changes in the intestinal mucosa of UC, and the inflammation is further exacerbated by the stimulation of inflammatory factors that cause NF-κB to be activated, so the expression of NF-κB in colonic tissues also responds to the UC severity of the disease.[53,54] HIF-1α, COX-2, IL-6, IL-1β, and TNF-α are influenced by NF-κB and induce and regulate the development of body immunity and inflammation through different pathways.[55] HIF-1α can be produced in large amounts in hypoxic environments, regulates the neovascularization, and participates in several response processes such as inflammation,[56] and UC The large number of immune cells aggregating at the site of intestinal inflammation creates a local hypoxic environment where HIF-1α is activated, which in turn promotes the synthesis of IL-10 and COX-2 and enhances the inflammatory response.[57,58]COX-2, the rate-limiting enzyme that catalyzes the synthesis of prostaglandins from arachidonic acid, is highly expressed in the UC mucosal epithelium and crypt, which can lead to an increase in prostaglandin E2,[59] COX-2 expression is also regulated by the NF-κB pathway, and its expression is enhanced under conditions of tissue injury and inflammation, resulting in increased prostaglandins in UC patients, causing vasodilation, increased permeability, mucosal congestion and edema, and symptoms of abdominal pain and diarrhea.[60] TNF-α, a proinflammatory factor mediating the pathogenesis of UC, is first seen in intestinal inflammation and promotes the C-reactive protein release, amplifying the inflammatory response, and can also affect vascular microcirculation by inducing apoptosis of intestinal epithelial cells, thereby inhibiting the repair of ulcerated surfaces in the intestine.[61] IL-6 is at the center of the inflammatory factor burst along with TNF-α, and UC colonic tissue injury is accompanied by overexpression of these inflammatory factors, while IL-10 is an inflammatory suppressor with a significant inhibitory effect on inflammation in UC.[62]
2.4.2. PI3K/Akt signaling pathway.
The PI3K/Akt signaling pathway is also involved in the regulation and release of inflammatory factors, and it can indirectly activate the transcription factor NF-κB through phosphorylated IKK, thus interconnecting with the NF-κB pathway to promote enhanced expression and secretion of inflammatory factors, leading to damage of the colonic mucosa.[63] PI3K is a lipid second messenger of intracellular signaling,[64] which consists of a catalytic structural domain P110 and a regulatory structural domain P85.[65] Akt is a serine/threonine protein kinase encoded by the proto-oncogene c-akt and is a direct target protein of PI3K. PI3K can signal through tyrosine kinase-linked receptors or G protein-linked receptors,[66] causing conformational changes in Akt, with simultaneous phosphorylation of the Ser473 site and Thr308 site, activating or inhibiting downstream signaling molecules, thereby regulating cell differentiation, proliferation, and apoptosis. TLR4 is a pattern recognition receptor with an important role in intestinal intrinsic immunity, recognizing lipopolysaccharide as a protein. TLR4 recognizes lipopolysaccharide and mediates transmembrane signaling,[67] and is usually overexpressed in the colonic epithelium of UC patients. TLR4 induces intestinal inflammation mainly by recognizing pathogen surface-associated molecules to induce activation of downstream signaling pathways, which in turn activates the expression of inflammatory factors.[68] TLR4 signaling activates the PI3K/Akt signaling pathway and causes downstream mTOR activation, and it was found[69] that mTOR was aberrantly activated in colonic tissues of UC patients and DSS-induced UC mice, and mTOR inhibitors had good inhibitory effects on DSS-induced UC. PlGF (placental growth factor), as an angiogenic protein,[70] is closely related to pathological angiogenesis and inflammatory responses in UC patients, and studies have confirmed that PlGF-induced angiogenic responses in UC patients are dependent on the migration and sprouting of HIMECs (human intestinal microvascular endothelial cells) in the PI3K/Akt signaling pathway,[71] and that serum PlGF concentrations were significantly higher in UC patients compared to healthy controls,[72,73] with increased vascularity occurring only in areas containing active inflammatory infiltrates and concomitant increased histopathology.[74]
In addition, patients with long-term chronic recurrent UC are at increased risk of carcinogenesis, and the PI3K/Akt signaling pathway is one of the pathways of inflammatory carcinogenesis and a key link in the development of UC to CRC (colorectal cancer).[75] It has been suggested that IL23R variants in IBD may act as a protective variant or contribute to inflammation, and IL23R polymorphisms may also increase the risk of CRC.[76] Razali et al[77] detected 13 cytokine-induced somatic mutations in PI3K-related genes in long-standing UC, CAC, and CRC patients and found that most of these variants appeared in the IL23R, IL12Rß1, and IL12Rß2 genes, and the IL23R variant rs10889677, as a possible mutation,[78] helps to gain insight into how the cytokine-induced PI3K pathway induces UC to develop into CRC, and it may be a breakthrough in the treatment of UC.
2.4.3. JAK/STAT signaling pathway.
The JAK/STAT signaling pathway uses second messengers to transmit extracellular information to the nucleus, influencing target gene expression and cellular responses, coordinating intracellular signaling of more than 50 different ligands, and consisting of 3 parts: tyrosine-associated receptors, JAK/STAT, and the intracellular segment of tyrosine kinase-coupled tyrosine-associated receptors. It is believed that abnormal JAK/STAT signaling pathway can lead to abnormal t-cell differentiation as well as defective t-cell regulatory activity, which is an important mechanism in the pathogenesis of UC.[79] JAK family (JAK1, JAK2, JAK3, TYK2) are non-covalently bound to cytokine receptors, while STATs, as substrates of JAK, can couple to tyrosine phosphorylation signaling pathway.[80] Depending on the ligand and receptor, different combinations of JAK and STATs are highly specific activated to exert specific transcriptional regulation and mediate a variety of biological processes such as apoptosis, proliferation, differentiation, and migration of cells.[81] The expression of cytokines (IL-6, IL-10, IFN-γ, IL-12, and IL-23) required for the maintenance of homeostasis in intestinal immune cells and stromal cells are mediated through the JAK/STAT signaling pathway,[82] where IL-6 to gp130 receptors activate STAT3 through JAK1, JAK2, and TYK2,[83] and IL-12 can stimulate JAK2 and TYK2 activity, leading mainly to STAT4 homodimer phosphorylation, and IL-23 activates mainly STAT3.[84] Activation of STAT3 promotes pathogenic differentiation of Th17 cells and suppression of regulatory T cells, accelerating UC inflammation,[85] and it was found that inhibition of STAT3 phosphorylation restores DSS-induced UC mice cellular homeostasis of Treg/Th17 in colonic tissue and alleviates the clinical symptoms of UC.[86] Whereas JAK2 and TYK2-dependent STAT4 phosphorylation are only present in the transduction response of certain specific signals (e.g., IL-12R and IL-23R) in the gp130 receptor family,[87] STAT4 phosphorylation is associated with Th1 activity and regulates IFN-γ expression, accelerating intestinal mucosal injury in UC, and studies have also confirmed that STAT4 levels in the cytoplasm of UC patients and increased levels of STAT4 phosphorylation in the nuclei of mucosal cells.[88] And it was found that polymorphisms of single nucleotides containing JAK2, TYK2, STAT1, STAT3, and STAT4 genes increase the risk of developing UC.[23]
IL-27 and IL-35 are inhibitory cytokines that signal through JAK1 and JAK2 and play an important role in the immune regulation of UC,[89] IL-27 induces the expression of IL-10 and also inhibits the expression of Th1, Th2, and Th17 cells,[90] IL-35 downregulates the levels of TNF-α, IFN-γ pro-inflammatory cytokines and increase the response of Th1 and Th17 to exert anti-inflammatory effects to improve UC.[91] Because the JAK/STAT pathway is such that JAKs can be linked to multiple STATs and STATs are the final effectors of signaling, studies explaining UC from a JAK family perspective are more limited. In previous studies, we can know that JAK1 and JAK3 are mainly expressed in T and B cells, JAK1-deficient mice die prenatally or perinatally,[92] JAK2-deficient mice are mutationally lethal due to lack of erythropoiesis,[93] and JAK3 knockout mice develop severe combined immunity that affects T and B cell development, resulting in intestinal epithelial cell differentiation defects and impaired intestinal barrier function, thereby increasing susceptibility to UC.[94]
3. UC molecular targeting drugs
With advances in drug development, the drug options for UC have become more diverse. The resulting targeted therapeutic agents include anti-TNF-α monoclonal antibodies, integrin antagonists, IL-12/IL-23 antagonists, JAK inhibitors, and SIP receptor agonists (Table 1). This article now introduces the mechanism of action of these drugs to bring thoughts to clinical UC drug selection.
Table 1.
Targeted drugs for UC and characteristics of drug effects.
| Type | Drugs | Characteristics of drug action |
|---|---|---|
| Anti-TNF-α monoclonal antibody | Infliximab (IFX) | Human-mouse chimeric IgG1 monoclonal antibody, targeting TNF-α, was administered intravenously. |
| Adalimumab (ADA) | Fully human, recombinant IgG1 monoclonal antibody, targeting TNF-α, is better tolerated, especially in patients who have lost response to or are intolerant to IFX monoclonal antibody. | |
| Golimumab (GLM) | The fully humanized monoclonal antibody, targeting Tnf-α, has a much higher affinity for TNF than IFX and ADA. GLM is suitable for IFX - and ADA-refractory UC patients and can be injected subcutaneously. | |
| Anti-integrin monoclonal antibodies | Vedolizumab | It specifically binds to α4β7 integrin and blocks the binding of α4β7 to MAdCAM-1. |
| Etrolizumab | Bidirectional inhibition of α4β7 integrin and MAdCAM-1 and αEβ7 integrin and β7 subunit of E-cadherin. | |
| Natalizumab | Targeted blocking of the binding of α4β1 integrin to VCAM-1 and the binding of α4β7 integrin to MAdCAM-1 has the risk of PML. | |
| IL-12/IL-23 antagonists | Ustekinumab | The fully human IgG1 monoclonal antibody targets the p40 subunit shared by both IL-12 and IL-23, thereby inhibiting IL-12 signaling and further activation of Th1 subsets of T cells, and blocking IL-23-mediated immune response and downstream activation of Th17 subsets of T cells. |
| Risankizumab | Novel biological agents selectively target the p19 subunit of IL-23 and produce specific inhibition of IL-23 without interfering with the host immune response involved in IL-12. Phase II and III clinical trials are ongoing | |
| Brazikumab | ||
| Guselkumab | ||
| JAK inhibitor | Tofacitinib | Inhibition of JAK1 and JAK3 reduces the activity of JAK signaling. |
| Filgotinib | Inhibition of JAK1. | |
| Upadacitinib | There was a stronger inhibitory effect on JAK1. | |
| SIP receptor agonists | Ozanimod | Selective binding of SIPR1 and SIPR5 |
| Etrasimod | Full agonist of S1PR1 and partial agonist of S1PR4 and S1PR5. |
MAdCAM-1 = mucusmembrane address element cell adhesion molecule 1, PML = progressive multifocal leukoencephalopathy, TNF = tumor necrosis factor, UC = ulcerative colitis, VCAM-1 = vascular cell adhesion molecules.
3.1. Anti-TNF-α monoclonal antibody
TNF-α is produced by a subpopulation of immune cells in the gut of UC patients, associated with Th17 differentiation and involved in the regulation of innate and adaptive immunity. TNF-α mainly induces pro-inflammatory factors, and activates macrophages and T cells, causing epithelial cell damage and intestinal mucosal destruction.[95] The anti-TNF-α monoclonal antibodies currently approved for UC treatment are mainly infliximab (IFX), adalimumab (ADA), and golimumab. Infliximab, as a TNF-α targeted drug, is a human-murine chimera IgG1 monoclonal antibody, which plays an important role in the treatment of UC, and the 2020 American Gastroenterology Association guidelines recommend it as a first-line drug to save acute severe UC.[96] Animal experiments[97]have demonstrated that anti-TNF-α treatment improves UC activity, which may be related to the immune response triggered by IFX treatment and indirectly affects the intestinal flora. A prospective clinical study[98,99]showed that maintenance IFX treatment significantly reduced C-reactive protein and fecal calprotectin levels in UC patients and reduced the surgery rate from 27% to 11%.
ADA, a fully human, recombinant monoclonal IgG1 antibody injected subcutaneously, binds soluble and membrane-bound TNF-α, causing cell-mediated dependent cytotoxicity, repairing complement, and inducing T-cell apoptosis, thus acting as a therapeutic agent for UC.[100] In a real-world study,[101] adalimumab achieved clinical remission and mucosal healing in nearly one-third of patients with moderately to severely active UC within 52 weeks. And adalimumab was better tolerated, especially in patients who lost response or were intolerant to IFX monotherapy.[102] The rate of clinical remission at week 8 was 16.5% in the ADA group and 9.3% in the placebo group (P < .05). The rate of mucosal healing at week 8 was 41.1% in the ADA group and 31.7% in the placebo group (P < .05).[103]
Godamumab (GDM), the third TNF-α drug approved for the treatment of UC, is synthesized from TNF-immunized transgenic mice. Studies have shown that GDM has a much higher affinity for TNF than IFX and ADA, and it is also superior to IFX and ADA in terms of conformational stability and inhibition of tumor necrosis factor-induced cytotoxicity.[104] In a multicenter, randomized, double-blind, controlled study, the proportion of patients in clinical remission and mucosal healing at weeks 30 and 54 was higher in patients receiving 100 mg GDM (27.8% and 42.4%) than in those receiving placebo (15.6% and 26.6%; P = .004 and P = .002 respectively).[105]
3.2. Anti-integrin monoclonal antibodies
There are still patients who fail to achieve clinical improvement after treatment with TNF-α monoclonal antibodies or small molecule immunomodulators, which is related to patients’ insensitivity to the above drugs or drug off-target. UC patients have a large number of leukocyte aggregates in the intestinal tract after lesions occur, resulting in immune dysregulation in the intestine. Integrins are transmembrane receptors on the surface of leukocytes and consist of α and β subunits. α4β1 integrin mediates vascular cell adhesion molecules (VCAM-1) on gastrointestinal endothelial cells; Binding of α4β7 integrin to mucusmembrane address element cell adhesion molecule 1 (MAdCAM-1) of intestinal endothelial cells; αEβ7 integrin binds to E-calmodulin of mucosal epithelial cells,[106] thus binding to memory T lymphocytes and blocking inflammation production, thus also being one of the ideal targets for UC therapy.[107]
Vedolizumab (VDZ) is a humanized IgG1 monoclonal antibody that selectively blocks the binding of α4β7 integrin to MAdCAM-1 of intestinal endothelial cells, reducing leukocyte adhesion to the intestinal epithelium and has high intestinal selectivity, which can better promote intestinal mucosal repair and thus effectively induce symptom relief in UC patients.[108] In a 52-week-long clinical study of patients with active UC,[109] 374 patients were given intravenous placebo at weeks 0 and 2, and 521 patients were treated with intravenous VDZ at weeks 0 and 2. During the DAI assessment at week 6, it was found that the response rate was 47.1% in the VDZ group and 25.5% in the placebo group (difference with adjustment for stratification factors, 21.7 percentage points; 95% confidence interval [CI], 11.6–31.7; P < .001). VDZ was significantly superior to placebo as an induction and maintenance treatment for UC.
Etrolizumab monoclonal antibody inhibits bidirectionally α4β7 integrin with MAdCAM-1 and αEβ7 integrin with the β7 subunit on epithelial calcium adhesion protein.[110] In a phase III trial evaluating the efficacy and safety of etrolizumab in maintaining moderate to severely active UC remission, 31% of patients in the etrolizumab group achieved endoscopic remission at week 62 compared with 17% in the placebo group (P = .029 < 0.05) and was well tolerated in this population.[111]
In addition, Natalizumab, which targets the binding of α4β1 integrin to VCAM-1 and α4β7 integrin to MAdCAM-1, has a high clinical response rate, but the risk of progressive multifocal leukoencephalopathy,[112] a serious and fatal adverse effect, has led to the gradual withdrawal of natalizumab from the market.
3.3. IL-12/IL-23 antagonists
IL-12 (composed of p40 and p35 subunits) and IL-23 (composed of p40 and p19 subunits) are pro-inflammatory factors produced by intestinal pathogens and are essential for the differentiation of CD4 naive cells.[113] Ustekinumab (UST) is a fully human IgG-type monoclonal antibody that inhibits IL-12 signaling and further activation of the Th1 subpopulation of T cells by binding to the p40 subunit shared by IL-12 and IL-23, and, at the same time, blocks IL-23-mediated immune responses and activation of the Th17 subpopulation of downstream T cells. One study evaluated the efficacy of UST as induction and maintenance therapy in patients with moderate to severe UC,[114] the remission rate was 38.4% when UST was used every 12 weeks and 43.8% when UST was used every 8 weeks. The effective rate was significantly higher than that of the placebo group (24.0%) (P = .002 and P < .001, respectively). In a real-world prospective study[115] involving 95 UC patients, which was designed to evaluate the efficacy and safety of UST in the real world, 53% of UC patients responded at week 16.39% and 33% achieved remission at week 24 and week 52, respectively. In addition, the proportion of patients with elevated C-reactive protein and severe endoscopic activity in patients who achieved remission at week 16 was significantly lower than that in patients who did not achieve remission (52% vs 75%, P < .05, and 50% vs 74%, P < .05, respectively), suggesting that UST is effective in both short-and long-term treatment in real life.
Risankizumab, Brazikumab, and Guselkumab, as novel biologics, selectively target the p19 subunit of IL-23 and do not interfere with the host immune response in which IL-12 is involved, thus theoretically allowing for better efficacy. Phase II and Phase III clinical trials of these drugs are ongoing, and further studies are needed regarding their clinical efficacy, safety, advantages over other biologics, and optimal dosing strategies.[116]
3.4. JAK inhibitor
The JAK-STAT signaling pathway is involved in the regulation of innate and adaptive immunity and hematopoiesis, and it enables the production of cytokines by helper T cells and induces the onset of inflammatory responses in UC.[117] Tofacitinib, a nonselective inhibitor of JAK, is the most widely used clinically as a non-immunogenic oral small molecule drug that mainly inhibits JAK1 and JAK3, and is now approved by the FDA and EMA for the treatment of moderate to severe UC.[118] Animal and human organoid experiments[119,120] found that Tofacitinib may exert clinical effects by reducing the activity of the JAK signaling pathway, disrupting the normalization of tight junction protein expression, correcting the function of PTPN2 in macrophages or intestinal epithelial cells, and restoring the integrity of the epithelial barrier. In the subsequent phase III trial to explore its efficacy, it was found that patients with moderate to severe UC who received Tofacitinib clinically had a remission rate of 16.6% at 8 weeks of induction therapy, which was significantly higher than that of 3.6% in the placebo group (P < .001). In the trial with 52-week maintenance therapy, 34.3% of the patients had remission at week 5, as compared with 11.1% of those in the placebo group (P < .001). A real-world study[121] that included 260 UC patients found that 15.7% of UC patients experienced adverse reactions with Tofacitinib, with infection being the most common, 5.8% of the cohort reported serious adverse events, and 4.2% of the cohort required discontinuation of treatment. An analysis of clinical data from 1157 UC patients found[122] that patients treated with Tofacitinib experienced reversible elevated lipid levels during dosing and that lipid levels were negatively correlated with high-sensitivity C-reactive protein levels, but cardiovascular adverse events occurred less frequently and were not related to the dose of Tofacitinib used.
Filgotinib, a second-generation selective JAK1 inhibitor, has been approved for UC treatment in Europe and Japan.[123] In a randomized, double-blind, phase 2b/3 study,[124] 26.1% of UC patients receiving 200mgFilgotinib achieved clinical remission at week 10, which was significantly higher than the remission rate of 15.3% in the placebo group (difference rate, 10.8%; 95% CI: 2.1–19.5, P = .0157). Regardless of experience with biologics, was effective in inducing and maintaining clinical remission, and Filgotinib was well tolerated, with a similar incidence of adverse events to the placebo group, with nasopharyngitis and headache being the most common.
Upadacitinib has a stronger inhibitory effect on JAK1. In a phase 2b trial,[125] upadacitinib was used as induction therapy in patients with moderately to severely active UC, and at week 8, clinical responses were achieved in 5.19%, 6.7%, 5.15%, and 30.45% of patients receiving 8.5, 14, 3, and 13 mg, respectively. In contrast, patients who received a placebo did not have clinical remission (P = .052, P = .013, P = .011, and P = .002) and had the best efficacy at 45 mg of upadacitinib once daily. However, the incidence of herpes zoster was higher in patients using upadacitinib, and the study was not sufficient to assess the safety of upadacitinib due to the limitations of the study in terms of sample size, dose range, and duration of use. A subsequent multicenter, phase 3 study[126] showed that the clinical remission rate at week 8 was 26% in UC patients receiving induction therapy with upadacitinib at a dose of 45 mg once daily, which was higher than the remission rate of 5% in the placebo group (P < .0001). The rate of clinical remission at week 52 was significantly higher with maintenance upadacitinib at a dose of 30 mg (52%) than with placebo (12%) (P < .0001), and induction and maintenance therapy was well tolerated, and no new safety risks were observed.
3.5. SIP receptor agonists
SIP is a membrane-derived lysophospholipid signaling molecule that specifically binds G protein-coupled receptors (SIPR1-SIPR5), and SIP receptor agonists target lymphocyte recirculation and block lymphocyte migration from lymphoid organs to the intestine along a chemotactic gradient, thereby effectively reducing intestinal inflammation,[127] leading to the therapeutic drugs Ozanimod and Etrasimod. Ozanimod can optionally combine SIPR1 and SIPR5, and a phase 3, multicenter, randomized, double-blind controlled trial[128] found that the clinical response rate of induction therapy at week 10 of Ozanimod (18.4%) was significantly higher than that of placebo group (6.0%) (P < .001), and the response rate of maintenance therapy at week 52 (37.0%) was significantly higher than that of the control group (18.5%) (P < .001). Endoscopic changes and mucosal healing were significantly improved, and the incidence of adverse effects was similar in the Ozanimod group and the placebo group, with elevated hepatic aminotransferases being the most common.
Etrasimod is a full agonist of S1PR1 and a partial agonist of S1PR4 and S1PR5, which controls the level of immune cells in the blood and reduces inflammation in the lining of the colon.[129] In the phase 2 randomized trial of patients with moderately to severely active UC,[130] 41.8% of UC patients in the Etrasimod2mg group had endoscopic improvement compared with 17.8% of those in the placebo group (P = .003). The primary and secondary clinical endpoints were met with Etrasimod 2 mg at week 12, with very few patients experiencing atrioventricular block. Further phase III trials of Etrasimod need to be conducted to clarify the safety and optimal treatment regimen of Etrasimod in UC.
4. Conclusion
The pathogenesis of UC is still unclear, and it is mainly related to genetic, environmental, immune, and related signaling pathway overexpression and activation, etc. Moreover, the pathogenesis of UC is not caused by a single factor, but by the interaction of multiple factors. Therefore, an in-depth investigation of the pathogenesis of UC and the analysis of the intrinsic connection of each factor can help to develop effective and safe new therapeutic drugs for UC. In the past decades, targeted drugs represented by TNF-α monoclonal antibodies have achieved certain therapeutic effects. With continuous research on the immune system, new therapeutic targets have been discovered, and drugs such as integrin antagonists, IL-12/IL-23 antagonists, and JAK inhibitors have shown better therapeutic effects. Novel therapeutic tools based on the pathogenesis of UC can be used as a breakthrough in the treatment of UC. Targeted therapies for UC have good clinical application prospects, but these drugs still have potential adverse effects, and real trials and long-term clinical data are needed to improve their safety and optimal dosing strategies in the treatment of UC.
Author contributions
Conceptualization: Xiaoyan Wang.
Data curation: Meitong Guo.
Writing – original draft: Meitong Guo.
Writing – review & editing: Xiaoyan Wang.
Abbreviations:
- ADA
- adalimumab
- CRC
- colorectal cancer
- GDM
- Godamumab
- IBD
- inflammatory bowel disease
- IFX
- infliximab
- IKK
- inhibition of NF-κBkinase
- IL
- interleukin
- ILCs
- innate lymphoid-like cells
- MAdCAM-1
- mucusmembrane address element cell adhesion molecule 1
- PlGF
- placental growth factor
- TNF
- tumor necrosis factor
- UC
- ulcerative colitis
- UST
- Ustekinumab
- VCAM-1
- vascular cell adhesion molecules
- VDZ
- vedolizumab
The authors have no funding and conflicts of interest to disclose.
The datasets generated during and/or analyzed during the current study are publicly available.
How to cite this article: Guo M, Wang X. Pathological mechanism and targeted drugs of ulcerative colitis: A review. Medicine 2023;102:37(e35020).
References
- [1].Ordás I, Eckmann L, Talamini M, et al. Ulcerative colitis. Lancet. 2012;380:1606–19. [DOI] [PubMed] [Google Scholar]
- [2].Du L, Ha C. Epidemiology and pathogenesis of ulcerative colitis. Gastroenterol Clin North Am. 2020;49:643–54. [DOI] [PubMed] [Google Scholar]
- [3].Ng SC, Shi HY, Hamidi N, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies [published correction appears in Lancet. 2020 Oct 3;396(10256):e56]. Lancet. 2017;390:2769–78. [DOI] [PubMed] [Google Scholar]
- [4].Kaplan GG, Ng SC. Understanding and preventing the global increase of inflammatory bowel disease [published correction appears in Gastroenterology. 2017 Jun;152(8):2084]. Gastroenterology. 2017;152:313–321.e2. [DOI] [PubMed] [Google Scholar]
- [5].Annese V. Genetics and epigenetics of IBD. Pharmacol Res. 2020;159:104892. [DOI] [PubMed] [Google Scholar]
- [6].Hodson R. Inflammatory bowel disease. Nature. 2016;540:S97. [DOI] [PubMed] [Google Scholar]
- [7].Mourad FH, Hashash JG, Kariyawasam VC, et al. Ulcerative colitis and cytomegalovirus infection: from A to Z. J Crohns Colitis. 2020;14:1162–71. [DOI] [PubMed] [Google Scholar]
- [8].Tatiya-Aphiradee N, Chatuphonprasert W, Jarukamjorn K. Immune response and inflammatory pathway of ulcerative colitis. J Basic Clin Physiol Pharmacol. 2018;30:1–10. [DOI] [PubMed] [Google Scholar]
- [9].Wang K, Wu LY, Dou CZ, et al. Research advance in intestinal mucosal barrier and pathogenesis of Crohn’s disease. Gastroenterol Res Pract. 2016;2016:9686238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Antoni L, Nuding S, Wehkamp J, et al. Intestinal barrier in inflammatory bowel disease. World J Gastroenterol. 2014;20:1165–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Feuerstein JD, Cheifetz AS. Ulcerative colitis: epidemiology, diagnosis, and management. Mayo Clin Proc. 2014;89:1553–63. [DOI] [PubMed] [Google Scholar]
- [12].Eisenstein M. Ulcerative colitis: towards remission. Nature. 2018;563:S33. [DOI] [PubMed] [Google Scholar]
- [13].Linares V, Alonso V, Domingo JL. Oxidative stress as a mechanism underlying sulfasalazine-induced toxicity. Expert Opin Drug Saf. 2011;10:253–63. [DOI] [PubMed] [Google Scholar]
- [14].Kucharzik T, Koletzko S, Kannengiesser K, et al. Ulcerative colitis-diagnostic and therapeutic algorithms. Dtsch Arztebl Int. 2020;117:564–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Bruscoli S, Febo M, Riccardi C, et al. Glucocorticoid therapy in inflammatory bowel disease: mechanisms and clinical practice. Front Immunol. 2021;12:691480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Oray M, Abu Samra K, Ebrahimiadib N, et al. Long-term side effects of glucocorticoids. Expert Opin Drug Saf. 2016;15:457–65. [DOI] [PubMed] [Google Scholar]
- [17].Nielsen OH, Ainsworth MA, Steenholdt C. Methotrexate for inflammatory bowel disease: time for reconsideration. Expert Rev Gastroenterol Hepatol. 2019;13:407–9. [DOI] [PubMed] [Google Scholar]
- [18].Nielsen OH, Coskun M, Steenholdt C, et al. The role and advances of immunomodulator therapy for inflammatory bowel disease. Expert Rev Gastroenterol Hepatol. 2015;9:177–89. [DOI] [PubMed] [Google Scholar]
- [19].Childers RE, Eluri S, Vazquez C, et al. Family history of inflammatory bowel disease among patients with ulcerative colitis: a systematic review and meta-analysis. J Crohns Colitis. 2014;8:1480–97. [DOI] [PubMed] [Google Scholar]
- [20].Barnes EL, Loftus EV, Jr, Kappelman MD. Effects of race and ethnicity on diagnosis and management of inflammatory bowel diseases. Gastroenterology. 2021;160:677–89. [DOI] [PubMed] [Google Scholar]
- [21].Misra R, Faiz O, Munkholm P, et al. Epidemiology of inflammatory bowel disease in racial and ethnic migrant groups. World J Gastroenterol. 2018;24:424–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Saadati HR, Wittig M, Helbig I, et al. Genome-wide rare copy number variation screening in ulcerative colitis identifies potential susceptibility loci. BMC Med Genet. 2016;17:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Jostins L, Ripke S, Weersma RK, et al.; International IBD Genetics Consortium (IIBDGC). Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491:119–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Liu JZ, van Sommeren S, Huang H, et al.; International Multiple Sclerosis Genetics Consortium. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 2015;47:979–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Uniken Venema WT, Voskuil MD, Dijkstra G, et al. The genetic background of inflammatory bowel disease: from correlation to causality. J Pathol. 2017;241:146–58. [DOI] [PubMed] [Google Scholar]
- [26].Moon CM, Kim SW, Ahn JB, et al. Deep resequencing of ulcerative colitis-associated genes identifies novel variants in candidate genes in the Korean population. Inflamm Bowel Dis. 2018;24:1706–17. [DOI] [PubMed] [Google Scholar]
- [27].Degenhardt F, Mayr G, Wendorff M, et al.; MAAIS Recruitment Center. Transethnic analysis of the human leukocyte antigen region for ulcerative colitis reveals not only shared but also ethnicity-specific disease associations. Hum Mol Genet. 2021;30:356–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Wei SC, Sollano J, Hui YT, et al. Epidemiology, burden of disease, and unmet needs in the treatment of ulcerative colitis in Asia. Expert Rev Gastroenterol Hepatol. 2021;15:275–89. [DOI] [PubMed] [Google Scholar]
- [29].Sharara AI, Al Awadhi S, Alharbi O, et al. Epidemiology, disease burden, and treatment challenges of ulcerative colitis in Africa and the Middle East. Expert Rev Gastroenterol Hepatol. 2018;12:883–97. [DOI] [PubMed] [Google Scholar]
- [30].Timmer A. Environmental influences on inflammatory bowel disease manifestations. lessons from epidemiology. Dig Dis. 2003;21:91–104. [DOI] [PubMed] [Google Scholar]
- [31].Keshteli AH, Madsen KL, Dieleman LA. Diet in the pathogenesis and management of ulcerative colitis; a review of randomized controlled dietary interventions. Nutrients. 2019;11:1498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Lunney PC, Leong RW. Review article: ulcerative colitis, smoking and nicotine therapy. Aliment Pharmacol Ther. 2012;36:997–1008. [DOI] [PubMed] [Google Scholar]
- [33].Ianiro G, Tilg H, Gasbarrini A. Antibiotics as deep modulators of gut microbiota: between good and evil. Gut. 2016;65:1906–15. [DOI] [PubMed] [Google Scholar]
- [34].Levine A, Rhodes JM, Lindsay JO, et al. Dietary guidance from the international organization for the study of inflammatory bowel diseases. Clin Gastroenterol Hepatol. 2020;18:1381–92. [DOI] [PubMed] [Google Scholar]
- [35].Ananthakrishnan AN, Khalili H, Konijeti GG, et al. Long-term intake of dietary fat and risk of ulcerative colitis and Crohn’s disease. Gut. 2014;63:776–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Khalili H, Chan SSM, Lochhead P, et al. The role of diet in the aetiopathogenesis of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2018;15:525–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Gill SK, Rossi M, Bajka B, et al. Dietary fibre in gastrointestinal health and disease. Nat Rev Gastroenterol Hepatol. 2021;18:101–16. [DOI] [PubMed] [Google Scholar]
- [38].O’Grady J, O’Connor EM, Shanahan F. Review article: dietary fibre in the era of microbiome science. Aliment Pharmacol Ther. 2019;49:506–15. [DOI] [PubMed] [Google Scholar]
- [39].Porter RJ, Kalla R, Ho GT. Ulcerative colitis: Recent advances in the understanding of disease pathogenesis. F1000Res. 2020;9:F1000 Faculty Rev–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Wu Y, Wu B, Zhang Z, et al. Heme protects intestinal mucosal barrier in DSS-induced colitis through regulating macrophage polarization in both HO-1-dependent and HO-1-independent way. FASEB J. 2020;34:8028–43. [DOI] [PubMed] [Google Scholar]
- [41].Scott JR, Cukiernik MA, Ott MC, et al. Low-dose inhaled carbon monoxide attenuates the remote intestinal inflammatory response elicited by hindlimb ischemia-reperfusion. Am J Physiol Gastrointest Liver Physiol. 2009;296:G9–G14. [DOI] [PubMed] [Google Scholar]
- [42].Tripathi MK, Pratap CB, Dixit VK, et al. Ulcerative colitis and its association with salmonella species. Interdiscip Perspect Infect Dis. 2016;2016:5854285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Weaver KN, Herfarth HH, Barnes EL. Antibiotic use patterns in the management of chronic pouchitis. Inflamm Bowel Dis. 2022;28:e92–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Peterson CT, Sharma V, Elmén L, et al. Immune homeostasis, dysbiosis and therapeutic modulation of the gut microbiota. Clin Exp Immunol. 2015;179:363–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Rees WD, Sly LM, Steiner TS. How do immune and mesenchymal cells influence the intestinal epithelial cell compartment in inflammatory bowel disease? Let’s crosstalk about it! J Leukoc Biol. 2020;108:309–21. [DOI] [PubMed] [Google Scholar]
- [46].Wallace KL, Zheng LB, Kanazawa Y, et al. Immunopathology of inflammatory bowel disease. World J Gastroenterol. 2014;20:6–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Mowat AM, Agace WW. Regional specialization within the intestinal immune system. Nat Rev Immunol. 2014;14:667–85. [DOI] [PubMed] [Google Scholar]
- [48].Pelaseyed T, Bergström JH, Gustafsson JK, et al. The mucus and mucins of the goblet cells and enterocytes provide the first defense line of the gastrointestinal tract and interact with the immune system. Immunol Rev. 2014;260:8–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Fuss IJ. Is the Th1/Th2 paradigm of immune regulation applicable to IBD? [published correction appears in Inflamm Bowel Dis. 2009 Sep;15(9):1438–47]. Inflamm Bowel Dis. 2008;14(Suppl 2):S110–2. [DOI] [PubMed] [Google Scholar]
- [50].De Salvo C, Buela KA, Pizarro TT. Cytokine-mediated regulation of innate lymphoid cell plasticity in gut mucosal immunity. Front Immunol. 2020;11:585319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Siebenlist U, Brown K, Claudio E. Control of lymphocyte development by nuclear factor-kappaB. Nat Rev Immunol. 2005;5:435–45. [DOI] [PubMed] [Google Scholar]
- [52].Perkins ND. Post-translational modifications regulating the activity and function of the nuclear factor kappa B pathway. Oncogene. 2006;25:6717–30. [DOI] [PubMed] [Google Scholar]
- [53].Shen J, Cheng J, Zhu S, et al. Regulating effect of baicalin on IKK/IKB/NF-kB signaling pathway and apoptosis-related proteins in rats with ulcerative colitis. Int Immunopharmacol. 2019;73:193–200. [DOI] [PubMed] [Google Scholar]
- [54].Wei YY, Fan YM, Ga Y, et al. Shaoyao decoction attenuates DSS-induced ulcerative colitis, macrophage and NLRP3 inflammasome activation through the MKP1/NF-κB pathway. Phytomedicine. 2021;92:153743. [DOI] [PubMed] [Google Scholar]
- [55].Xiong T, Zheng X, Zhang K, et al. Ganluyin ameliorates DSS-induced ulcerative colitis by inhibiting the enteric-origin LPS/TLR4/NF-κB pathway. J Ethnopharmacol. 2022;289:115001. [DOI] [PubMed] [Google Scholar]
- [56].Abou Zaid ES, Mansour SZ, El-Sonbaty SM, et al. Boswellic acid coated zinc nanoparticles attenuate NF-κB-mediated inflammation in DSS-induced ulcerative colitis in rats. Int J Immunopathol Pharmacol. 2023;37:3946320221150720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Liu N, Zhao N, Cai N. The effect and mechanism of celecoxib in hypoxia-induced survivin up-regulation in HUVECs. Cell Physiol Biochem. 2015;37:991–1001. [DOI] [PubMed] [Google Scholar]
- [58].Sheu SY, Hong YW, Sun JS, et al. Radix Scrophulariae extracts (harpagoside) suppresses hypoxia-induced microglial activation and neurotoxicity. BMC Complement Altern Med. 2015;15:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Li Y, Soendergaard C, Bergenheim FH, et al. COX-2-PGE2 signaling impairs intestinal epithelial regeneration and associates with TNF inhibitor responsiveness in ulcerative colitis. EBioMedicine. 2018;36:497–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Granja AG, Nogal ML, Hurtado C, et al. The viral protein A238L inhibits cyclooxygenase-2 expression through a nuclear factor of activated T cell-dependent transactivation pathway. J Biol Chem. 2004;279:53736–46. [DOI] [PubMed] [Google Scholar]
- [61].Wang C, Li W, Wang H, et al. Saccharomyces boulardii alleviates ulcerative colitis carcinogenesis in mice by reducing TNF-α and IL-6 levels and functions and by rebalancing intestinal microbiota. BMC Microbiol. 2019;19:246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Xuan-Qing CHEN, Xiang-Yu LV, Shi-Jia LIU. Baitouweng decoction alleviates dextran sulfate sodium-induced ulcerative colitis by regulating intestinal microbiota and the IL-6/STAT3 signaling pathway. J Ethnopharmacol. 2021;265:113357. [DOI] [PubMed] [Google Scholar]
- [63].Fouad MR, Salama RM, Zaki HF, et al. Vildagliptin attenuates acetic acid-induced colitis in rats via targeting PI3K/Akt/NFκB, Nrf2 and CREB signaling pathways and the expression of lncRNA IFNG-AS1 and miR-146a. Int Immunopharmacol. 2021;92:107354. [DOI] [PubMed] [Google Scholar]
- [64].Fruman DA, Chiu H, Hopkins BD, et al. The PI3K pathway in human disease. Cell. 2017;170:605–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Wang Q, Zhang P, Zhang W, et al. PI3K activation is enhanced by FOXM1D binding to p110 and p85 subunits. Signal Transduct Target Ther. 2020;5:105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Fox M, Mott HR, Owen D. Class IA PI3K regulatory subunits: p110-independent roles and structures. Biochem Soc Trans. 2020;48:1397–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Płóciennikowska A, Hromada-Judycka A, Borzęcka K, et al. Co-operation of TLR4 and raft proteins in LPS-induced pro-inflammatory signaling. Cell Mol Life Sci. 2015;72:557–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Liu B, Piao X, Niu W, et al. Kuijieyuan decoction improved intestinal barrier injury of ulcerative colitis by affecting TLR4-dependent PI3K/AKT/NF-κB oxidative and inflammatory signaling and gut microbiota. Front Pharmacol. 2020;11:1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Peng KY, Gu JF, Su SL, et al. Salvia miltiorrhiza stems and leaves total phenolic acids combination with tanshinone protect against DSS-induced ulcerative colitis through inhibiting TLR4/PI3K/AKT/mTOR signaling pathway in mice. J Ethnopharmacol. 2021;264:113052. [DOI] [PubMed] [Google Scholar]
- [70].Luttun A, Tjwa M, Moons L, et al. Revascularization of ischemic tissues by PlGF treatment, and inhibition of tumor angiogenesis, arthritis and atherosclerosis by anti-Flt1. Nat Med. 2002;8:831–40. [DOI] [PubMed] [Google Scholar]
- [71].Zhou Y, Tu C, Zhao Y, et al. Placental growth factor enhances angiogenesis in human intestinal microvascular endothelial cells via PI3K/Akt pathway: potential implications of inflammation bowel disease. Biochem Biophys Res Commun. 2016;470:967–74. [DOI] [PubMed] [Google Scholar]
- [72].Hindryckx P, Waeytens A, Laukens D, et al. Absence of placental growth factor blocks dextran sodium sulfate-induced colonic mucosal angiogenesis, increases mucosal hypoxia and aggravates acute colonic injury. Lab Invest. 2010;90:566–76. [DOI] [PubMed] [Google Scholar]
- [73].Algaba A, Linares PM, Fernández-Contreras ME, et al. Relationship between levels of angiogenic and lymphangiogenic factors and the endoscopic, histological and clinical activity, and acute-phase reactants in patients with inflammatory bowel disease. J Crohns Colitis. 2013;7:e569–79. [DOI] [PubMed] [Google Scholar]
- [74].Danese S, Sans M, Spencer DM, et al. Angiogenesis blockade as a new therapeutic approach to experimental colitis. Gut. 2007;56:855–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Grivennikov SI, Cominelli F. Colitis-associated and sporadic colon cancers: different diseases, different mutations? Gastroenterology. 2016;150:808–10. [DOI] [PubMed] [Google Scholar]
- [76].Zhu Y, Jiang H, Chen Z, et al. Genetic association between IL23R rs11209026 and rs10889677 polymorphisms and risk of Crohn’s disease and ulcerative colitis: evidence from 41 studies. Inflamm Res. 2020;69:87–103. [DOI] [PubMed] [Google Scholar]
- [77].Razali NN, Raja Ali RA, Muhammad Nawawi KN, et al. Targeted sequencing of cytokine-induced PI3K-related genes in ulcerative colitis, colorectal cancer and colitis-associated cancer. Int J Mol Sci. 2022;23:11472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Yaeger R, Shah MA, Miller VA, et al. Genomic alterations observed in colitis-associated cancers are distinct from those found in sporadic colorectal cancers and vary by type of inflammatory bowel disease. Gastroenterology. 2016;151:278–287.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Salas A, Hernandez-Rocha C, Duijvestein M, et al. JAK-STAT pathway targeting for the treatment of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2020;17:323–37. [DOI] [PubMed] [Google Scholar]
- [80].Cordes F, Foell D, Ding JN, et al. Differential regulation of JAK/STAT-signaling in patients with ulcerative colitis and Crohn’s disease. World J Gastroenterol. 2020;26:4055–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Villarino AV, Kanno Y, O’Shea JJ. Mechanisms and consequences of Jak-STAT signaling in the immune system. Nat Immunol. 2017;18:374–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Harris C, Cummings JRF. JAK1 inhibition and inflammatory bowel disease. Rheumatology (Oxford). 2021;60(Suppl 2):ii45–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Galien R. Janus kinases in inflammatory bowel disease: Four kinases for multiple purposes. Pharmacol Rep. 2016;68:789–96. [DOI] [PubMed] [Google Scholar]
- [84].Sohn SJ, Barrett K, Van Abbema A, et al. A restricted role for TYK2 catalytic activity in human cytokine responses revealed by novel TYK2-selective inhibitors. J Immunol. 2013;191:2205–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Lu D, Liu L, Ji X, et al. The phosphatase DUSP2 controls the activity of the transcription activator STAT3 and regulates TH17 differentiation. Nat Immunol. 2015;16:1263–73. [DOI] [PubMed] [Google Scholar]
- [86].Zhao Y, Luan H, Jiang H, et al. Gegen Qinlian decoction relieved DSS-induced ulcerative colitis in mice by modulating Th17/Treg cell homeostasis via suppressing IL-6/JAK2/STAT3 signaling. Phytomedicine. 2021;84:153519. [DOI] [PubMed] [Google Scholar]
- [87].Zundler S, Neurath MF. Integrating immunologic signaling networks: the JAK/STAT pathway in colitis and colitis-associated cancer. Vaccines (Basel). 2016;4:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Pang YH, Zheng CQ, Yang XZ, et al. Increased expression and activation of IL-12-induced Stat4 signaling in the mucosa of ulcerative colitis patients. Cell Immunol. 2007;248:115–20. [DOI] [PubMed] [Google Scholar]
- [89].Soendergaard C, Bergenheim FH, Bjerrum JT, et al. Targeting JAK-STAT signal transduction in IBD. Pharmacol Ther. 2018;192:100–11. [DOI] [PubMed] [Google Scholar]
- [90].Wynick C, Petes C, Gee K. Interleukin-27 mediates inflammation during chronic disease. J Interferon Cytokine Res. 2014;34:741–9. [DOI] [PubMed] [Google Scholar]
- [91].Yan Y, Zhao N, He X, et al. Mesenchymal stem cell expression of interleukin-35 protects against ulcerative colitis by suppressing mucosal immune responses. Cytotherapy. 2018;20:911–8. [DOI] [PubMed] [Google Scholar]
- [92].Rodig SJ, Meraz MA, White JM, et al. Disruption of the Jak1 gene demonstrates obligatory and nonredundant roles of the Jaks in cytokine-induced biologic responses. Cell. 1998;93:373–83. [DOI] [PubMed] [Google Scholar]
- [93].Parganas E, Wang D, Stravopodis D, et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell. 1998;93:385–95. [DOI] [PubMed] [Google Scholar]
- [94].Mishra J, Verma RK, Alpini G, et al. Role of Janus kinase 3 in mucosal differentiation and predisposition to colitis. J Biol Chem. 2013;288:31795–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Nakase H, Sato N, Mizuno N, et al. The influence of cytokines on the complex pathology of ulcerative colitis. Autoimmun Rev. 2022;21:103017. [DOI] [PubMed] [Google Scholar]
- [96].Feuerstein JD, Isaacs KL, Schneider Y, et al.; AGA Institute Clinical Guidelines Committee. AGA clinical practice guidelines on the management of moderate to severe ulcerative colitis. Gastroenterology. 2020;158:1450–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Petito V, Graziani C, Lopetuso LR, et al. Anti-tumor necrosis factor α therapy associates to type 17 helper T lymphocytes immunological shift and significant microbial changes in dextran sodium sulphate colitis. World J Gastroenterol. 2019;25:1465–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Hossain A, Lördal M, Olsson AE, et al. Sustained clinical benefit, improved quality of life, and reduced intestinal surgery from maintenance infliximab treatment in inflammatory bowel disease. Scand J Gastroenterol. 2020;55:178–83. [DOI] [PubMed] [Google Scholar]
- [99].Engström J, Lönnkvist M, Befrits R, et al. Comparison of fecal calprotectin and serum C-reactive protein in early prediction of outcome to infliximab induction therapy. Scand J Gastroenterol. 2019;54:1081–8. [DOI] [PubMed] [Google Scholar]
- [100].Sparrow MP. Adalimumab in ulcerative colitis – efficacy, safety and optimization in the era of treat-to target. Expert Opin Biol Ther. 2017;17:613–21. [DOI] [PubMed] [Google Scholar]
- [101].Fernández-Blanco JI, Fernández-Díaz G, Cara C, et al. Adalimumab for induction of histological remission in moderately to severely active ulcerative colitis. Dig Dis Sci. 2018;63:731–7. [DOI] [PubMed] [Google Scholar]
- [102].Danese S. Adalimumab in ulcerative colitis: ready for prime time. Dig Liver Dis. 2013;45:8–13. [DOI] [PubMed] [Google Scholar]
- [103].Bhattacharya A, Osterman MT. Biologic therapy for ulcerative colitis. Gastroenterol Clin North Am. 2020;49:717–29. [DOI] [PubMed] [Google Scholar]
- [104].Kedia S, Ahuja V, Makharia GK. Golimumab for moderately to severely active ulcerative colitis. Expert Rev Clin Pharmacol. 2016;9:1273–82. [DOI] [PubMed] [Google Scholar]
- [105].Sandborn WJ, Feagan BG, Marano C, et al.; PURSUIT-Maintenance Study Group. Subcutaneous golimumab maintains clinical response in patients with moderate-to-severe ulcerative colitis. Gastroenterology. 2014;146:96–109.e1. [DOI] [PubMed] [Google Scholar]
- [106].Park SC, Jeen YT. Anti-integrin therapy for inflammatory bowel disease. World J Gastroenterol. 2018;24:1868–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Gubatan J, Keyashian K, Rubin SJS, et al. Anti-integrins for the treatment of inflammatory bowel disease: current evidence and perspectives. Clin Exp Gastroenterol. 2021;14:333–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Panés J, Salas A. Past, present and future of therapeutic interventions targeting leukocyte trafficking in inflammatory bowel disease. J Crohns Colitis. 2018;12(suppl_2):S633–40. [DOI] [PubMed] [Google Scholar]
- [109].Feagan BG, Rutgeerts P, Sands BE, et al.; GEMINI 1 Study Group. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2013;369:699–710. [DOI] [PubMed] [Google Scholar]
- [110].D’Amico F, Danese S, Peyrin-Biroulet L. Vedolizumab and etrolizumab for ulcerative colitis: twins or simple cousins? Expert Opin Biol Ther. 2020;20:353–61. [DOI] [PubMed] [Google Scholar]
- [111].Vermeire S, Lakatos PL, Ritter T, et al.; LAUREL Study Group. Etrolizumab for maintenance therapy in patients with moderately to severely active ulcerative colitis (LAUREL): a randomised, placebo-controlled, double-blind, phase 3 study. Lancet Gastroenterol Hepatol. 2022;7:28–37. [DOI] [PubMed] [Google Scholar]
- [112].Bellaguarda E, Keyashian K, Pekow J, et al. Prevalence of antibodies against JC virus in patients with refractory Crohn’s disease and effects of natalizumab therapy. Clin Gastroenterol Hepatol. 2015;13:1919–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Gee K, Guzzo C, Che Mat NF, et al. The IL-12 family of cytokines in infection, inflammation and autoimmune disorders. Inflamm Allergy Drug Targets. 2009;8:40–52. [DOI] [PubMed] [Google Scholar]
- [114].Sands BE, Sandborn WJ, Panaccione R, et al.; UNIFI Study Group. Ustekinumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2019;381:1201–14. [DOI] [PubMed] [Google Scholar]
- [115].Chaparro M, Garre A, Iborra M, et al. Effectiveness and safety of ustekinumab in ulcerative colitis: real-world evidence from the ENEIDA registry. J Crohns Colitis. 2021;15:1846–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Hanžel J, D’Haens GR. Anti-interleukin-23 agents for the treatment of ulcerative colitis. Expert Opin Biol Ther. 2020;20:399–406. [DOI] [PubMed] [Google Scholar]
- [117].Morris R, Kershaw NJ, Babon JJ. The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci. 2018;27:1984–2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].López-Sanromán A, Esplugues JV, Domènech E. Pharmacology and safety of tofacitinib in ulcerative colitis. Farmacología y seguridad de tofacitinib en colitis ulcerosa. Gastroenterol Hepatol. 2021;44:39–48. [DOI] [PubMed] [Google Scholar]
- [119].Spalinger MR, Sayoc-Becerra A, Ordookhanian C, et al. The JAK inhibitor tofacitinib rescues intestinal barrier defects caused by disrupted epithelial-macrophage interactions. J Crohns Colitis. 2021;15:471–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Sayoc-Becerra A, Krishnan M, Fan S, et al. The JAK-inhibitor tofacitinib rescues human intestinal epithelial cells and colonoids from cytokine-induced barrier dysfunction. Inflamm Bowel Dis. 2020;26:407–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Sandborn WJ, Su C, Sands BE, et al.; OCTAVE Induction 1, OCTAVE Induction 2, and OCTAVE Sustain Investigators. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2017;376:1723–36. [DOI] [PubMed] [Google Scholar]
- [122].Panés J, Vermeire S, Lindsay JO, et al. Tofacitinib in patients with ulcerative colitis: health-related quality of life in phase 3 randomised controlled induction and maintenance studies [published correction appears in J Crohns Colitis. 2019 Jan 1;13(1):139–140]. J Crohns Colitis. 2018;12:145–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Namour F, Anderson K, Nelson C, et al. Filgotinib: a clinical pharmacology review. Clin Pharmacokinet. 2022;61:819–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Feagan BG, Danese S, Loftus EV, Jr, et al. Filgotinib as induction and maintenance therapy for ulcerative colitis (SELECTION): a phase 2b/3 double-blind, randomised, placebo-controlled trial. Lancet. 2021;397:2372–84. [DOI] [PubMed] [Google Scholar]
- [125].Sandborn WJ, Ghosh S, Panes J, et al. Efficacy of upadacitinib in a randomized trial of patients with active ulcerative colitis [published correction appears in Gastroenterology. 2020 Sep;159(3):1192]. Gastroenterology. 2020;158:2139–2149.e14. [DOI] [PubMed] [Google Scholar]
- [126].Danese S, Vermeire S, Zhou W, et al. Upadacitinib as induction and maintenance therapy for moderately to severely active ulcerative colitis: results from three phase 3, multicentre, double-blind, randomised trials [published correction appears in Lancet. 2022 Sep 24;400(10357):996]. Lancet. 2022;399:2113–28. [DOI] [PubMed] [Google Scholar]
- [127].Verstockt B, Vetrano S, Salas A, et al.; Alimentiv Translational Research Consortium (ATRC). Sphingosine 1-phosphate modulation and immune cell trafficking in inflammatory bowel disease [published correction appears in Nat Rev Gastroenterol Hepatol. 2022 Mar 7]. Nat Rev Gastroenterol Hepatol. 2022;19:351–66. [DOI] [PubMed] [Google Scholar]
- [128].Sandborn WJ, Feagan BG, D’Haens G, et al.; True North Study Group. Ozanimod as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2021;385:1280–91. [DOI] [PubMed] [Google Scholar]
- [129].Wils P, Peyrin-Biroulet L. Etrasimod for the treatment of ulcerative colitis. Immunotherapy. 2023;15:311–21. [DOI] [PubMed] [Google Scholar]
- [130].Sandborn WJ, Peyrin-Biroulet L, Zhang J, et al. Efficacy and safety of etrasimod in a phase 2 randomized trial of patients with ulcerative colitis. Gastroenterology. 2020;158:550–61. [DOI] [PubMed] [Google Scholar]
