Background: The specific etiopathogenesis of inflammatory bowel disease (IBD) is still unknown. Although the conventional anti-inflammatory or immunomodulatory drugs relatively nonspecific to pathogenesis have been quite useful in many cases, elucidating the pathogenesis has gradually facilitated developments of disease-specific therapies for refractory cases in the last 2 decades. Summary: With a greater understanding of the multiple overactive signaling pathways of the gut mucosal immune response and enhanced leukocyte trafficking, several biological agents or small molecule drugs following the first novel biologic, anti-tumor necrosis factor α (anti-TNFα), have been developed against several modes of action including adhesion molecules, sphingosine-1-phospate receptors, cytokines (IL-12/23, TL1A, and IL-36), Janus kinase (JAK), and phosphodiesterase. Although preceding biological agents have dramatically changed the IBD treatment strategy, many patients still require alternative therapies due to failure or side effects. Newer treatments are now expected to be provided for better efficacy with an improved adverse event profile. In addition, translational studies have highlighted the new therapeutic concepts’ potential, including modulation of host-microbiome interactions, stem therapy for perianal fistula, regulation of fibrosis, regulation of the gut-brain axis, and control of previously less targeted immune cells (B cells and innate lymphoid cells). This paper comprehensively reviewed not only the latest already or shortly available therapies but also emerging promising treatments that will be hopefully established in the future for IBD. Key Messages: Many kinds of new treatments are available, and promising treatments with new perspectives are expected to emerge for refractory IBD in the future.

Inflammatory bowel disease (IBD) is a chronic or remitting/relapsing inflammatory disease of the intestinal tract, including ulcerative colitis (UC) and Crohn’s disease (CD); the number of patients with IBD has been increasing worldwide [1]. Although the etiology of IBD remains unclear, elucidating its mechanism has facilitated therapeutic development, especially in the last 2 decades. Among the recently available therapies, anti-tumor necrosis factor α (anti-TNFα) agents are the most notable predecessors (infliximab, adalimumab, etc.), resulting in improved health outcomes and decreased need for surgical intervention [2]. However, treatment failure is observed in many patients treated with anti-TNFα agents, including primary and secondary nonresponders. Additionally, anti-TNF-α agents are associated with rare but serious adverse effects, including serious infection, paradoxical autoimmune reactions, and a small but increased risk of malignancy. In this situation, other novel anti-inflammatory therapies targeting inflammatory pathways, including cellular signaling (e.g., anti-interleukin (IL) 12/23 agents and Janus kinase (JAK) inhibitors) and leukocyte trafficking (anti-integrin agents), have been developed with improved safety or pharmacokinetics (i.e., less frequent injection, short administration time, oral therapy). In addition, translational studies have highlighted the potential of various new therapeutic targets, including B cells, innate lymphoid cells (ILCs), bile acids (BA), the brain-gut axis, and microbiota. This article comprehensively reviewed already or shortly available new treatments (1. New drugs) and a wide range of emerging promising therapies that will be hopefully established for IBD in the new era (2. Future perspectives). The brief mechanisms and roles of the new and emerging treatments are summarized in Figure 1.

Fig. 1.

Schematic summary of new and emerging treatments for IBD. ACh acetylcholine, α7nAChR; α7-nicotinic acetylcholine receptors; DCs, dendritic cells; DR3, death receptor 3; HEV, high endothelial venule; ICAM-1, intercellular adhesion molecule-1; IELs, intraepithelial cells; ILCs, innate lymphoid cells; IL-1RAcP, interleukin-1 receptor accessory protein; PDE4, phosphodiesterase 4; PSGL-1, P-selectin glycoprotein ligand-1; S1PR1, sphingosine-1-phosphate receptor 1; TL1A, TNF-like ligand 1A; TNF-α, tumor necrosis factor α; TNFRs, TNF receptors; VCAM-1, vascular cellular adhesion molecule-1; VN, vagus nerve. Orange boxes: new drugs. Blue boxes: future perspectives.

Fig. 1.

Schematic summary of new and emerging treatments for IBD. ACh acetylcholine, α7nAChR; α7-nicotinic acetylcholine receptors; DCs, dendritic cells; DR3, death receptor 3; HEV, high endothelial venule; ICAM-1, intercellular adhesion molecule-1; IELs, intraepithelial cells; ILCs, innate lymphoid cells; IL-1RAcP, interleukin-1 receptor accessory protein; PDE4, phosphodiesterase 4; PSGL-1, P-selectin glycoprotein ligand-1; S1PR1, sphingosine-1-phosphate receptor 1; TL1A, TNF-like ligand 1A; TNF-α, tumor necrosis factor α; TNFRs, TNF receptors; VCAM-1, vascular cellular adhesion molecule-1; VN, vagus nerve. Orange boxes: new drugs. Blue boxes: future perspectives.

Close modal

The pathogenesis of chronic inflamed intestinal mucosa in IBD consists of multistep processes, including migrating lymphocytes from secondary lymphoid organs (SLOs) to the bloodstream, into the gut mucosa through high endothelial venules (HEVs), and proliferation and activation of inflammatory cells [3]. Most new treatments specifically inhibit these points of action. Table 1 shows the summary of new drugs with mode of action and development status.

Table 1.

Summary of new treatments for IBD

 Summary of new treatments for IBD
 Summary of new treatments for IBD

Inhibition of Immune Cell Trafficking

Anti-Adhesion Agents

The trafficking of T lymphocytes from SLOs to the site of inflamed intestine in IBD is mediated by several enhanced chemokines and selectins, leading to the adhesion of integrins expressed on T cells to ligands on HEVs, followed by their transmigration into intestinal tissue. New pharmacological agents have been developed to prevent leukocyte trafficking by selectively targeting the adhesion molecules involved in the pathogenesis of IBD [3]. Among several adhesion molecules, integrin α4β7 is preferentially expressed on lymphocytes activated in gut SLOs and interacts with mucosal addressin cell adhesion molecule-1 (MAdCAM-1), which is highly expressed on HEVs [3]. Natalizumab, a recombinant humanized monoclonal antibody against α4-integrin, was initially approved to treat multiple sclerosis (MS), followed by CD [4]. However, the α4-integrin subunit is critical for the α4β1-dependent central nervous system (CNS) homing of lymphocytes, and natalizumab’s use has been drastically limited by its association with life-threatening CNS infection and progressive multifocal leukoencephalopathy (PML, incidence: 1/100-1/1000) due to JC virus reactivation in cases administered for over 8 months (85% of PML cases were administered for more than 24 months) [5]. Vedolizumab is a humanized monoclonal antibody against α4β7 that selectively prevents lymphocyte migration and improves chronic intestinal inflammation. Vedolizumab effectively induces and maintains remission in IBD patients refractory to conventional therapies, with a generally favorable long-term safety profile (GEMINI LTS study), offering an advantage over natalizumab because of no association with the occurrence of PML [6]. Etrolizumab is a humanized monoclonal antibody against the β7 subunit of the heterodimeric integrins α4β7 and αEβ7 expressed on lymphocytes, thus concurrently inhibiting their interaction with MAdCAM-1 and E-cadherin, respectively, leading to the reduction of inflammatory T cells and cytotoxic intraepithelial lymphocytes in the gut mucosa via blockade of intestinal tissue entry and retention [7]. Carotegrast methyl is an orally active, small molecule with a similar action mechanism to natalizumab and was approved in Japan for UC treatment in 2022 [8]. Theoretically, carotegrast methyl can lead to an increased risk of PML as mentioned above. Therefore, the treatment duration was limited to <6 months and there was no case of PML reported. Ontamalimab is a monoclonal IgG2 antibody against MAdCAM-1, thus preventing lymphocyte migration into sites of intestinal inflammation and is efficacious for IBD treatment [4]. Other potential targets of adhesion molecules for the treatment of IBD include intercellular adhesion molecules, vascular cellular adhesion molecule-1, P-selectin, and P-selectin glycoprotein ligand-1 [9].

Sphingosine-1-Phosphate Modulators

The sphingosine-1-phosphate receptor (S1PR) comprises 5 subtypes and plays several important roles, including cell proliferation and migration, intercellular communication, vascular tone maintenance, and other cardiovascular effects. S1PR1 plays an essential role in controlling lymphocyte egress from the thymus, SLOs, and bone marrow [10]. Several S1P modulators have been developed to treat immune-mediated diseases, including IBD, MS, rheumatoid arthritis (RA), systemic lupus erythematosus, and psoriasis, with promising results [10]. Ozanimod, an orally administered small molecule and selective immunomodulator of the S1PR1 and S1PR5 receptors, is effective as an induction and maintenance therapy for IBD by receptor internalization and degradation, thus inhibiting the migration of lymphocytes along a gradient of S1P concentration from SLOs into the systemic circulation [10]. Other S1P modulators, including etrasimod and amiselimod, have also been developed and used in clinical trials [10].

Inhibition of Cytokines

Targeting IL-12/23 Pathways

IL-12, a heterodimer of p40 and p35, and IL-23, a heterodimer of p40 and p19, are released from myeloid cells, including dendritic cells (DCs) or macrophages, induce differentiation of naïve CD4+ T cells into T-helper 1 (Th1) and T-helper 17 (Th17) cells, and play important roles in the pathogenesis of IBD [11]. Ustekinumab is a fully human IgG1 monoclonal antibody that targets the IL-12/23 shared p40 subunit, preventing its binding to the receptors on cells [11]. UNITI and UNIFI studies have shown ustekinumab’s long-term efficacy and safety for IBD. Risankizumab is a humanized IgG1 monoclonal antibody that selectively binds the IL-23 p19 subunit with a high affinity. Theoretically, risankizumab may confer fewer side effects than ustekinumab by specifically targeting the IL-23-mediated inflammatory pathway without blocking the IL-12-dependent T cell activation pathway, which is vital for infection and cancer immunity. Studies targeting the IL-23 p19 subunit by other biologics, such as mirikizumab, brazikumab, and guselkumab, also showed effectiveness in controlling intestinal inflammation [11].

After the successful results of these ant-IL-23 p40 and p19 monoclonal antibodies, targeting the IL-23/Th17 axis was expected to be a good strategy for IBD. In the IL-17 subclass, IL-17A and IL-17F are the representative cytokines, and Secukinumab (anti-IL-17A antibody) and Brodalumab (anti-IL-17RA antibody) are effective against psoriasis. However, they showed paradoxical gastrointestinal effects and were not approved for IBD therapy [11], since it is mainly assumed that IL-17F is normally pathogenic in enteritis but IL-17A is protective for intestinal epithelial barrier integrity [12].

JAK Inhibitors

Cytokines promote intracellular signaling through induction of the JAK/signal transducer and activator of transcription (JAK/STAT) signaling pathway. JAKs, a family of intracellular tyrosine protein kinases, are comprised of 4 members: JAK1, JAK2, JAK3, and tyrosine kinase 2 (TYK2). Inhibition of the JAK/STAT signaling pathway gets a lot of attention for developing novel IBD therapies [13]. JAK inhibitors are small molecules and the first IBD-targeted therapy administered orally. JAK inhibitors are characterized by a rapid onset of action after entering systemic circulation and may provide fast symptomatic relief. In contrast to the aforementioned monoclonal antibodies that target a single cytokine (TNF-α, IL-12, or IL-23), JAK inhibitors have the potential to affect multiple cytokine-dependent immune pathways (for example, JAK1, JAK2, and TYK2 mediate signaling of IL-6, while JAK2 and TYK2 also mediate signaling of IL-23) [13]. Several JAK inhibitors have already been approved for treating RA. Among them, tofacitinib is a highly effective nonselective JAK inhibitor (preferentially inhibits JAK1 and JAK3) that was first approved for induction and maintenance therapy in UC. Remission rates at 8 weeks in OCTAVE induction 1 and 2 compared to placebo were 18.5% versus 8.2% (p < 0.01) and 16.6% versus 3.6% (p < 0.001), respectively. Remission rates at 52 weeks in the OCTAVE Sustain trial were 34.3% in 5 mg and 40.6% in 10 mg versus 11.1% in placebo (p < 0.0001 in both). The risk of herpes zoster infection increases in patients aged >65 years with previous failure of TNFα agents and Asian ethnicity [13]. In phase 2b clinical trials of CD, primary efficacy endpoints were not significantly different from placebo. The selective JAK1 inhibitors, filgotinib and upadacitinib, are also efficacious for IBD [14, 15]. In the maintenance study of filgotinib (SELECTION trial), clinical remission rate at 58 weeks of moderately and severe UC patients taking 200 mg were 37.2% compared to 11.2% in placebo (p < 0.0001). Filgotinib was assessed in CD in the phase 2 FITZROY study, which showed 47% of patients taking 200 mg achieved clinical remission at week 10 compared to 23% in placebo (p = 0.0077). In the U-ACHIEVE study, clinical remission rate at 8 weeks of moderately and severely UC patients taking upadacitinib 45 mg was 19.6% compared to 0% in placebo (p = 0.002). In the CELEST trial assessing upadacitinib in patients with moderate to severe CD, the clinical remission at week 16 was 27% of patients receiving 6 mg upadacitinib twice daily compared to 11% in placebo (p < 0.1). Deucravacitinib in phase 2 clinical trials of CD and UC is a highly selective TYK2 inhibitor that targets IL-12, IL-23, and type 1 interferon (IFN) [13].

TLI1A Inhibitor

Genome-wide association studies have identified that one of the strongest genetic variants related to IBD is the TNF superfamily member 15 (TNFSF15) locus [16]. The TNFSF15 gene is known as TNF-like ligand 1A (TL1A), which is secreted from antigen-presenting cells upon stimulation and plays a central role in activating innate and adaptive immune pathways by binding to death receptor 3 (DR3). TL1A-DR3 signaling is involved with proinflammatory effects including T cell activation and profibrotic pathways [16], both of which are critical for IBD pathogenesis. An anti-TL1A antibody, PF-06480605, downregulates tissue Th1 and Th17 cytokine responses along with significant endoscopic improvements in moderate to severe UC [17].

Phosphodiesterase Inhibitor

Phosphodiesterases (PDEs) are a heterogeneous and large family of enzymes that catalyze the degradation of cAMP and cGMP. PDE4 is expressed in DCs, macrophages, monocytes, and T cells. The inhibition of PDE4 elevates intracellular cAMP levels, reducing the expression of the inflammatory cytokines TNFα, IL-17, IFN-γ, and IL-23, while increasing regulatory cytokine levels, such as IL10. Therefore, the PDE4 inhibitor apremilast has been evaluated as a therapeutic agent in treating active UC and proved effective [18].

IL-36 Inhibitor

The expression of the IL-36 family in IBD is up-regulated during intestinal inflammation. Full-length IL-36α and IL-36γ are secreted by different gut-residing cells (epithelium, lymphocytes, and macrophages) and enzymatically processed by extracellular neutrophil proteases into highly active IL-36R agonists. Ligand binding to the IL-36R complex leads to activation of proinflammatory signaling in several cells, including DCs, macrophages, and T lymphocytes. The humanized monoclonal IgG1 antibody spesolimab that targets IL-36R is a potential therapy for IBD and in clinical trials [19].

Since multiple inflammatory pathways are activated in the inflamed intestine, blocking one of them might not be sufficient to control inflammation, as we now do with targeted monotherapies. Therefore, in the future, we will need to establish treatment strategies, such as sequential/combination therapy, to optimize the efficacy of each drug [20]; for example, using combination therapies in the induction phase of early IBD followed by monotherapy in remission. Meanwhile, therapeutic approaches with different mechanisms of action have been investigated from various perspectives. In this section, we discuss several possible future therapeutic targets.

Gut Microbiota Modulation

In this decade, a complex interaction between intestinal epithelial cells, the host immune system, and the gut microbiota has been investigated worldwide. Although the diversity and composition of human gut microbiota are believed to play a critical role in human health and the development of several diseases, there remain so many things to be clarified. The intestinal microbiota of patients has less biodiversity (dysbiosis) than healthy subjects, which is supposed to contribute to the onset of IBD. Therefore, microbiome-modulation gets attention as a new therapeutic approach. The beneficial effects mostly depend on their metabolism and metabolic by-products, including short-chain fatty acids, bacteriocins, hydroperoxides, secondary BA, and lactic acids [21]. Potential therapeutic methods include probiotics, prebiotics, fecal microbiota transplantation (FMT), synthetic combinations of specific bacteria, personalized therapies based on individual microbiome profiles, and regulation of BA [21]. Meta-analysis showed that FMT is a safe, effective, and well-tolerated therapy, although long-term effectiveness and safety need to be assessed more [22]. In IBD, the proportion of conjugated primary BA in feces increases, while the secondary BA proportion decreases due to blocked transformation by dysbiosis (conversion of secondary BA is attributed to limited bacteria with BA-inducible enzymes). Secondary BA exerts anti-inflammatory effects, partly by promoting T cell differentiation and inhibiting Th17 cell differentiation. In addition, BA is a natural ligand for several receptors (FXR, TGR5, PXR, and VDR) that regulate metabolic and immune processes. Based on these facts, several therapies targeting the BA-gut microbiota axis might be novel options for treating IBD [23]. Tryptophan is another metabolite involved in host-microbiota interactions. Tryptophan is metabolized by intestinal microorganisms into indole and its derivatives, many of which are ligands for aryl hydrocarbon receptor (AhR), activating the AhR/IL-22 pathway crucial for intestinal homeostasis partly through enhancing barrier integrity. Another metabolism pathway of tryptophan in microorganisms produces peripheral serotonin, an important gastrointestinal signaling molecule for intestinal peristalsis and motility, secretion, vasodilatation, and the absorption of nutrients. Therefore, microorganisms that manipulate tryptophan metabolism may have beneficial effects [24].

Stem Cell Therapy for Perianal Fistula

Perianal fistulas are a common complication of CD, and approximately one-third of patients develop this complication within 20 years after diagnosis. They deeply impair the quality of life and are poorly responsive to treatment. Closure of the fistula by surgery for a long time is not easy, with an additional risk of incontinence. Thus, effective treatments for perianal fistulas with fewer side effects have been investigated, and local injection of mesenchymal stem cells has recently been found to have a higher efficacy in managing perianal fistula in CD [25].

Regulation of Fibrosis

Gastrointestinal stricture is the pathological fibrotic stenosis of the gastrointestinal tract and is a common complication of CD. Chronic inflammation activates fibroblast, followed by excessive accumulation of extracellular matrix (ECM) and expansion of mesenchymal cells. Matrix metalloproteinases (MMPs) and MMP inhibitors control the generation and degradation of the ECM, and fibrosis progresses when ECM production exceeds the degradation rate. Recent studies also suggest some potential therapies, including anti-fibrotic drugs, anti-MMP9 antibodies, and inhibition of pH-sensing ovarian cancer G-protein-coupled receptor 1 and BCL2 [26].

Regulation of Gut-Brain Axis

The gut-brain axis is bidirectional communication between the gut and nervous system, including the CNS, autonomic nervous system, enteric nervous system, and hypothalamus-pituitary-adrenal axis, and highly complex and driven by neural, hormonal, metabolic, immunological, and microbial signals [27]. The vagus nerve (VN), a key component of the autonomic nervous system, is a pivotal element in brain-gut axis. Blunted vagal tone is related to high TNFα levels in IBD, and VN stimulation is a potential therapy for IBD via 2 pathways [27]. VN activates the hypothalamus-pituitary-adrenal axis through its afferent fibers, leading to the release of glucocorticoids followed by suppression of peripheral inflammation. Another anti-inflammatory pathway is the release of acetylcholine (ACh) at the distal end of the VN, which inhibits the release of TNFα from macrophages by binding to α7-nicotinic ACh receptors. In addition, dysregulation of the gut-brain axis is related to gastrointestinal and neurological diseases and a high risk of developing Parkinson’s disease and MS in IBD [28]. Mechanistically, enteric dysbiosis, translocation of microbial metabolites, and inflammatory factors derived from the inflamed intestinal mucosa across the gut epithelial barrier (leaky gut) and blood-brain barrier have been generally supposed to be major factors for altered structure and function in the CNS.

Regulation of B Cells

B cells at the intestinal mucosal surface are critical for immune homeostasis, and the dysregulated B-cell response may be a potential therapeutic target in IBD patients [29]. Since IBD patients show increased T cell-dependent mucosal IgG1+ plasma cells, inhibition of CD40L, which is pivotal for T cell-dependent class switching, may reduce inflammatory IgG1 responses while T cell-independent IgA responses are preserved. B-cell-activating factor (BAFF) is a TNF superfamily cytokine and is released by several immune cells upon stimulation for plasma cell and B-cell survival, B-cell maturation, and class-switch recombination. Since BAFF is overexpressed in IBD and Belimumab, a human monoclonal anti-BAFF IgG1 antibody, is effective in systemic lupus erythematosus and RA by reducing naïve and activated B cells as well as plasma cells, BAFF antagonists may ameliorate intestinal inflammation in IBD. Vedolizumab may inhibit the infiltration of circulating α4β7+ plasmablasts into inflamed mucosa. In addition, since some studies have shown a beneficial role of intravenous immunoglobulin in steroid-refractory IBD patients, blocking the Fcγ receptor function may be an attractive approach, although precise mechanism remains to be clarified [29].

Regulation of ILCs

ILCs are involved in IBD pathogenesis through interaction with microbiota, modulation of epithelial barrier integrity, and production of cytokines [30]. ILC3s are the major component of intestinal ILCs in normal state, but during active state, IFN-γ-producing ILC1s and IL-17-producing ILC3s increase while IL-22-producing ILC3s decrease. ILC2s are involved in IBD through their ability to sense microbiota and maintain the intestinal barrier. To develop new approaches for IBD, further research on the mechanisms that change the ILCs subpopulation and function and how these ILCs exert their protective and inflammatory effects is required [30].

New and emerging treatments for IBD were reviewed, and novel therapeutic approaches were presented to foresee future treatments. In these 2 decades, many novel drugs have been developed, and the therapeutic algorithms for IBD patients have drastically changed. However, there still remain many refractory patients and the number of IBD patients is increasing worldwide. Over the next 2 decades, where will we be? We hope that the number of patients with severe IBD will be reduced with many novel emerging treatments.

The authors have no conflict of interest to declare.

A grant from National Defense Medical College was received for the study.

Masaaki Higashiyama and Ryota Hokari drafted the manuscript.

1.
Kaplan
GG
,
Ng
SC
.
Understanding and preventing the global increase of inflammatory bowel disease
.
Gastroenterology
.
2017
;
152
(
2
):
313
21.e2
.
2.
Costa
J
,
Magro
F
,
Caldeira
D
,
Alarcão
J
,
Sousa
R
,
Vaz-Carneiro
A
.
Infliximab reduces hospitalizations and surgery interventions in patients with inflammatory bowel disease: a systematic review and meta-analysis
.
Inflamm Bowel Dis
.
2013
;
19
(
10
):
2098
110
.
3.
Habtezion
A
,
Nguyen
LP
,
Hadeiba
H
,
Butcher
EC
.
Leukocyte trafficking to the small intestine and colon
.
Gastroenterology
.
2016
;
150
(
2
):
340
54
.
4.
Gubatan
J
,
Keyashian
K
,
Rubin
SJS
,
Wang
J
,
Buckman
CA
,
Sinha
S
.
Anti-integrins for the treatment of inflammatory bowel disease: current evidence and perspectives
.
Clin Exp Gastroenterol
.
2021
;
14
:
333
42
.
5.
Berger
JR
.
Classifying PML risk with disease modifying therapies
.
Mult Scler Relat Disord
.
2017
;
12
:
59
63
.
6.
Loftus
EV
, Jr
,
Feagan
BG
,
Panaccione
R
,
Colombel
JF
,
Sandborn
WJ
,
Sands
BE
,
.
Long-term safety of vedolizumab for inflammatory bowel disease
.
Aliment Pharmacol Ther
.
2020
;
52
(
8
):
1353
65
.
7.
Dai
B
,
Hackney
JA
,
Ichikawa
R
,
Nguyen
A
,
Elstrott
J
,
Orozco
LD
,
.
Dual targeting of lymphocyte homing and retention through α4β7 and αEβ7 inhibition in inflammatory bowel disease
.
Cell Rep Med
.
2021
;
2
(
8
):
100381
.
8.
Matsuoka
K
,
Watanabe
M
,
Ohmori
T
,
Nakajima
K
,
Ishida
T
,
Ishiguro
Y
,
.
AJM300 (carotegrast methyl), an oral antagonist of α4-integrin, as induction therapy for patients with moderately active ulcerative colitis: a multicentre, randomised, double-blind, placebo-controlled, phase 3 study
.
Lancet Gastroenterol Hepatol
.
2022
;
7
(
7
):
648
57
.
9.
Liu
P
,
Gao
C
,
Chen
H
,
Vong
CT
,
Wu
X
,
Tang
X
,
.
Receptor-mediated targeted drug delivery systems for treatment of inflammatory bowel disease: opportunities and emerging strategies
.
Acta Pharm Sin B
.
2021
;
11
(
9
):
2798
818
.
10.
Perez-Jeldres
T
,
Alvarez-Lobos
M
,
Rivera-Nieves
J
.
Targeting sphingosine-1-phosphate signaling in immune-mediated diseases: beyond multiple sclerosis
.
Drugs
.
2021
;
81
(
9
):
985
1002
.
11.
Moschen
AR
,
Tilg
H
,
Raine
T
.
IL-12, IL-23 and IL-17 in IBD: immunobiology and therapeutic targeting
.
Nat Rev Gastroenterol Hepatol
.
2019
;
16
(
3
):
185
96
.
12.
Zhou
C
,
Wu
D
,
Jawale
C
,
Li
Y
,
Biswas
PS
,
McGeachy
MJ
,
.
Divergent functions of IL-17-family cytokines in DSS colitis: insights from a naturally-occurring human mutation in IL-17F
.
Cytokine
.
2021
;
148
:
155715
.
13.
Dudek
P
,
Fabisiak
A
,
Zatorski
H
,
Malecka-Wojciesko
E
,
Talar-Wojnarowska
R
.
Efficacy, safety and future perspectives of JAK inhibitors in the IBD treatment
.
J Clin Med
.
2021
;
10
(
23
):
5660
.
14.
Feagan
BG
,
Danese
S
,
Loftus
EV
Jr
,
Vermeire
S
,
Schreiber
S
,
Ritter
T
,
.
Filgotinib as induction and maintenance therapy for ulcerative colitis (SELECTION): a phase 2b/3 double-blind, randomised, placebo-controlled trial
.
Lancet
.
2021
;
397
(
10292
):
2372
84
.
15.
Sandborn
WJ
,
Ghosh
S
,
Panes
J
,
Schreiber
S
,
D'Haens
G
,
Tanida
S
,
.
Efficacy of Upadacitinib in a randomized trial of patients with active ulcerative colitis
.
Gastroenterology
.
2020
;
158
(
8
):
2139
49.e14
.
16.
Siakavellas
SI
,
Bamias
G
.
Tumor necrosis factor-like cytokine TL1A and its receptors DR3 and DcR3: important new factors in mucosal homeostasis and inflammation
.
Inflamm Bowel Dis
.
2015
;
21
(
10
):
2441
52
.
17.
Furfaro
F
,
Alfarone
L
,
Gilardi
D
,
Correale
C
,
Allocca
M
,
Fiorino
G
,
.
TL1A: a new potential target in the treatment of inflammatory bowel disease
.
Curr Drug Targets
.
2021
;
22
(
7
):
760
9
.
18.
Danese
S
,
Neurath
MF
,
Kopoń
A
,
Zakko
SF
,
Simmons
TC
,
Fogel
R
,
.
Effects of Apremilast, an oral inhibitor of phosphodiesterase 4, in a randomized trial of patients with active ulcerative colitis
.
Clin Gastroenterol Hepatol
.
2020
;
18
(
11
):
2526
34.e9
.
19.
Neufert
C
,
Neurath
MF
,
Atreya
R
.
Rationale for IL-36 receptor antibodies in ulcerative colitis
.
Expert Opin Biol Ther
.
2020
;
20
(
4
):
339
42
.
20.
Privitera
G
,
Pugliese
D
,
Lopetuso
LR
,
Scaldaferri
F
,
Neri
M
,
Guidi
L
,
.
Novel trends with biologics in inflammatory bowel disease: sequential and combined approaches
.
Therap Adv Gastroenterol
.
2021
;
14
:
175628482110066
.
21.
Amoroso
C
,
Perillo
F
,
Strati
F
,
Fantini
MC
,
Caprioli
F
,
Facciotti
F
.
The role of gut microbiota biomodulators on mucosal immunity and intestinal inflammation
.
Cells
.
2020
;
9
(
5
):
1234
.
22.
Tan
XY
,
Xie
YJ
,
Liu
XL
,
Li
XY
,
Jia
B
.
A systematic review and meta-analysis of randomized controlled trials of fecal microbiota transplantation for the treatment of inflammatory bowel disease
.
Evid Based Complement Alternat Med
.
2022
;
2022
:
8266793
.
23.
Yang
M
,
Gu
Y
,
Li
L
,
Liu
T
,
Song
X
,
Sun
Y
,
.
Bile acid-gut microbiota axis in inflammatory bowel disease: from bench to bedside
.
Nutrients
.
2021
;
13
(
9
):
3143
.
24.
Agus
A
,
Planchais
J
,
Sokol
H
.
Gut microbiota regulation of tryptophan metabolism in health and disease
.
Cell Host Microbe
.
2018
;
23
(
6
):
716
24
.
25.
Cao
Y
,
Su
Q
,
Zhang
B
,
Shen
F
,
Li
S
.
Efficacy of stem cells therapy for Crohn’s fistula: a meta-analysis and systematic review
.
Stem Cell Res Ther
.
2021
;
12
(
1
):
32
.
26.
Iwata
K
,
Mikami
Y
,
Kato
M
,
Yahagi
N
,
Kanai
T
.
Pathogenesis and management of gastrointestinal inflammation and fibrosis: from inflammatory bowel diseases to endoscopic surgery
.
Inflamm Regen
.
2021
;
41
(
1
):
21
.
27.
Bonaz
B
.
Is-there a place for vagus nerve stimulation in inflammatory bowel diseases
.
Bioelectron Med
.
2018
;
4
(
1
):
4
.
28.
Gunther
C
,
Rothhammer
V
,
Karow
M
,
Neurath
M
,
Winner
B
.
The gut-brain Axis in inflammatory bowel disease-current and future perspectives
.
Int J Mol Sci
.
2021
;
22
(
16
):
8870
.
29.
Castro-Dopico
T
,
Colombel
JF
,
Mehandru
S
.
Targeting B cells for inflammatory bowel disease treatment: back to the future
.
Curr Opin Pharmacol
.
2020
;
55
:
90
8
.
30.
Saez
A
,
Gomez-Bris
R
,
Herrero-Fernandez
B
,
Mingorance
C
,
Rius
C
,
Gonzalez-Granado
JM
.
Innate lymphoid cells in intestinal homeostasis and inflammatory bowel disease
.
Int J Mol Sci
.
2021
;
22
(
14
):
7618
.