Skip to main content
Journal of Crohn's & Colitis logoLink to Journal of Crohn's & Colitis
. 2018 Feb 23;12(Suppl 2):S641–S652. doi: 10.1093/ecco-jcc/jjx145

Chemokines and Chemokine Receptors as Therapeutic Targets in Inflammatory Bowel Disease; Pitfalls and Promise

Palak J Trivedi 1,2,3,4, David H Adams 1,2,
PMCID: PMC6104621  PMID: 30137309

Abstract

The principal targets for anti-chemokine therapy in inflammatory bowel disease (IBD) have been the receptors CCR9 and CXCR3 and their respective ligands CCL25 and CXCL10. More recently CCR6 and its ligand CCL20 have also received attention, the expression of the latter in enterocytes being manipulated through Smad7 signalling. These pathways, selected based on their fundamental role in regulating mucosal immunity, have led to the development of several therapeutic candidates that have been tested in early phase clinical trials with variable clinical efficacy. In this article, we appraise the status of chemokine-directed therapy in IBD, review recent developments, and nominate future areas for therapeutic focus.

Keywords: Eldelumab, Mongersen, Vercirnon

1. Introduction

Effective immune surveillance involves the continuous patrolling of tissues by leukocytes able to respond to foreign antigens and thereby mount protective immune responses. This process is dependent upon the ability of leukocytes to reach tissues via the blood and then traverse vascular beds in response to local signals presented by endothelial cells. An initial capture step, classically mediated by selectins, brings a leukocyte flowing in the blood into contact with endothelium.1 This induces tethering or rolling, an inherently unstable transition state, that is converted into firm leukocyte arrest by chemokine-mediated triggering of leukocyte integrins to bind immunoglobulin superfamily members. Once stable adhesion is achieved, chemokines are able to provide directional migratory signals that direct leukocyte crawling within, through, and beyond the vasculature towards targets within tissue.

Chemokines encompass a group of small (8–12 kDa) heparin-binding cytokines with the ability to induce leukocyte migration.2 Virtually any cell type can express and secrete chemokines upon stimulation, including endothelial, epithelial, and stromal cells as well as leukocytes. Chemokines can operate as a soluble gradient to induce chemotaxis or after being immobilised on, for instance, the endothelial glycocalyx via glycosaminoglycan binding or within the extracellular matrix. This process allows multiple chemokines to be sequestered, often in multimeric forms, at sites of inflammation and prevents them from being ‘washed away’, facilitates chemokine localisation by leukocytes, and increases their affinity toward cognate receptors.3 Immobilised chemokines can also persist at higher local concentrations than the freely diffusible state,2 while enhancing the function of one another in a phenomenon referred to as chemokine cooperativity.4

Classification of chemokines can be based on structure and the position of conserved cysteine residues (Table 1), or functionally as to whether they play a role predominantly in the context of inflammation, or constitutively expressed under homeostatic conditions. Inflammatory chemokines are released by a wide range of cell types in response to pro-inflammatory cytokines, tissue injury, or contact with pathogens. Such chemokines are secreted early in response to pathogen recognition receptor activation on epithelial, stromal, and immune cells,2 to recruit the first wave of innate immune effectors including neutrophils, monocytes, natural killer (NK cells), and NKT cells. Chemokines also promote the migration of activated dendritic cells (DC) into secondary lymphoid tissues to activate adaptive immunity and the recruitment of T effectors (Teff) to the site of injury. Regulatory T cells (Treg) are recruited in a similar manner, and the balance between effector and regulatory cell recruitment will determine the outcome of the immune response. Homeostatic chemokines are constitutively expressed in lymphoid and non-lymphoid tissues where they mediate the physiological trafficking and positioning of cells involved in antigen sampling and immune surveillance. Functional distinctions are not absolute, and chemokines previously thought to be homeostatic are often induced and up-regulated at sites of chronic inflammation.2

Table 1.

Chemokine-mediated recruitment in the small bowel and colon.

Site Constitutively expressed Increased in response to inflammation
Small bowel CCR6 – CCL20
CCR9 – CCL25
CCR10 – CCL28
CXCR1 – CXCL5/6/8
CXCR2 – CXCL1/2/5/6
CXCR6 – CXCL16
CX3CR1 – CX3CL1
α4β7 – MAdCAM-1 (increased)
VAP-1
α4β1 – VCAM-1
αLβ2 – ICAM-1
CCR2 – CCL2/7/8
CCR6 – CCL20
CCR5 – CCL3/4/5/8
CXCR3 – CXCL9/10/11
CX3CR1 – CX3CL1 (increased)
Colon CCR5 – CCL3/4/5/8
CCR6 – CCL20
CCR10 – CCL28
CX3CR1 – CX3CL1
CCR2–CCL2/7/8
CCR3 – CCL11 (UC)
CCR4 – CCL17 (murine colitis)
CCR6 – CCL20
CCR9 – CCL25 (UC)
CXCR1 – CXCL5/6/8
CXCR2 – CXCL1/2/5/6
CXCR3 – CXCL9/10/11

UC, ulcerative colitis.

In the gut, different intestinal sites show distinct patterns of chemokine expression, which serves to compartmentalise leukocyte recruitment, thereby regulating regional immunity. For example, under non-inflammatory conditions the chemokine CCL25 is expressed by small intestinal but not colonic epithelium, where it supports the recruitment of T cells and B cells expressing its receptor CCR9;5 whereas CCR10 and G protein-coupled receptor (GPR)-15 are involved in recruiting IgA+ plasmablasts and T cells, respectively, to the colon.6–8 In response to inflammation however, other chemokines can predominate; for instance interferon-induced CXCL10 can recruit CXCR3-expressing effector cells to the small bowel.9 Conversely in active ulcerative colitis (UC), CCL25 which was previously believed to be involved in small bowel immune homeostasis becomes detectable in the human colon, with expression levels correlating with disease activity.10 These findings suggest that expression of chemokines shape regional differences in immune composition along the normal intestine and determine the nature of inflammation in disease.11–14

2. CCR9 and CCL25

A greater understanding of the role of chemokines in coordinating the development and maintenance of mucosal immunity, in addition to the nature of pathological inflammation in the gut, has suggested therapeutic strategies for targeting leukocyte recruitment pathways in inflammatory bowel disease (IBD).

The chemokine CCL25 and its receptor CCR9 are essential for optimal mucosal immune development and function, with the latter being expressed by 58–97% of lymphocytes imprinted with gut-tropism.15–18 Activation of CCR9 by CCL25 induces pro-migratory responses, including the activation of α4β1 and α4β7 integrins to bind vascular cell adhesion molecule (VCAM)-1 and mucosal addressin cell adhesion molecule (MAdCAM)-1, respectively, on mucosal vessels.19,20 CCR9-CCL25 interactions shape the gut lamina propria and the development of gut cryptopatches and the intra-epithelial lymphoid compartment through recruitment of CD4 and CD8 T cells, IgA-secreting B cells, and subsets of dendritic cells.21–26 However, whereas lymphocytes derived from knockout animals are hampered in their ability to enter the gut in competitive homing experiments,22Ccr9–/– mice exhibit only a mild phenotype under basal conditions with only modest reductions in numbers of intraepithelial lymphocytes (IELs),27 suggesting that T cells can enter the intestine through CCR9-independent as well as dependent mechanisms. This is particularly the case in the context of inflammation when other pathways including those involving CXCR3 play a more dominant role.9

The only known ligand for CCR9 is CCL25, expression of which is largely restricted to thymic and small bowel epithelium under normal conditions, although in response to inflammation, CCL25 is also detected in the colon and liver.10,28 Early growth response protein (EGR)-1 and caudal-related homeobox transcription factor (CDX) have been reported to increase CCL25 transcription in endothelial cells, but the role of conventional inflammatory stimuli such as tumour necrosis factor alpha (TNFα) or interferon (IFN)-γ remain controversial.11,17 Murine studies report a gradient of CCL25 expression throughout the normal gut, with highest tissue levels detected proximally in the duodenum and lowest in the ileum and very little if any detected in the non-inflamed colon.22 Furthermore, there appears to be differential dependence on CCR9 and CCL25 for recruitment to the lamina propria (LP) and the intestinal epithelium dependent (in part) on cell type.9,15

Studies of the CCR9-CCL25 axis in IBD have given different results depending on the model studied and the time course of inflammation. Neutralising antibodies to CCL25 reduced the adhesion of adoptively transferred intestinal lamina propria lymphocytes (LPL) and intraepithelial lymphocytes (IEL) to TNF-activated small intestinal post-capillary venules, but not to colonic vessels.17 In the SAMP1/YitFc model of murine ileitis, a significantly greater proportion of CD4+ and CD8+ lymphocytes are CCR9+ compared with wild-types, and histological indices of early but not late intestinal inflammation are lessened following administration of antibodies targeting the CCR9-CCL25 axis.29 The ileitis seen in Rag2-deficient mice is also attenuated in the absence of CCR9, although these findings conflict with the TnfΔARE model of IBD, wherein CCR9 deficiency is associated with a reduction in gut Treg and persistent intestinal inflammation.30–32 Consistent with the latter, CCR9 can be expressed by subsets of Treg and immunosuppressive DC populations.25,30,31,33 Indeed, following adoptive transfer of CD4+ T-cells, Ccr9-/-Rag2-/- mice develop small bowel inflammation, suggesting that non-T cell CCR9+ populations are partially responsible for maintaining tolerance in this model. Furthering the argument for an immune modulatory role of CCR9 cells, Wurbel et al. reported that CCR9-CCL25 interactions are critical for protective responses against large intestinal inflammation in acute and chronic models of dextran sulphate sodium (DSS)-induced colitis.34,35

There are several potential explanations for these different findings. Imprinting of CCR9 is retinoic acid dependent, and in the presence of transforming growth factor (TGF)-β is associated with induction of FoxP3. Indeed, CD4+ Treg preferentially express CCR9 when compared with effector counterparts in the TnfΔARE model, and CCR9+ CD8+ subsets can inhibit of proliferation of CD4+ T-cells in vitro. Wermers et al. report highly concordant expression of CCR9, FoxP3, and CD103 in mice, suggesting that CCR9 is required for T cell-mediated regulation of chronic ileitis.30 Moreover, CCR9-CCL25 interactions are critical for establishing early thymocyte colonisation and selective maturation of T cells that are tolerant to self-antigens (hence the synonym for CCL25: thymic expressed chemokine; TECK).26 However, clinical studies illustrate increased numbers of CCR9+ T cells in blood from individuals with small bowel inflammation;36 and CCR9+ T cells isolated from mesenteric lymph nodes (MLN) of patients with Crohn’s disease are more activated and show enhanced IFN-γ and interleukin (IL)-17 secretion when compared with lymphocytes extracted from normal MLN.36,37

Collectively, these findings imply that although congenital absence has potentially harmful consequences, targeted therapy against CCR9 or CCL25 may be beneficial in the prevention or treatment of small bowel inflammation. This hypothesis is supported by clinical trial data in which clinical and endoscopic indicators of small-intestinal Crohn’s disease improved following treatment with the CCR9-antagonist Vercirnon.38

Whereas a role for lymphocyte CCR9 is accepted in the small bowel, controversy has existed over the roles of CCR9 and CCL25 in the colon.37,39 Murine studies suggest that under non-inflammatory conditions, Ccl25 gene expression is restricted to the small intestine in uninjured wild-type mice9,24,40 and that is also the case in some IBD models, including the Samp1/YitFc model of ileitis,29 the TnfΔARE model of small bowel Crohn’s disease,30 and spontaneous ileitis in Rag2-/- mice.31 Studies in pigs again suggest differential expression along the small intestine which is dependent on the microbiota.41 Nevertheless, colonic CCL25 transcripts have been reported in spontaneous murine models of colitis,42 as well as colonic inflammation induced by dextran sulphate sodium34,35 and oxazolone.43

In humans there has been even more variability in findings. In some studies, fewer than 10% of cultured T cells isolated from colonic lymphoid tissue expressed CCR9 irrespective of inflammatory status, and lower frequencies of circulating CD4+ T cells expressed CCR9 in patients with colonic versus small bowel Crohn’s disease.39 Consistent with this, northern blotting was able to detect CCR9 mRNA in small bowel but not colonic tissue.16 In other studies, increased CCR9 expression gauged by median fluorescence intensity (MFI) has been reported on subsets of CD3+ T cells and CD14+ monocytes in the blood of a small number of UC patients.44,45 IL-17+ CCR9+ γΔ T cells have also been implicated in the mediation of extraintestinal inflammation in humans, and are increased in the blood of patients with active UC.46,47 Recent studies from our group and others have extended upon these observations, to confirm elevated serum levels of CCL25 in patients with UC48 and, most significantly, a strong positive correlation between CCL25 gene expression in the colonic mucosa and both the Mayo endoscopic sub-score and mucosal TNFα levels in ulcerative colitis patients.10 This was validated by CCL25 protein expression in the inflamed colon and a high frequency of CCR9+ colon-infiltrating effector T cells.10,42 Such findings may contribute to the increased colonic cancer risk in colitis which relates to inflammatory burden, given the ability of CCR9-CCL25 interactions to mediate colonic tumour growth, invasion, and metastasis.49

2.1. Therapeutic targeting of CCR9/CCL25

The biological role of the CCR9-CCL25 axis and the evidence implicating CCR9, and CCL25 in gut inflammation in both human and murine IBD, suggest this is an appropriate pathway to target therapeutically.10,29,39,50,51 However, the involvement of CCR9 and CCL25 in immune regulation and tolerance raises valid concerns that under some circumstances inhibition could be detrimental. Studies of immunoblockade of CCR9 and CCL25 in the Samp1/Yit model, showing an effect on early but not late inflammation, suggest that timing of intervention may also be important, with the possibility of a role in preventing or inducing rather than maintaining remission.29 The only orally bioavailable CCR9 antagonist, CCX282-B (Chemocentryx, USA)52 is a potent inhibitor of CCR9+ T cell-mediated chemotaxis in vitro, and results in near complete protection against ileitis in the TnfΔARE model.34 Additionally CCX282-B attenuated colitis in oxazolone-treated animals.43

The encouraging results from preclinical models have thus led to clinical study in IBD. The PROTECT-1 phase IIb trial randomly allocated patients with Crohn’s disease to placebo or one of three treatment dosages, organised into: an induction phase (induction of clinical response at Weeks 8 and 12); an active, open-label study phase (4 weeks) in which all eligible participants received CCX282-B at 250 mg twice daily; and a maintenance period in which patients who showed clinical response during the active phase were re-randomised to receive placebo or CCX282-B at a dose of 250 mg twice daily. The induction phase of PROTECT-1 failed to attain its primary endpoint of a significant reduction in the CD Activity Index (CDAI) of 70 points at Week 8 of treatment, although a greater number of patients on the 500 mg daily dose regimen achieved clinical response compared with placebo (Table 2). The fact that response rates were similar between placebo and the 250 mg treatment groups suggests this dose may be suboptimal, although 41% of subjects on CCX282-B were in corticosteroid-free remission compared with 28% on placebo (P = 0.041). The active treatment phase of PROTECT-I gave all patients an opportunity to receive active treatment, following which patients who showed a clinical response were re-randomised to receive placebo or CCX282-B (250 mg twice daily) as maintenance therapy. Remission was achieved in 47% of patients on CCX282-B treatment compared with 31% of those on placebo (P = 0.012), together with a reduction in median overall CDAI.

Table 2.

Clinical trials of anti-CCR9 therapy in IBD.

Compound Entry criteria Cohort size Organisation Primary endpoint Potential expedients
A) Phase II (PROTECT)
CCX282-B (oral) Inclusion:
Moderate to severe Crohn’s disease
- CDAI 250450
- Fasting CRP > 7.5 mg/L
Exclusion:
- Fluctuating dosages of immunosuppression 4 weeksa
- > 20 mg prednisolone (or equivalent) 4 w previouslya
- Anti-TNFα or anti-α4 integrin treatment 12 w previouslya
- Placebo twice daily (n = 144)
- 250 mg CCX282-B once daily (n = 98)
- 250 mg CCX282-B twice daily (n = 96)
- 500 mg CCX282-B once daily (n = 97)
Induction phase; 12 w
- Measuring CR at 8 and 12 w
Active (open-label) period; 4 w
- Eligible participants received CCX282-B (250 mg twice daily)
Maintenance period; 36 w
- Subjects with clinical response following active period re-randomised to receive placebo or CCX282-B (250 mg twice daily)
Induction period
CDAI to ≤ 70 by 8 w
- Placebo: 49%
- 250 mg o.d.: 52%
- 250 mg b.d.: 48%
- 500 mg: 60%
P = N.S.; all comparisons
Maintenance period
Sustained CDAI ≤.70
- Placebo: 31%
- 250 mg b.d.: 47%
P = 0.012
CDAI ≤ 70 by 12 w
- Placebo vs 500 mg: 47% vs 61%; P = 0.039
CDAI decrease by ≥ 100 points by 12 w
- Placebo vs 500 mg: 40% vs 55%; P = 0.029
Mean change in CDEIS at 12 w
- Placebo: - 3.0
- 250 mg o.d.: - 8.7
- 250 mg b.d.: 2.0
- 500 mg: - 10.8
P = 0.049 for 500 mg vs placebo
B) Phase III (SHIELD-1)54
CCX282-B (oral): - Vercirnon Inclusion:
Moderate to severe Crohn’s disease
- CDAI 220450
- Active inflammationb
- Inadequate response to immunosuppression
Exclusion:
- Fluctuating dosages of immunosuppressiona
- > 20 mg prednisolone (or equivalent)
- No concurrent dependence on anti-TNFα therapy during trial
- EC, abdominal or pelvic fistulas with abscess
- Placebo once daily (n = 203)
- 500 mg CCX282-B once daily (n = 203)
- 500 mg CCX282-B twice daily (n = 202)
Induction phase only CDAI dec. ≥100 by 12 w
- Placebo: 25%
- 500 mg o.d.: 27%
- 500 mg b.d.: 27%
P = N.S.; all comparisons
CDAI dec. ≥ 100 by 12 w in patients with colitis
- Placebo: 13%
- 500 mg o.d.: 25%
- 500 mg b.d.: 29%
p < 0.05; for 500 mg b.d. vs placebo
C) Phase II55,56
CCR9-targeted leukapheresis Inclusion:
UC; moderate to severe activity
- Mayo score 6–11
- Prednisolone ≤ 20 mg daily (stable dose at least 2 w)
- Naïve to anti-TNFα antibodies
- Placebo (n = 9)
- Treatment (n = 14)
Alternate day leukapheresis
(5 sessions; 10 days)
Reduction of CCR9 + HLA-DR hi monocytes
- 11% to 12% (P = 0.469) vs 14% to 10% (P = 0.039); placebo vs treatment groups, respectively
Dec. in overall Mayo score:
8.0 to 6.3 (P = 0.125) vs 8.8 to 5.7 (P = 0.016);
placebo vs treatment groups, respectively

IBD, inflammatory bowel disease; CRP, C-reactive protein; EC, enterocutaneous; TNF, tumour necrosis factor; CDAI,Crohn’s Disease Activity Index; CDEIS, Crohn’s Disease Endoscopic Index of Severity; w, weeks; m, months; UC, ulcerative colitis; N.S., not significant; dec., decrease; o.d., once daily; b.d., twice daily.

aBefore randomisation.

bBy endoscopic assessment or a CRP ≥.3 mg/L or faecal calprotectin > 200 µg/g in stool.

Following the results of the phase IIb study, CCX282-B was reformulated and renamed (initially GSK1605786 Formulation A, and later Vercirnon) and studied in a phase III double-blind randomised placebo-controlled trial. SHIELD-1 was conducted over 162 centres in 23 countries between 2010 and 2013, and included patients with a CDAI of 220–450, evidence of active Crohn’s disease endoscopically, or with elevated inflammatory markers, and if they had failed corticosteroid or immunosuppressive therapy.54 No significant difference in remission rates (defined as a reduction in CDAI of ≥ 100 points by Week 12) was observed between either of the treatment arms compared with placebo (Table 2).

The reasons for the discrepancy in outcome between the Phase IIb PROTECT-1 study and the Phase III SHIELD-1 study are unclear but could reflect differences in the patients recruited; previous anti-TNFα treatment was reported in 69% of patients comprising SHIELD-1 versus 26% in PROTECT-1. Another plausible reason for failure in the phase III Vercirnon study may stem from differences in clinical trial design. For instance, PROTECT-I comprised a 12-week placebo-controlled lead-in followed by 4 weeks of open-label treatment.

It is also possible that 12 weeks is insufficient time for the drug to deplete the lamina propria of effector populations, because previously recruited cells may be retained for several weeks before they die in situ. This would be consistent with the positive effects on maintenance of remission reported in PROTECT-1.51 Although results from the maintenance phase of the SHIELD programme have not been published, the aim of SHIELD-4—a randomised double-blind induction study for patients with moderate-to-severe Crohn’s disease—was to treat patients with one of two dose regimens of Vercirnon, and enrol clinical responders to a Phase III maintenance clinical trial (SHIELD-2).55 The primary endpoint of the trial was the proportion of patients with a Crohn’s disease activity index (CDAI) ≥ 100-point response (100-point decrease in CDAI score) at Week 12. The secondary endpoint was the proportion of patients in clinical remission (CDAI < 150) at Week 12. Adult patients with a baseline CDAI of 220 to 450, C-reactive protein (CRP) ≥ 3 mg/L or faecal calprotectin > 200 μg/g were randomised to receive either 500 mg four times daily (q.d.s.) or 500 mg twice daily (b.d.) Vercirnon for 12 weeks. Baseline CDAI was 323 (± 56) with a range of 220–450. In those who completed the trial before premature study termination (n = 118), the CDAI ≥ 100-point response at Week 12 was 56% and 69% in the 500-mg q.d.s. and in the 500-mg b.d. groups, respectively, similar to the PROTECT-1 data. Within the same observed population, rates of remission (defined as CDAI < 150) at Week 12, were 26% and 36%, respectively. To the authors’ knowledge, no further data relating to the SHIELD-2 maintenance study are available.

The failure to reach primary endpoints in the phase III clinical trial has delayed further development of anti-CCR9 therapy in IBD. However, pre-specified subgroup analysis in SHIELD-1 did show a higher clinical response for Vercirnon treatment compared with placebo, specifically in patients with colonic disease: CDAI response at Week 12 of 25.4%, 28.8%, and 13% in the once daily, twice daily, and placebo groups, respectively.57 Although the investigators state a lower placebo response rate as one factor that could explain this observation, it is also possible that this was a real effect consistent with the fact that CCR9+ T cell frequencies do not correlate with inflammatory activity in the small bowel, whereas they correlate strongly with disease activity in the colon.10 The current evidence therefore indicates that a randomised placebo-controlled trial of Vercirnon, specifically for patients with ulcerative colitis, is justified.

In addition to circulating gut homing T cells, subsets of pro-inflammatory monocytes also express high levels of CCR9,44 and their frequencies in the circulation correlate with clinical activity in some IBD patients. A recently published study reports the successful treatment of a patient with refractory UC, by depleting CCR9 cells from the blood using leukapheresis. Clinical and endoscopic remission was associated with a greater reduction in CCR9 expression on circulating T cells and monocytes.45 This led to a subsequent double-blind placebo-controlled trial of leukapheresis to deplete CCR9+ monocytes in ulcerative colitis, which was associated with improvement in the endoscopic Mayo score compared with placebo (Table 2).56

3. CXCR3 and CXCL10

CCL25 and CCR9 drive gut specific leukocyte recruitment under homeostatic and inflammatory conditions, but in the context of IBD their role can be overshadowed by inflammatory chemokines.11,12 Human LPL and IEL express the chemokine receptors CCR2, CCR5, and CXCR3 that play an important role during leukocyte recruitment to the inflamed intestine.11 Under normal circumstances the colonic epithelium expresses low levels of the chemokine ligands for CXCR3 (namely CXCL9, CXCL10, and CXCL11). However, the expression of one of these, CXCL10, which is an IFN-γ inducible chemokine, increases markedly in colitis.57,58 CXCR3 is expressed at high levels on tissue-infiltrating, activated T cells in a number of inflammatory disorders, and mucosal biopsies taken from patients with active UC and Crohn’s disease demonstrate increased frequencies of CXCR3+ cells associated with strong expression of CXCL10.59–61 Of note, the latter has been implicated in the induction of Th1 responses across inductive sites such as MLNs, as well as the promotion of effector cell recruitment to inflamed gut tissue. Zhao et al. have also demonstrated that the CXCL10/CXCR3 axis mediates monocyte activation and drives tissue inflammation by innate immunity distinct from the conventional paradigm as a regulator of immune cell recruitment.62 In contrast to CXCL9 and CXCL11, CXCL10 alone was necessary and sufficient for IFN-γ primed monocytes to promote pro-inflammatory cytokine release, indicating a non-redundant role of CXCL10 in inflammation. Moreover, CXCL10 signalling was able to synergise with lipopolysaccharide to potentiate much greater amounts IL-12 and IL-23 release by monocytes.

A therapeutic role targeting CXCR3 is supported by studies in the Il-10/ model of IBD. Frequencies of CXCR3+ cells increase in mesenteric lymph nodes and Peyer’s patches associated with Th1-mediated injury, and inhibition of CXCL10 attenuates the rate and intensity of colitis and intestinal inflammation.63,64 CXCR3-deficient mice also develop a much milder colitis in response to dextran sulphate sodium. This is associated not only with reduced inflammation but also changes in epithelial cell proliferation, suggesting direct effects of CXCR3 chemokines on epithelial cells in the gut.65,66

3.1. Clinical trials targeting CXCR3/CXCL10

Eldelumab (BMS-936557, previously named MDX-110067) is a fully human monoclonal antibody to CXCL10 which has been investigated for the treatment of moderate to severe UC in a phase IIa multicentre, double-blind randomised placebo-controlled trial.68 Despite a trend toward increased clinical response at Day 57, as evidenced by clinical remission and mucosal healing, there were no significant differences in the primary or secondary endpoints between treatment versus placebo groups (Table 3). However, the study yielded a notable exposure-response relationship, with Eldelumab clinical response and mucosal healing rates being greatest in patients with the highest trough concentrations.68 We have observed similar outcomes in a trial of NI-0801, a monoclonal antibody against CXCL10, in patients with the immune-mediated liver disease primary biliary cholangitis (PBC), which suggests that the high production rate of CXCL10 by inflamed tissues makes it difficult to achieve therapeutic drug levels and sustained neutralisation of the chemokine in vivo (K. de Graaf et al; submitted).

Table 3.

Clinical trials targeting CXCR3/CXCL10 in IBD.

Compound Entry criteria Cohort size and organisation Primary endpoint Potential expedients
A) Phase IIa68
BMS-936557 (IV) Inclusion:
UC; active flare despite 5-ASA or immunosuppressiona
- Mayo score 6–10
- Mayo endoscopic sub-score ≥ 2
Exclusion:
- Fulminant UC
- Anti-TNFα therapy in preceding 8 w
- Placebo (n = 54)
- BMS-936557 10 mg/kg (n = 55)
Induction period; 8 w
Dec. in Mayo score ≥ 3 at 57 d + dec. in rectal bleed score ≥ 1 or absolute bleed score ≤ 1
- Placebo 35%
- 10 mg/kg: 53%
P = 0.083
Attaining primary endpoint by dose-response (drug trough drug values)
- Placebo: 37%
- 26–79 µg/mL: 53%
- 79–105 µg/mL: 63%
- 108–235 µg/mL: 88%
Mucosal healing
- Placebo: 35%
- 26–79 µg/mL: 29%
- 79–105 µg/mL: 44%
- 108–235 µg/mL: 69%
B) Phase IIb69
BMS-936557 (IV)
- Eldelumab
Inclusion:
UC; moderate to severe activitya
- Mayo score ≥ 6
- Mayo endoscopic subscore ≥ 2
- Inadequate response to existing medical therapy; any type
Exclusion:
- As above
- Placebo (n = 83)
- Eldelumab 15 mg/kg (n = 84)
- Eldelumab 25 mg/kg (n = 85)
Induction period; 11 w
Mayo score < 2 by 11 w + no individual component > 1
- Placebo: 9.6% vs
- 15 mg/kg: 13.1%; P = 0.515)
- 25 mg/kg: 17.6%; P = 0.158)
Subgroup analysis of primary endpoint and mucosal healing greater compared with placebo in:
(A) anti-TNFα naïve; and
(B) immunomodulator-exposed subgroups
C) Phase IIa70
BMS-936557 (IV)
- Eldelumab
Inclusion:
Moderate to severe Crohn’s disease
- CDAI 220 to 450
- CRP ≥ 5 mg/L, or
- Faecal calprotectin > 250 µg/g, or
- SES-CD of 2 to 3
- Insufficient response/intolerance to
prednisolone (≥ 40 mg/d for 2 w (or equivalent) or immunomodulators
Exclusion:
- Penetrating disease or fibrotic stenosis
- Surgery within 6 m of screening
- Anti-TNFα therapy in preceding 8 w
- Placebo (n = 40)
- Eldelumab 10 mg/kg (n = 40)
- Eldelumab 20 mg/kg (n = 41)
1) Steady-state plasma concentration for induction- No statistically significant exposure-remission relationship with Eldelumab at 11 w
OR: 1.06 (90% CI 0.96–1.18)
2) Efficacy: CDAI < 150 by 11 w; treatment differences
- 10 mg/kg vs placebo: 9%
- 20 mg/kg vs placebo: 13%
Composite endpoint analysis showed increased response rate for both doses vs placebo

5-ASA, 5-aminosalicylic acid; CDAI, Crohn’s Disease Activity Index; CI, confidence interval; d, day; dec., decrease; IV, intravenous; m, month; SES-CD, simplified endoscopic score of Crohn’s disease; TNF, tumour necrosis factor; OR, odds ratio; UC, ulcerative colitis; w, week, IBD, inflammatory bowel disease; CRP, C-reactive protein.

a2 w before drug administration and of ≥ 6 m duration.

Based on the phase IIa data, a 100-µg/mL trough value was considered the ‘target for efficacy’ in a phase IIb dose-ranging induction study in UC (Table 3).69 The proportion of patients achieving clinical remission by Week 11 (primary endpoint) was no different between placebo versus either the Eldelumab 15 mg/kg or the Eldelumab 25 mg/kg group. This was despite 79% and 97% of patients in the treatment groups maintaining a trough plasma concentration of 100 µg/mL, respectively. Although a numerically higher proportion of patients in the Eldelumab treatment arms achieved clinical response, rates of mucosal healing were similar when compared with placebo. In subgroup analysis, greater efficacy was observed in Eldelumab-treated patients who were anti-TNFα naïve,69 and for the 25 mg/kg subgroup who were receiving concurrent immunomodulators.

Results from a phase IIa trial in Crohn’s disease were also recently published.70 The primary objective was to demonstrate dose response; no exposure-remission relationship was seen with Eldelumab, although a trend towards clinical and endoscopic efficacy was observed. There was no statistically significant relationship between trough drug levels and clinical remission by Week 11, suggesting that at the doses of 10 and 20 mg/kg studied, clinical efficacy had been maximised.70 Moreover, there were no significant differences in the rate of remission or CDAI response rates between either the 10 mg/kg or 20 mg/kg treatment groups versus placebo (Table 3). As with previous clinical trials in IBD, clinical response was more pronounced for treatment subgroups who were anti-TNFα naïve or receiving concurrent immunomodulatory treatment.70

This was one of the first placebo-controlled, prospective studies with central endoscopy reading for patients with moderate to severe Crohn’s disease. This follows lessons from the earlier UC trial of Eldelumab as well as others, in which high but variable mucosal response rates were seen with placebo.69 Treatment with both Eldelumab doses resulted in mean reductions in the endoscopic degree of inflammatory activity measured by the simplified endoscopic score in Crohn’s disease (SES-CD): 3.44, –3.57, and –0.94, for 10 mg/kg, 20 mg/kg, and placebo groups, respectively).70 Unlike with clinical response, endoscopic activity following Eldelumab did not differ in those patients with previous exposure to biologic therapy,70 neither did this vary according to baseline severity of endoscopic disease. Although the trial was not powered to test efficacy endpoints, given the lack of agreement between clinical and endoscopic findings a composite post hoc endpoint analysis was conducted. Therein, response was determined if ≥ 30% improvement in clinical symptoms occurred together with either: (i) a 3-point decrease in SES-CD score; or (ii) an absolute SES-CD score of zero. This did show significant treatment differences between Eldelumab and placebo for both the biologic naïve and biologic failure subgroups, albeit with very low placebo response rates.

The above studies all indicate that inhibition of the CXCR3-CXCL10 pathway appears to be safe but fails to demonstrate overall efficacy. However, the suggestion of responses in specific patient populations are encouraging. It is possible that IBD results in the activation of several inflammatory pathways and thus inhibition of CXCR3 alone is insufficient to induce clinical responses. However further attempts to define patients based on clinical or endoscopic features, or perhaps more pertinent immunophenotypes, might allow treatment to be targeted to patient subgroups more likely to experience therapeutic benefit.

4. CCR6 and CCL20

The ability of intestinal epithelial cells to secrete cytokines and chemokines in response to commensal bacteria, pathogens, or injury allows them to play an active role in shaping the nature of the local immune response.71 IL-17-secreting innate lymphoid cells and Th17 CD4 T cells are abundant in the gut, and the epithelium-secreted chemokine CCL20 plays a major part in attracting IL-17-secreting cells that express its sole receptor CCR6.72 Although CCL20 is secreted by the epithelium in response to inflammation, it is also constitutively expressed by the follicle-associated epithelium overlying Peyer’s patches and isolated lymphoid follicles through which it contributes to the homeostatic development and maintenance of the mucosal immune system. In Ccr6 knockout mice, there is a failure of full development of mucosal inductive sites leading to decreased IgA production to oral antigens. Thus, CCL20 functions as both an inflammatory and a homeostatic chemokine.73

Following epithelial damage, infection, or changes in the microbiota, there is a shift in local mucosal immune responses away from regulation and towards pro-inflammatory Type-1 or Type-17 responses, driven in part by increased production of CCL20 from inflamed epithelium.12,74–78 A role for CCL20 in IBD pathogenesis is also supported by the findings of genome association and gene expression studies,79,80 and the fact that intestinal CCL20 mRNA expression is up-regulated several-fold during active inflammation,77,78 enhanced through stimulation of toll-like receptor (TLR)-1 and TLR3.77,81 Murine models have also shown that Th17-cells can be actively redirected from the blood to the small bowel in response to CCL20,82 and several studies report that IL-17A in turn up-regulates CCL20 release from the intestinal epithelium, resulting in a positive feedback circuit that drives ongoing inflammation.82 Furthermore, neutralisation of CCL20 reduces T cell recruitment and attenuates colitis in the 2,4,6-trinitrobenzene sulphonic acid (TNBS)-induced murine model.83

Persistent intestinal inflammation requires not only local pro-inflammatory signals such as TNFα that induces CCL20 secretion by epithelial cells,10 but also the presence of induced or constitutive defects in counter-regulatory mechanisms that allow inflammation to develop unchecked.84 One such mechanism involves TGF-β1, levels of which are reduced in CD and UC due to increased expression of the intracellular protein Smad7.85 TGF-β1 activity can be restored in isolates of lamina propria mucosal cells by treatment with an anti-sense oligonucleotide to Smad7 termed Mongersen (previously GED0301), the administration of which attenuates colitis in the TNBS mouse model.86 Smad7-transgenic animals do not develop an IBD phenotype under normal circumstances but exhibit more severe colitis in response to DSS than wild-types.87 Moreover, Smad7-overexpressing CD4+ T-cells induce severe colitis that is resistant to Treg-mediated rescue when transferred to immunodeficient mice.88 Marafini and colleagues have recently shown that CCL20 expression by colonic epithelial cells was inhibited by TGF-β1, but that this was overridden in epithelial cells that over-expressed Smad7.89 Moreover, Mongersen administered directly to the luminal side of mucosa down-regulated Smad7 in a dose-dependent manner, which correlated with the reduction in colonic CCL20 expression.89

4.1. Targeting Smad7 and the impact on CCL20 expression

In a small open-label and dose-finding Phase I study in Crohn’s disease,90,91 treatment with Mongersen was associated with a reduction in CDAI of > 70 points within 8 days across all three tested doses (40 mg, 80 mg, 160 mg; n = five participants per group). These results were swiftly followed by efficacy evaluation in a Phase II, multicentre, double-blind, placebo-controlled trial.92 A total of 166 active, steroid-dependent or steroid-resistant Crohn’s disease patients were assigned to receive one of three doses of Mongersen (10, 40, or 160 mg per day) or placebo daily for 2 weeks. The primary endpoint was the proportion of individuals who reached clinical remission at Day 15 (CDAI < 150) maintained for at least 2 weeks. Patients receiving the highest doses of Mongersen experienced significantly higher rates of remission than those treated with 10 mg or placebo (Table 4). Moreover, at the end of follow-up on Day 84, the percentage of patients who entered glucocorticoid-free remission was significantly greater in the 160 mg group versus placebo. Colonic Smad7 expression and CCL20 levels were decreased in treated patients, and serum values were also reduced in those achieving clinical response as early as Day 15 of treatment,89 suggesting that circulating CCL20 may be a useful biomarker to monitor early response, efficacy, and dose titration.

Table 4.

Summary data from the Mongersen Phase II clinical trial in Crohn’s disease (orally administered).

Phase II 92
Entry criteria Cohort size and organisation Primary endpoint Additional expedients
Inclusion:
- Moderate to severe Crohn’s disease (CDAI 220400)
- Disease located to the terminal ileum of right colon
- Steroid-dependent / refractory disease
Exclusion:
- Disease affecting the upper GI tract, proximal small intestine, transverse colon, or left colon.
- Stricturing disease
- Penetrating disease
- Extraintestinal disease
- Anti-TNFα therapy in preceding 90 da
- Placebo (n = 42)
- Mongersen 10 mg (n = 41)
- Mongersen 40 mg (n = 40)
- Mongersen 160 mg (n = 43)
Daily treatment for 2 w
- Clinical evaluation at 15 d, 28 d, and 84 d
CDAI < 150 at 15 d, and sustained until 28 d
- Placebo: 10%
- 10 mg: 12%
- 40 mg: 55%
- 160 mg: 65%
P < 0.001 for 40 mg or 160 mg groups vs either 10 mg or placebo groups
100-point CDAI response at 15d
- Placebo: 26%
- 10 mg: 22%
- 40 mg: 45%
- 160 mg: 65%
P < 0.05 for 40 mg or 160 mg groups vs either 10 mg or placebo groups
100-point CDAI response at 28d
- Placebo: 17%
- 10 mg: 37%
- 40 mg: 58%
- 160 mg: 72%
P ≤ 0.001 for 40 mg or 160 mg groups vs either 10 mg or placebo groups
P < 0.05 for 10 mg vs placebo groups

CDAI, Crohn’s disease activity index; d, days; TNF, tumour necrosis factor; w, weeks; GI, gastrointestinal.

aBefore the date of enrolment. However, patients with a worsening of disease (increase in CDAI ≥ 70 points) after 2 weeks into the trial were eligible for steroid, immunomodulator or anti-TNFα rescue.

A post hoc subgroup analysis of the Mongersen phase II data divided the cohort according to baseline disease duration (cut-point of 5 years), CDAI (cut-point of 260), and CRP level (cut-point of 3 mg/L).93 The authors found that clinical remission rates (CDAI < 150) at Weeks 2, 4, and 12 were significantly higher with Mongersen 160 mg/day and 40 mg/day compared with placebo for each disease duration and CRP subgroup. Furthermore, the rate of remission was significantly greater with Mongersen 40 mg/day compared with placebo at Weeks 2, 4, and 12 in the subgroup of patients with CDAI ≤ 260. The authors concluded that disease duration and baseline CRP did not appear to significantly affect drug efficacy. However, endoscopic indices of disease severity were not evaluated, and data from other trials suggest that baseline CDAI does not correlate with endoscopic remission.94 A phase III trial of Mongersen is ongoing, and these studies have also encouraged interest in direct anti-CCL20 therapy, which is now being explored by GSK in a Phase I clinical trial of an anti-CCL20 antibody GSK3050002 (NCT01984047).

5. Critical Appraisal and Future Directions

Although it is clear that chemokines and their receptors are critical in the development of the mucosal immune system and in orchestrating inflammatory responses to tissue injury and infection, only a few have been explored as targets for IBD.95 This may partly reflect current therapeutic focus within the field in targeting endothelial adhesion molecules, with agents such as vedolizumab. Additionally, concerns surround safety of targeting chemokine receptors in light of their widespread expression on a broad range of cell types.96 For instance, a potential ligand for GPR15 (GPR15L) was recently characterised as being developmentally determined, only modestly affected by inflammation or the presence of microbiota, and constitutively expression in the colon.97 These findings called into question the therapeutic implications of targeting GPR15, given its role in mediating colitis in T cell-dependent murine models, and high expression levels on Th2 cells in the lamina propria of patients with UC.7,8 However, GPR15 is critical for regulating the migration of FoxP3 Treg to the large intestine, and is also expressed on vascular endothelium at similar levels to T cells, where it binds to thrombomodulin, is cytoprotective, and mediates critical angiogenic functions in vivo.98 In a similar vein, maraviroc, which was developed as a therapy for HIV infection, is a safe and effective inhibitor of CCR5.99 The latter is also expressed at high frequencies by gut infiltrating lymphocytes and monocytes in IBD,100 suggesting it could be a promising target. However, its distribution on a broad range of leukocytes including activated, highly proliferative immunosuppressive Treg populations in the gut, suggest that inhibition may also disrupt critical regulatory pathways. Other potentials implicated in IBD pathogenesis include CCL17 and its only known receptor CCR4. CCL17 is increased in the inflamed mucosa of murine colitis,101,102 and CCL17-deficient animals are protected from colitis induced by DSS,103 although the ability of mice lacking CCR4+ T-cells to develop florid colitis suggests contribution by CCR4+ innate effector cell types.

There is also a strong rationale to target gut-tropic chemokines in patients with extraintestinal manifestations of IBD. Of particularly relevance is primary sclerosing cholangitis (PSC),12 a progressive biliary disease that is associated with IBD in 70–80% of cases and which affects ~8% of patients with IBD, particularly colitis.104,105 Under normal circumstances, expression of CCL25 and MAdCAM-1 are absent from the liver, but in PSC both are detectable on hepatic endothelium28,106 and support the aberrant recruitment of α4β7+ CCR9+ effector lymphocytes from the gut.10,28,106 These effector cells can then exploit CCR6 to localise to biliary epithelium, where they drive injury upon reactivation within the liver.

Despite advances in our understanding, strategies that target chemokine receptors do provide unique challenges,96 and have not yet achieved the same level of efficacy as those directed toward integrins or endothelial adhesion molecules. Although safe and reasonably well tolerated in most studies (Table 5), it has proved difficult to deliver consistent inhibition with antibodies, which may reflect the high levels of chemokines secreted by inflamed tissues or their sequestration on the endothelial glycocalyx. Indeed, targeting of chemokine-receptor interactions may be less efficacious than therapies which inhibit binding of chemokines to glycosaminoglycans in vivo.107

Table 5.

Safety and tolerability profiles of existing chemokine-directed agents.

Agent Commentary
CCX282-B (oral); Vercirnon - No specific safety concerns in the Phase II study38
- In the Phase III trial,54 gastrointestinal adverse events were observed at an increased frequency in the Vercirnon treatment groups: 30%, 37%, and 48% for placebo, Vercirnon once daily, and Vercirnon twice daily respectively (P < 0.001, 500 mg twice daily vs placebo), with the most common AEs being abdominal pain, Crohn’s disease worsening, nausea, and dyspepsia
BMS-936557 (IV); Eldelumab Ulcerative colitis phase II study69
- Infusion reactions occurred in 19%, 14%, and 5% in the 25 mg/kg, 15 mg/kg, and placebo groups, respectively. None with an acute infusion reactions exhibited anti-Eldelumab antibodies
- Higher proportion of infectious complications in the 25 mg/kg group patients (26%) vs 15 mg/kg (17.9%) and placebo (18%) groups. The most common type of infection was nasopharyngitis (10.6%, 3.6%, and 3.6%, respectively)
Crohn’s disease phase II study70
- Infusion reactions in 10% and 27% of patients receiving 10 mg/kg and 20 mg/kg arms; in three cases (all in the 20-kg arm), infusion reactions were considered serious
Mongersen (oral) - No serious concerns in the Phase II trial92
- Theoretical risk of fibrotic stricturing disease, attributable to the role of TGF-β1 in collagen synthesis, extracellular matrix dynamics, and fibrogenesis. However, no patient developed small bowel strictures in a 6-month phase I study91

AE, adverse event.

Additional limitations, relevant to cytokine-directed therapies more broadly, include acceptance that therapeutic drug monitoring in peripheral blood may not necessarily reflect levels in mucosal tissue. This was highlighted by the ATLAS study, which revealed no correlation between anti-TNFα values in matched specimens of serum versus mucosal biopsies from patients with active IBD.108 Moreover, the ratio of anti-TNF to TNF in tissue was greatest in non-inflamed areas and lowest in the most severely afflicted regions. These observations have not been conclusively validated,109 but the ATLAS investigators propose that local, progressive inflammation characterised by high tissue levels of TNFα serves as a ‘sink’ for anti-TNFα. It is plausible that the reason why some therapies fail in clinic is thus because of potency issues, wherein high levels of compound are required to ‘draw out’ chemokines from out of tissue compartments.

In future, it will be important to look at new ways of modelling pharmacokinetics for such antibodies, to ensure that trials are not failing due to suboptimal dosing.52,53 Furthermore, the large numbers of different players secreted in the context of disease suggest a degree of redundancy in inflammation-induced pathways, which could potentially reduce the efficacy of therapy targeted at single chemokines or receptors. This suggests that targeting chemokines that play a specific role in the gut or in the recruitment of particular, critical immune subsets may be more effective than targeting receptors involved more broadly during an inflammatory response, such as CXCR3 and CCR5. Furthermore, some chemokine recruitment pathways are up-regulated relatively early in the inflammatory process and, because most clinical trials include patients with active disease refractory to existing medical therapy, the crucial role of this pathway may have been missed. Consistent with this hypothesis in both the phase II and phase III anti-CCR9 trials in Crohn’s, clinical remission rates were greater in the group who did not have previous or recent exposure to biologics.38,54,69,70

It is particularly important to consider heterogeneity in the IBD phenotypes recruited for clinical trials, and to take in to account clinical, genetic, and immunophenotypic factors that might define patient subsets most likely to respond. An obvious stratification in IBD is between Crohn’s disease and ulcerative colitis, but it may be equally important to take in to account the predominant site of disease, given the regional differences in the chemokines involved in immune cell recruitment between the small bowel and colon. For instance, in the small bowel the proportion of infiltrating CCR9+ T-cells remains relatively stable or may actually lessen as inflammation progresses,39 whereas in colitis CCL25 expression is increased in the colon and strongly correlates with inflammatory activity.10 The potential importance of this factor is emphasised by SHIELD-1 where differences between Vercirnon and placebo in CD were seen in patients with colitis but not small bowel disease.54 The Mongersen Phase II trial was considerably refined in light of these concerns.92 Given that the active compound is released in the terminal ileum and ascending colon, patients with lesions localised to the upper gastrointestinal tract or distal large bowel (beyond the transverse colon) were rightly excluded.92,93

Finally, we will increasingly use sophisticated, often unsupervised, data gleaned from genomics and deep immunophenotyping, to determine much more precisely the immune pathways activated in particular patients and to use this information to target patients for specific therapy—for instance, patients with IL-17-dominated responses may respond better to therapy targeted at CCR6 or CCL20. Current approaches are likely to be an oversimplification, and detailed probing of immune pathways using novel techniques in patients may allow a much more nuanced approach to therapy selection. Herein, IBD has a real advantage over many other diseases in that analysis can be carried out on the target tissue rather than having to be inferred from changes in blood. A recent study in rheumatoid arthritis shows the potential of such approaches. The investigators used multidimensional cytometry and single-cell transcriptomics to define a new population of helper T cells in the joints of some patients with active disease.110 These cells were associated with disease activity and expressed distinct combinations of chemokines and chemokine receptors. Thus, patients in whom these cells are shown to be present and activated could be targeted with specific combinations of anti-chemokine treatment aimed at preventing the recruitment of this specific cell type early in disease.

Funding

PJT and DHA received institutional funding from the NIHR BRC. This article is independent research supported by the NIHR Birmingham Liver Biomedical Research Centre. The views expressed in this publication are of the authors and not necessarily those of the National Health Service, the NIHR, or the Department of Health.

Conflict of Interest

There are no conflicts of interest to disclose.

Acknowledgments

This paper was published as part of a supplement supported by an educational grant from F. Hoffmann-La Roche Ltd & Takeda Development Center Americas, Inc.

References

  • 1. Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol 2007;7:678–89. [DOI] [PubMed] [Google Scholar]
  • 2. Rot A, von Andrian UH. Chemokines in innate and adaptive host defense: basic chemokinese grammar for immune cells. Annu Rev Immunol 2004;22:891–928. [DOI] [PubMed] [Google Scholar]
  • 3. Baltus T, Weber KS, Johnson Z, Proudfoot AE, Weber C. Oligomerization of RANTES is required for CCR1-mediated arrest but not CCR5-mediated transmigration of leukocytes on inflamed endothelium. Blood 2003;102:1985–8. [DOI] [PubMed] [Google Scholar]
  • 4. Verkaar F, van Offenbeek J, van der Lee MMC, et al. . Chemokine cooperativity is caused by competitive glycosaminoglycan binding. J Immunol 2014;192:3908–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Svensson M, Agace WW. Role of CCL25/CCR9 in immune homeostasis and disease. Expert Rev Clin Immunol 2006;2:759–73. [DOI] [PubMed] [Google Scholar]
  • 6. Kunkel EJ, Kim CH, Lazarus NH, et al. . CCR10 expression is a common feature of circulating and mucosal epithelial tissue IgA Ab-secreting cells. J Clin Invest 2003;111:1001–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Kim SV, Xiang WV, Kwak C, et al. . GPR15-mediated homing controls immune homeostasis in the large intestine mucosa. Science 2013;340:1456–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Nguyen LP, Pan J, Dinh TT, et al. . Role and species-specific expression of colon T cell homing receptor GPR15 in colitis. Nat Immunol 2015;16:207–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Stenstad H, Svensson M, Cucak H, Kotarsky K, Agace WW. Differential homing mechanisms regulate regionalized effector CD8alphabeta+ T cell accumulation within the small intestine. Proc Natl Acad Sci U S A 2007;104:10122–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Trivedi PJ, Bruns T, Ward S, et al. . Intestinal CCL25 expression is increased in colitis and correlates with inflammatory activity. J Autoimmun 2016;68:98–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Eksteen B, Liaskou E, Adams DH. Lymphocyte homing and its role in the pathogenesis of IBD. Inflamm Bowel Dis 2008;14:1298–312. [DOI] [PubMed] [Google Scholar]
  • 12. Trivedi PJ, Adams DH. Mucosal immunity in liver autoimmunity: a comprehensive review. J Autoimmun 2013;46:97–111. [DOI] [PubMed] [Google Scholar]
  • 13. Mosli MH, Rivera-Nieves J, Feagan BG. T-cell trafficking and anti-adhesion strategies in inflammatory bowel disease: current and future prospects. Drugs 2014;74:297–311. [DOI] [PubMed] [Google Scholar]
  • 14. Habtezion A, Nguyen LP, Hadeiba H, Butcher EC. Leukocyte trafficking to the small intestine and colon. Gastroenterology 2016;150:340–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Kunkel EJ, Campbell JJ, Haraldsen G, et al. . Lymphocyte CC chemokine receptor 9 and epithelial thymus-expressed chemokine (TECK) expression distinguish the small intestinal immune compartment: Epithelial expression of tissue-specific chemokines as an organizing principle in regional immunity. J Exp Med 2000;192:761–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Papadakis KA, Prehn J, Nelson V, et al. . The role of thymus-expressed chemokine and its receptor CCR9 on lymphocytes in the regional specialization of the mucosal immune system. J Immunol 2000;165:5069–76. [DOI] [PubMed] [Google Scholar]
  • 17. Hosoe N, Miura S, Watanabe C, et al. . Demonstration of functional role of TECK/CCL25 in T lymphocyte-endothelium interaction in inflamed and uninflamed intestinal mucosa. Am J Physiol Gastrointest Liver Physiol 2004;286:G458–66. [DOI] [PubMed] [Google Scholar]
  • 18. Mora JR, Iwata M, Eksteen B, et al. . Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Science 2006;314:1157–60. [DOI] [PubMed] [Google Scholar]
  • 19. Parmo-Cabañas M, García-Bernal D, García-Verdugo R, Kremer L, Márquez G, Teixidó J. Intracellular signaling required for CCL25-stimulated T cell adhesion mediated by the integrin alpha4beta1. J Leukoc Biol 2007;82:380–91. [DOI] [PubMed] [Google Scholar]
  • 20. Miles A, Liaskou E, Eksteen B, Lalor PF, Adams DH. CCL25 and CCL28 promote alpha4 beta7-integrin-dependent adhesion of lymphocytes to MAdCAM-1 under shear flow. Am J Physiol Gastrointest Liver Physiol 2008;294:G1257–67. [DOI] [PubMed] [Google Scholar]
  • 21. Feng N, Jaimes MC, Lazarus NH, et al. . Redundant role of chemokines CCL25/TECK and CCL28/MEC in IgA+ plasmablast recruitment to the intestinal lamina propria after rotavirus infection. J Immunol 2006;176:5749–59. [DOI] [PubMed] [Google Scholar]
  • 22. Stenstad H, Ericsson A, Johansson-Lindbom B, et al. . Gut-associated lymphoid tissue-primed CD4+ T cells display CCR9-dependent and -independent homing to the small intestine. Blood 2006;107:3447–54. [DOI] [PubMed] [Google Scholar]
  • 23. Wurbel MA, Malissen M, Guy-Grand D, Malissen B, Campbell JJ. Impaired accumulation of antigen-specific CD8 lymphocytes in chemokine CCL25-deficient intestinal epithelium and lamina propria. J Immunol 2007;178:7598–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Svensson M, Marsal J, Ericsson A, et al. . CCL25 mediates the localization of recently activated CD8alphabeta(+) lymphocytes to the small-intestinal mucosa. J Clin Invest 2002;110:1113–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Wendland M, Czeloth N, Mach N, et al. . CCR9 is a homing receptor for plasmacytoid dendritic cells to the small intestine. Proc Natl Acad Sci U S A 2007;104:6347–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Hadeiba H, Lahl K, Edalati A, et al. . Plasmacytoid dendritic cells transport peripheral antigens to the thymus to promote central tolerance. Immunity 2012;36:438–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Mora JR, von Andrian UH. T-cell homing specificity and plasticity: new concepts and future challenges. Trends Immunol 2006;27:235–43. [DOI] [PubMed] [Google Scholar]
  • 28. Eksteen B, Grant AJ, Miles A, et al. . Hepatic endothelial CCL25 mediates the recruitment of CCR9+ gut-homing lymphocytes to the liver in primary sclerosing cholangitis. J Exp Med 2004;200:1511–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Rivera-Nieves J, Ho J, Bamias G, et al. . Antibody blockade of CCL25/CCR9 ameliorates early but not late chronic murine ileitis. Gastroenterology 2006;131:1518–29. [DOI] [PubMed] [Google Scholar]
  • 30. Wermers JD, McNamee EN, Wurbel MA, Jedlicka P, Rivera-Nieves J. The chemokine receptor CCR9 is required for the T-cell-mediated regulation of chronic ileitis in mice. Gastroenterology 2011;140:1526–35.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Mizuno S, Kanai T, Mikami Y, et al. . CCR9+ plasmacytoid dendritic cells in the small intestine suppress development of intestinal inflammation in mice. Immunol Lett 2012;146:64–9. [DOI] [PubMed] [Google Scholar]
  • 32. Apostolaki M, Manoloukos M, Roulis M, et al. . Role of beta7 integrin and the chemokine/chemokine receptor pair CCL25/CCR9 in modeled TNF-dependent Crohn’s disease. Gastroenterology 2008;134:2025–35. [DOI] [PubMed] [Google Scholar]
  • 33. Hadeiba H, Sato T, Habtezion A, Oderup C, Pan J, Butcher EC. CCR9 expression defines tolerogenic plasmacytoid dendritic cells able to suppress acute graft-versus-host disease. Nat Immunol 2008;9:1253–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Wurbel MA, McIntire MG, Dwyer P, Fiebiger E. CCL25/CCR9 interactions regulate large intestinal inflammation in a murine model of acute colitis. PLoS One 2011;6:e16442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Wurbel MA, Le Bras S, Ibourk M, et al. . CCL25/CCR9 interactions are not essential for colitis development but are required for innate immune cell protection from chronic experimental murine colitis. Inflamm Bowel Dis 2014;20:1165–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Papadakis KA, Prehn J, Moreno ST, et al. . CCR9-positive lymphocytes and thymus-expressed chemokine distinguish small bowel from colonic Crohn’s disease. Gastroenterology 2001;121:246–54. [DOI] [PubMed] [Google Scholar]
  • 37. Saruta M, Yu QT, Avanesyan A, Fleshner PR, Targan SR, Papadakis KA. Phenotype and effector function of CC chemokine receptor 9-expressing lymphocytes in small intestinal Crohn’s disease. J Immunol 2007;178:3293–300. [DOI] [PubMed] [Google Scholar]
  • 38. Keshav S, Vaňásek T, Niv Y, et al. ; Prospective Randomized Oral-Therapy Evaluation in Crohn’s Disease Trial-1 PROTECT-1 Study Group A randomized controlled trial of the efficacy and safety of CCX282-B, an orally-administered blocker of chemokine receptor CCR9, for patients with Crohn’s disease. PLoS One 2013;8:e60094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Papadakis KA, Prehn J, Moreno ST, et al. . CCR9-positive lymphocytes and thymus-expressed chemokine distinguish small bowel from colonic Crohn’s disease. Gastroenterology 2001;121:246–54. [DOI] [PubMed] [Google Scholar]
  • 40. Ericsson A, Kotarsky K, Svensson M, Sigvardsson M, Agace W. Functional characterization of the CCL25 promoter in small intestinal epithelial cells suggests a regulatory role for caudal-related homeobox (Cdx) transcription factors. J Immunol 2006;176:3642–51. [DOI] [PubMed] [Google Scholar]
  • 41. Meurens F, Berri M, Siggers RH, et al. . Commensal bacteria and expression of two major intestinal chemokines, TECK/CCL25 and MEC/CCL28, and their receptors. PLoS One 2007;2:e677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Bekker P, Ebsworth K, Walters MJ, et al. . CCR9 antagonists in the treatment of ulcerative colitis. Mediators Inflamm 2015;2015:628340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Zhu S, Bing Y, Wang X, et al. . CCL25/CCR9 interactions regulate the function of iNKT cells in oxazolone-induced colitis in mice. PLoS One 2014;9:e100167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Linton L, Karlsson M, Grundström J, et al. . HLA-DR(hi) and CCR9 define a pro-inflammatory monocyte subset in IBD. Clin Transl Gastroenterol 2012;3:e29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Eberhardson M, Marits P, Jones M, et al. . Treatment of inflammatory bowel disease by chemokine receptor-targeted leukapheresis. Clin Immunol 2013;149:73–82. [DOI] [PubMed] [Google Scholar]
  • 46. Mann ER, McCarthy NE, Peake ST, et al. . Skin- and gut-homing molecules on human circulating γδ T cells and their dysregulation in inflammatory bowel disease. Clin Exp Immunol 2012;170:122–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Costa MF, Bornstein VU, Candéa AL, Henriques-Pons A, Henriques MG, Penido C. CCL25 induces α₄β₇ integrin-dependent migration of IL-17⁺ γδ T lymphocytes during an allergic reaction. Eur J Immunol 2012;42:1250–60. [DOI] [PubMed] [Google Scholar]
  • 48. Singh UP, Singh NP, Murphy EA, et al. . Chemokine and cytokine levels in inflammatory bowel disease patients. Cytokine 2016;77:44–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Chen HJ, Edwards R, Tucci S, et al. . Chemokine 25-induced signaling suppresses colon cancer invasion and metastasis. J Clin Invest 2012;122:3184–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Wendt E, White GE, Ferry H, Huhn M, Greaves DR, Keshav S. Glucocorticoids suppress CCR9-mediated chemotaxis, calcium flux, and adhesion to MAdCAM-1 in human T cells. J Immunol 2016;196:3910–9. [DOI] [PubMed] [Google Scholar]
  • 51. Wendt E, Keshav S. CCR9 antagonism: potential in the treatment of inflammatory bowel disease. Clin Exp Gastroenterol 2015;8:119–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Kalindjian SB, Kadnur SV, Hewson CA, et al. . A new series of orally bioavailable chemokine receptor 9 (CCR9) antagonists; possible agents for the treatment of inflammatory bowel disease. J Med Chem 2016;59:3098–111. [DOI] [PubMed] [Google Scholar]
  • 53. Walters MJ, Wang Y, Lai N, et al. . Characterization of CCX282-B, an orally bioavailable antagonist of the CCR9 chemokine receptor, for treatment of inflammatory bowel disease. J Pharmacol Exp Ther 2010;335:61–9. [DOI] [PubMed] [Google Scholar]
  • 54. Feagan BG, Sandborn WJ, D’Haens G, et al. . Randomised clinical trial: vercirnon, an oral CCR9 antagonist, vs. placebo as induction therapy in active Crohn’s disease. Aliment Pharmacol Ther 2015;42:1170–81. [DOI] [PubMed] [Google Scholar]
  • 55. http://ir.chemocentryx.com/releasedetail.cfm?releaseid=876961. [Google Scholar]
  • 56. Eberhardson M, Karlén P, Linton L, et al. . Randomised, double-blind, placebo-controlled trial of CCR9-targeted leukapheresis treatment of ulcerative colitis patients. J Crohns Colitis 2017;11:534–42. [DOI] [PubMed] [Google Scholar]
  • 57. Shibahara T, Wilcox JN, Couse T, Madara JL. Characterization of epithelial chemoattractants for human intestinal intraepithelial lymphocytes. Gastroenterology 2001;120:60–70. [DOI] [PubMed] [Google Scholar]
  • 58. Dwinell MB, Lügering N, Eckmann L, Kagnoff MF. Regulated production of interferon-inducible T-cell chemoattractants by human intestinal epithelial cells. Gastroenterology 2001;120:49–59. [DOI] [PubMed] [Google Scholar]
  • 59. Uguccioni M, Gionchetti P, Robbiani DF, et al. . Increased expression of IP-10, IL-8, MCP-1, and MCP-3 in ulcerative colitis. Am J Pathol 1999;155:331–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Ostvik AE, Granlund AV, Bugge M, et al. . Enhanced expression of CXCL10 in inflammatory bowel disease: potential role of mucosal Toll-like receptor 3 stimulation. Inflamm Bowel Dis 2013;19:265–74. [DOI] [PubMed] [Google Scholar]
  • 61. Annunziato F, Cosmi L, Galli G, et al. . Assessment of chemokine receptor expression by human Th1 and Th2 cells in vitro and in vivo. J Leukoc Biol 1999;65:691–9. [DOI] [PubMed] [Google Scholar]
  • 62. Zhao Q, Kim T, Pang J, et al. . A novel function of CXCL10 in mediating monocyte production of proinflammatory cytokines. J Leukoc Biol 2017, Sep 12. doi: 10.1189/jlb.5A0717-302. [Epub ahead of print.] [DOI] [PubMed] [Google Scholar]
  • 63. Singh UP, Singh S, Taub DD, Lillard JW Jr. Inhibition of IFN-gamma-inducible protein-10 abrogates colitis in IL-10-/- mice. J Immunol 2003;171:1401–6. [DOI] [PubMed] [Google Scholar]
  • 64. Hyun JG, Lee G, Brown JB, et al. . Anti-interferon-inducible chemokine, CXCL10, reduces colitis by impairing T helper-1 induction and recruitment in mice. Inflamm Bowel Dis 2005;11:799–805. [DOI] [PubMed] [Google Scholar]
  • 65. Chami B, Yeung AW, van Vreden C, King NJ, Bao S. The role of CXCR3 in DSS-induced colitis. PLoS One 2014;9:e101622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Sasaki S, Yoneyama H, Suzuki K, et al. . Blockade of CXCL10 protects mice from acute colitis and enhances crypt cell survival. Eur J Immunol 2002;32:3197–205. [DOI] [PubMed] [Google Scholar]
  • 67. Kuhne M, Preston B, Wallace S, et al. . MDX-1100, a fully human anti-CXCL10 (IP-10) antibody, is a high affinity, neutralizing antibody that has entered Phase I clinical trials for the treatment of Ulcerative Colitis (UC). J Immunol 2007;178:S241. [Google Scholar]
  • 68. Mayer L, Sandborn WJ, Stepanov Y, et al. . Anti-IP-10 antibody (BMS-936557) for ulcerative colitis: a phase II randomised study. Gut 2014;63:442–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Sandborn WJ, Colombel JF, Ghosh S, et al. . Eldelumab [Anti-IP-10] induction therapy for ulcerative colitis: a randomised, placebo-controlled, phase 2b study. J Crohns Colitis 2016;10:418–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Sandborn WJ, Rutgeerts P, Colombel JF, et al. . Eldelumab [anti-interferon-γ-inducible protein-10 antibody] induction therapy for active Crohn’s disease: a randomised, double-blind, placebo-controlled phase IIa study. J Crohns Colitis 2017;11:811–9. [DOI] [PubMed] [Google Scholar]
  • 71. Swamy M, Jamora C, Havran W, Hayday A. Epithelial decision makers: in search of the ‘epimmunome’. Nat Immunol 2010;11:656–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Shao L, Serrano D, Mayer L. The role of epithelial cells in immune regulation in the gut. Semin Immunol 2001;13:163–76. [DOI] [PubMed] [Google Scholar]
  • 73. Cook DN, Prosser DM, Forster R, et al. . CCR6 mediates dendritic cell localization, lymphocyte homeostasis, and immune responses in mucosal tissue. Immunity 2000;12:495–503. [DOI] [PubMed] [Google Scholar]
  • 74. Becker C, Wirtz S, Blessing M, et al. . Constitutive p40 promoter activation and IL-23 production in the terminal ileum mediated by dendritic cells. J Clin Invest 2003;112:693–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Ivanov II, Atarashi K, Manel N, et al. . Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009;139:485–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Ohnmacht C, Park JH, Cording S, et al. . MUCOSAL IMMUNOLOGY. The microbiota regulates type 2 immunity through RORγt⁺ T cells. Science 2015;349:989–93. [DOI] [PubMed] [Google Scholar]
  • 77. Skovdahl HK, Granlund Av, Østvik AE, et al. . Expression of CCL20 and its corresponding receptor CCR6 is enhanced in active inflammatory bowel disease, and TLR3 mediates CCL20 expression in colonic epithelial cells. PLoS One 2015;10:e0141710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Kaser A, Ludwiczek O, Holzmann S, et al. . Increased expression of CCL20 in human inflammatory bowel disease. J Clin Immunol 2004;24:74–85. [DOI] [PubMed] [Google Scholar]
  • 79. Liu JZ, van Sommeren S, Huang H, et al. ; International Multiple Sclerosis Genetics Consortium; International IBD 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]
  • 80. Granlund Av, Flatberg A, Østvik AE, et al. . Whole genome gene expression meta-analysis of inflammatory bowel disease colon mucosa demonstrates lack of major differences between Crohn’s disease and ulcerative colitis. PLoS One 2013;8:e56818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Sugiura Y, Kamdar K, Khakpour S, Young G, Karpus WJ, DePaolo RW. TLR1-induced chemokine production is critical for mucosal immunity against Yersinia enterocolitica. Mucosal Immunol 2013;6:1101–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Esplugues E, Huber S, Gagliani N, et al. . Control of TH17 cells occurs in the small intestine. Nature 2011;475:514–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Katchar K, Kelly CP, Keates S, O’Brien MJ, Keates AC. MIP-3alpha neutralizing monoclonal antibody protects against TNBS-induced colonic injury and inflammation in mice. Am J Physiol Gastrointest Liver Physiol 2007;292:G1263–71. [DOI] [PubMed] [Google Scholar]
  • 84. Macdonald TT, Monteleone G. Immunity, inflammation, and allergy in the gut. Science 2005;307:1920–5. [DOI] [PubMed] [Google Scholar]
  • 85. Monteleone G, Kumberova A, Croft NM, McKenzie C, Steer HW, MacDonald TT. Blocking Smad7 restores TGF-beta1 signaling in chronic inflammatory bowel disease. J Clin Invest 2001;108:601–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Boirivant M, Pallone F, Di Giacinto C, et al. . Inhibition of Smad7 with a specific antisense oligonucleotide facilitates TGF-beta1-mediated suppression of colitis. Gastroenterology 2006;131:1786–98. [DOI] [PubMed] [Google Scholar]
  • 87. Rizzo A, Waldner MJ, Stolfi C, et al. . Smad7 expression in T cells prevents colitis-associated cancer. Cancer Res 2011;71:7423–32. [DOI] [PubMed] [Google Scholar]
  • 88. Fantini MC, Rizzo A, Fina D, et al. . Smad7 controls resistance of colitogenic T cells to regulatory T cell-mediated suppression. Gastroenterology 2009;136:1308–16, e1–3. [DOI] [PubMed] [Google Scholar]
  • 89. Marafini I, Monteleone I, Dinallo V, et al. . CCL20 is negatively regulated by TGF-β1 in intestinal epithelial cells and reduced in Crohn’s disease patients with a successful response to Mongersen, a Smad7 antisense oligonucleotide. J Crohns Colitis 2017;11:603–9. [DOI] [PubMed] [Google Scholar]
  • 90. Monteleone G, Fantini MC, Onali S, et al. . Phase I clinical trial of Smad7 knockdown using antisense oligonucleotide in patients with active Crohn’s disease. Mol Ther 2012;20:870–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Zorzi F, Calabrese E, Monteleone I, et al. . A phase 1 open-label trial shows that smad7 antisense oligonucleotide (GED0301) does not increase the risk of small bowel strictures in Crohn’s disease. Aliment Pharmacol Ther 2012;36:850–7. [DOI] [PubMed] [Google Scholar]
  • 92. Monteleone G, Neurath MF, Ardizzone S, et al. . Mongersen, an oral SMAD7 antisense oligonucleotide, and Crohn’s disease. N Engl J Med 2015;372:1104–13. [DOI] [PubMed] [Google Scholar]
  • 93. Monteleone G, Di Sabatino A, Ardizzone S, et al. . Impact of patient characteristics on the clinical efficacy of mongersen (GED-0301), an oral Smad7 antisense oligonucleotide, in active Crohn’s disease. Aliment Pharmacol Ther 2016;43:717–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Peyrin-Biroulet L, Reinisch W, Colombel JF, et al. . Clinical disease activity, C-reactive protein normalisation and mucosal healing in Crohn’s disease in the SONIC trial. Gut 2014;63:88–95. [DOI] [PubMed] [Google Scholar]
  • 95. Kulkarni N, Pathak M, Lal G. Role of chemokine receptors and intestinal epithelial cells in the mucosal inflammation and tolerance. J Leukoc Biol 2017;101:377–94. [DOI] [PubMed] [Google Scholar]
  • 96. Schall TJ, Proudfoot AE. Overcoming hurdles in developing successful drugs targeting chemokine receptors. Nat Rev Immunol 2011;11:355–63. [DOI] [PubMed] [Google Scholar]
  • 97. Ocón B, Pan J, Dinh TT, et al. . A mucosal and cutaneous chemokine ligand for the lymphocyte chemoattractant receptor GPR15. Front Immunol 2017;8:1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Pan B, Alam HB, Chong W, et al. . CitH3: a reliable blood biomarker for diagnosis and treatment of endotoxic shock. Sci Rep 2017;7:8972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Gulick RM, Lalezari J, Goodrich J, et al. ; MOTIVATE Study Teams Maraviroc for previously treated patients with R5 HIV-1 infection. N Engl J Med 2008;359:1429–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Ward ST, Li KK, Hepburn E, et al. . The effects of CCR5 inhibition on regulatory T-cell recruitment to colorectal cancer. Br J Cancer 2015;112:319–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Kristensen NN, Brudzewsky D, Gad M, Claesson MH. Chemokines involved in protection from colitis by CD4+CD25+ regulatory T cells. Inflamm Bowel Dis 2006;12:612–8. [DOI] [PubMed] [Google Scholar]
  • 102. Scheerens H, Hessel E, de Waal-Malefyt R, Leach MW, Rennick D. Characterization of chemokines and chemokine receptors in two murine models of inflammatory bowel disease: IL-10-/- mice and Rag-2-/- mice reconstituted with CD4+CD45RBhigh T cells. Eur J Immunol 2001;31:1465–74. [DOI] [PubMed] [Google Scholar]
  • 103. Heiseke AF, Faul AC, Lehr HA, et al. . CCL17 promotes intestinal inflammation in mice and counteracts regulatory T cell-mediated protection from colitis. Gastroenterology 2012;142:335–45. [DOI] [PubMed] [Google Scholar]
  • 104. Lunder AK, Hov JR, Borthne A, et al. . Prevalence of sclerosing cholangitis detected by magnetic resonance cholangiography in patients with long-term inflammatory bowel disease. Gastroenterology 2016;151:660–9.e4. [DOI] [PubMed] [Google Scholar]
  • 105. Trivedi PJ, Weismüller TJ, Bergquist A, et al. . Patient age, sex, and inflammatory bowel disease phenotype associate with course of primary sclerosing cholangitis. Gastroenterology 2017. doi: 10.1053/j.gastro.2017.02.038. [Google Scholar]
  • 106. Grant AJ, Lalor PF, Hübscher SG, Briskin M, Adams DH. MAdCAM-1 expressed in chronic inflammatory liver disease supports mucosal lymphocyte adhesion to hepatic endothelium (MAdCAM-1 in chronic inflammatory liver disease). Hepatology 2001;33:1065–72. [DOI] [PubMed] [Google Scholar]
  • 107. Proudfoot AE, Bonvin P, Power CA. Targeting chemokines: pathogens can, why can’t we?Cytokine 2015;74:259–67. [DOI] [PubMed] [Google Scholar]
  • 108. Yarur AJ, Jain A, Sussman DA, et al. . The association of tissue anti-TNF drug levels with serological and endoscopic disease activity in inflammatory bowel disease: the ATLAS study. Gut 2016;65: 249–55. [DOI] [PubMed] [Google Scholar]
  • 109. Yoshihara T, Shinzaki S, Kawai S, et al. . Tissue drug concentrations of anti-tumor necrosis factor agents are associated with the long-term outcome of patients with Crohn’s disease. Inflamm Bowel Dis 2017. doi: 10.1097/MIB.0000000000001260. [DOI] [PubMed] [Google Scholar]
  • 110. Rao DA, Gurish MF, Marshall JL, et al. . Pathologically expanded peripheral T helper cell subset drives B cells in rheumatoid arthritis. Nature 2017;542:110–4. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Crohn's & Colitis are provided here courtesy of Oxford University Press

RESOURCES