Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Dec 1.
Published in final edited form as: Ophthalmology. 2018 Jul 4;125(12):1977–1983. doi: 10.1016/j.ophtha.2018.05.014

Targeting Interleukin-23 in the Treatment of Noninfectious Uveitis

Kathryn L Pepple 1,*, Phoebe Lin 2,*
PMCID: PMC6538052  NIHMSID: NIHMS1029804  PMID: 30458922

Abstract

The interleukin (IL)-23/IL-17 axis plays a central role in the pathogenesis of immune-mediated diseases such as psoriasis, psoriatic arthritis, Crohn’s disease, and uveitis. Therefore, targeting the IL-23/IL-17 axis has become the focus of multiple clinical trials for drug development in patients with autoimmune diseases. We briefly describe the biology of the IL-23/IL-17 axis and its relevance to the pathogenesis of experimental and clinical uveitis, and review the monoclonal antibody therapies targeting this pathway. Finally, 2 ongoing phase 2 trials of the anti-IL-23 biologic therapy ustekinumab (STELARA, Janssen Biotech Inc, Horsham, PA) in patients with noninfectious uveitis are introduced.


The treatment of patients with uveitis has come a long way since glucocorticoids were first identified as an effective method for controlling ocular inflammation and preventing blindness.1 Through the use of immune-modulating medications, the detrimental side effects of systemic corticosteroids can now be largely avoided while maintaining durable disease remission.2 Unfortunately, the ideal therapy, one that perfectly controls active ocular inflammation and prevents relapses without serious systemic or ocular side effects, has remained an elusive goal. However, the fairly recent and fast-moving field of biologic therapies, in which monoclonal antibodies target specific immune mediators, has provided new hope and opportunities for realizing this goal. The successful VISUAL-I and VISUAL-II studies and subsequent Food and Drug Administration approval of the anti‒tumor necrosis factor-α agent adalimumab (Humira, AbbVie, Chicago, IL) led the way and provided uveitis specialists with a powerful new tool. In addition, these trials demonstrated that targeted biologic therapy can have a profound steroid-sparing effect in patients with uveitis.3,4

Since these studies were initiated, there has been an explosion of biologic therapies brought to market for the treatment of systemic inflammatory diseases. Only a handful of these new agents have been tested systematically in patients with uveitis.5,6 Moving forward, the growing challenge will be determining which of these therapies should be prioritized for clinical trials in uveitis. There simply are not enough resources available to support testing every option. So how do we choose? The evidence-based medicine approach calls for an ordered process including identification of central mechanisms of disease followed by development and testing of targeted therapies. In other fields where tissue collection from patients with active inflammatory disease is relatively safe and easy, and provides large samples for study, key pathogenic mechanisms have been identified by studying human samples. In ophthalmology, human samples are challenging to acquire during active uveitis flares and inherently limited in quantity. Thus, animal models of disease have had a central role in developing the foundations of our understanding of the mechanisms of ocular inflammation and generating the preclinical information supporting new treatments intended to save and preserve vision. However, this approach also has limitations because rodent eyes and immune systems are not identical to the human systems they model, and there can be layers of complexity in the roles of individual cytokines in immune health and disease that are not always appreciated until clinical trials fail to recapitulate the results seen in animal studies.7 Furthermore, classic animal models of uveitis require co-administration of a self-antigen peptide with certain adjuvants like mycobacterium, whereas the potential organisms or environmental products that might trigger uveitis in susceptible humans is unknown and limits generalizing what we learn in animal models to our human subjects. Other models in which animals are genetically modified with T-cell receptors against self-antigens to develop uveitis spontaneously perhaps are even less likely to represent a majority of cases of human uveitis. Despite these shortcomings, experimental autoimmune uveitis (EAU) and other animal models of uveitis are of great benefit, and with a larger depth of understanding of these models’ limitations and benefits, or similarities/dissimilarities to human disease, they could potentially guide successful treatment targets for human uveitis.

The key to moving forward will be the continued collaboration of basic and clinical scientists working together to untangle the complex immune mechanisms initiating and sustaining pathologic inflammation in the human eye. In this translational review, we will highlight the history and biology of the interleukin (IL)-23/IL-17 axis in uveitis, and provide an evidence-based rationale for continuing efforts to target this axis as a promising avenue for new targeted therapies in our patients with uveitis.

T-Helper 17 Cells in the Pathogenesis of Uveitis

At the center of the IL-23/IL-17 inflammatory axis lies the T-helper (Th) 17 cell.8 The Th17 cells are named for their signature cytokine, IL-17, which they produce in abundance once activated. T-helper 17 cells are an important subpopulation of CD4+ Th cells that were identified in 2005 and expanded the prior Th1 versus Th2 paradigm of Th cell subpopulations (reviewed in Tesmer et al9). T-helper 17 cells have been identified in the eyes of mice with EAU, and uveitis severity has been linked to the total burden of intraocular Th17 cells.1012 In humans, peripheral blood Th17 cells have been identified in patients with scleritis and active uveitis,10 and an elevated ratio of Th17 to regulatory T cells has been correlated to disease activity in patients with human leukocyte antigen B27eassociated uveitis.13

T-helper 17 cells are a heterogeneous group of cells that function both in autoimmunity and in mucosal immunity to protect from pathogens such as fungi, bacteria, and viruses. The pathologic and homeostatic functions of Th17 cells appear to segregate within at least 2 distinct subpopulations. One type of Th17 cell is highly pathogenic and can be identified by the expression of IL-17, interferon γ, granulocyte- macrophage colony-stimulating factor, and IL-6. The second population has beneficial homeostatic functions and can be identified by the expression of IL-17 and IL-10.1416 Although it is informative to consider these as 2 distinct subpopulations, the relationships are complex and fluid, because transdifferentiation of pathogenic Th17 cells into IL-10 expressing regulatory cells during resolution of inflammation has been demonstrated in multiple experimental models.17 In the absence of disease, the majority of vertical and horizontal cells reside in the intestines where their development is induced by the presence of certain types of commensal bacteria and where they act on intestinal epithelial cells to maintain a healthy mucosal barrier.18,19 The importance of these beneficial Th17 cells is highlighted by the development of infections, such as chronic mucocutaneous candidiasis, in patients with Th17 deficiency.20

In the case of autoimmunity, pathogenic Th17 cells deviate from their protective functions and drive destructive tissue inflammation. One of the main pathologic roles of Th17 cells is to produce and release IL-17. In the context of inflammation, IL-17 is a potent chemotactic agent for cells that mediate tissue injury, such as neutrophils, monocytes, and macrophages. Interleukin-17 also acts as an autocrine and paracrine signal to continue local production of inflammatory cytokines, chemokines, and matrix metalloproteases from T cells, epithelial cells, and stromal cells, which further causes additional inflammation and tissue injury.8

Considerable excitement was generated around targeting IL-17 as a possible therapy for uveitis after animal studies using the EAU model demonstrated that treatment with anti‒IL-17 antibodies ameliorated disease, and that there was an increase in serum levels of IL-17 in patients with uveitis associated with Behçet’s disease.2123 However, 3 clinical trials of the anti‒IL-17 antibody secukinumab, SHIELD, ENSURE, and ENDURE, in the treatment of patients with noninfectious uveitis failed to reach their primary end points.24 Of note, these were not the only trials of anti‒IL-17 therapy with unexpected clinical trial results. In patients with psoriasis treated with secukinumab, paradoxical exacerbations of inflammatory bowel disease were also reported.25

Subsequent studies in experimental models of uveitis have determined that the roles of IL-17 and Th17 cells in homeostasis and autoimmunity are more complex than initially appreciated. Early studies suggested that IL-17 had a purely pathogenic and prouveitic function, but more recent studies have identified a beneficial effect on EAU from treatment with recombinant IL-17.10,15,26 In EAU, the frequency of homeostatic Th17 cells outnumbers the retina-specific Th17 cells by 10:1, produce the anti-inflammatory cytokine IL-10, and neutralize the uveitogenic effect of pathogenic Th17 cells upon adoptive transfer.15,16 Thus, Th17 cells and IL-17 can contribute to both disease pathogenesis and recovery of homeostasis in experimental uveitis. This effect of IL-17 has also been described in other auto-immune disease models.8,2729 Therefore, the role of IL-17 is also likely to be complex in human uveitis, and these animal studies would imply that unless the inflammatory and anti-inflammatory roles of Th17 cells and IL-17 can be uncoupled, anti‒IL-17 therapy may not provide the desired therapeutic benefit. With the discovery of IL-23 and the subsequent development of anti‒IL-23 therapy, this may become a possibility.

Interleukin-23 in Autoimmunity

The cytokine environment in which a naïve T-cell experiences its cognate antigen helps determine what type of T-cell will ultimately develop. Multiple lines of evidence point to IL-23 as the key checkpoint used by naïve T cells to make the decision whether to become a homeostatic Th17 or pathogenic Th17 effector cell.23,30,31 To begin Th17 differentiation, naïve T-cells require exposure to transforming growth factor-b and IL-6, and in the absence of these cytokines Th17 cells fail to develop. Interleukin-23 is not necessary for the initial stages of Th17 differentiation, but when it is present it preferentially drives the developing Th17 cells into the highly inflammatory pathogenic subtype. Interleukin-23 also maintains this fate by upregulating the expression of its own receptor (interleukin-23 receptor [IL-23R]), which stabilizes and allows expansion of this harmful Th17 population.32,33

Interleukin-23 belongs to the IL-12 family of cytokines (IL-12, IL-23, IL-35, and fusokine) and is a heterodimeric protein made of p40 and p19 subunits (Fig 1).34 The p40 subunit is shared in common with IL-12 (p40/p35), whereas the p19 subunit is unique to IL-23 (p40/p19). Biological activity of IL-23 is mediated by binding to the IL-23R, which is expressed on activated T cells, including Th17 cells, γδ T cells, natural killer cells, and innate lymphoid cells.35

Figure 1.

Figure 1

Targeting interleukin (IL)-23. Interleukin-23 is a crucial checkpoint for the development of pathogenic T-helper (Th) 17 cells implicated in various types of immune-mediated conditions, including uveitis. Preventing IL-23 binding to its receptor by targeting the p40 subunit of IL-23, for instance, can disrupt formation of pathogenic Th17 cells without affecting potentially beneficial homeostatic Th17 cells. TGF = transforming growth factor.

Interleukin-23/IL-23R signaling activates the intracellular JAK/STAT signaling pathway (using JAK2, Tyk2, STAT3, and STAT4) in Th17 cells, resulting in the production of type 17 cytokines (IL-17, IL-22, and granulocyte-macrophage colony stimulating factor) that can drive chronic tissue inflammation. Interleukin-23 is expressed by dendritic cells, macrophages, B cells, and endothelial cells when they detect pathogen-associated molecular patterns generated by infectious organisms or host-associated innate signals known as “danger-associated molecular proteins” generated by inflammation-associated tissue injury.36

Evidence for Interleukin-23 as a Mediator of Uveitis

In human studies, increased IL-23 has been found in the serum and the supernatants of peripheral blood mononuclear cells from patients with active Vogt‒Koyonagi‒Harada and Behçet’s uveitis compared with patients with inactive uveitis and normal control subjects.21,37,38 Przepiera-Bedzak et al39 have shown that increased serum levels of IL-23 are associated with increased risk of acute anterior uveitis in patients with spondyloarthropathy (in addition to elevated IL-6, IL-18, and decreased endothelin-1 levels).39 Using proteomics of several hundred inflammatory markers, Velez et al40 found increased IL-23 production in the vitreous obtained from 15 patients with posterior uveitis compared with nonuveitic control subjects.40 Yang and Foster41 discovered elevated serum levels of IL-23 in patients with treatment-naïve birdshot chorioretinopathy, greater than levels found in normal subjects and patients with birdshot chorioretinopathy who had a history of immunosuppressive treatment.

Additional support for the importance of IL-23 in uveitis comes from multiple genetic association studies. Several single nucleotide polymorphisms (SNPs) in the IL-23R gene have been found to be associated with chronic inflammatory diseases, including inflammatory bowel disease,42 ankylosing spondylitis, uveitis,43 and psoriasis.44 Dong et al45 reported that although 3 IL-23R gene SNPs were no different between patients with ankylosing spondylitis and a control group, the homozygous GG genotype of SNP rs17375018 in the IL-23R gene was significantly associated with patients with ankylosing spondylitis and uveitis. This same SNP is increased in Behçet’s disease uveitis as well. Hou et al46 subsequently discovered that IL-23A and IL-17F gene copy number variants >2 were associated with Behçet’s disease uveitis and Vogt‒Koyonagi‒Harad uveitis. Kim et al44 demonstrated that the IL-23R gene SNPs rs11465804 and rs11209026 were associated with sarcoid uveitis compared with control subjects. These clinical data are also supported experimentally because Il-23‒deficient animals are resistant to EAU.23,36

Clinical Trials and Agents Targeting the Interleukin-23/17 Axis

Biologic agents targeting the IL-23/IL-17 axis have been studied in inflammatory bowel disease, psoriasis, ankylosing spondylitis, and uveitis. Because of the proximal relationship of IL-23 and IL-17, initially these 2 cytokines were believed to be interchangeable as targets for autoimmune disease. However, initial clinical results found some surprising differences in therapeutic efficacy of anti‒IL-17 and anti‒IL-23 agents. Continuing studies into the complexity of type-17 cell biology and the dichotomous effects that IL-17 and IL-23 have on immunity have provided some answers to help explain these clinical trial results, but ultimately determining the ability of these agents to affect noninfectious uveitis will require well-designed clinical trials.

The biologic agents specifically targeting the IL-17/IL-23 axis that are currently available or are under development are summarized in Table 1.4764 In patients with uveitis, secukinumab (an anti‒IL-17A antibody) is the only agent with completed trial results. Although secukinumab was not successful in several phase 3 clinical trials in patients with noninfectious uveitis, caveats in study design, including mode of administration of the drug, may have affected the study results.65 It remains to be determined whether or not targeting IL-17 or the IL-17 receptor using a different approach or drug will prove to be more efficacious.

Table 1.

Anti-Interleukin-17 and Anti-Interleukin-23 Monoclonal Antibody Therapies

Agent Drug Name Target Nonocular Indications with FDA Approval or in Active Clinical Trials Trials in Patients with Noninfectious Uveitis
Ixekizumab Taltz (Eli Lilly and Co., Indianapolis, IN) IL-17A Moderate to severe plaque psoriasis47 Active psoriatic arthritis48,49 None
Brodalumab Siliq (Valeant, Quebec, Canada) IL-17RA Moderate to severe plaque psoriasis5052 None
Secukinumab Cosentyx (Novartis, East Hanover, NH) IL-17A Plaque psoriasis,53,54 psoriatic arthritis,55,56 ankylosing spondylitis57 • One phase 2 trial of intravenous administration65
• Three phase 3 trials subcutaneous administration24 (SHIELD, INSURE, ENDURE)
Ustekinumab STELARA (Janssen Biotech Inc, Horsham, PA) p40 subunit of and IL-12 IL-23 Moderately to severely active Crohn’s disease,58,59 moderate to severe plaque psoriasis,6062 active psoriatic arthritis63,64 NCT02648581: Phase 2 trials on the efficacy and safety of ustekinumab, in patients with Behçet disease (STELABEC-1) and active posterior and panuveitis
NCT02911116: Phase 2 STELARA for the Treatment of Active Sight-Threatening Uveitis trial
Guselkumab Tremfya (Janssen Biotech, Inc) IL-23 p19 Moderate to severe plaque psoriasis6870 none
Tildrakizumab IL-23 p19 Moderate to severe plaque psoriasis71 none
Brazikumab (MEDI2070) IL-23 p19 In development for Crohn’s disease (NCT01714726)72 none
Risankizumab IL-23 p19 In development for psoriasis,73 Crohn’s disease,74 psoriatic arthritis (NCT02986373) none

FDA = Food and Drug Administration; IL = interleukin.

Ustekinumab, which targets the p40 subunit that is shared by both IL-23 and IL-12, has been reported to effectively control uveitis in 2 patients treated for coexisting severe psoriatic arthritis and plaque psoriasis or Behçet’s disease.66,67 In 2016, 2 phase 2 studies studying the efficacy of subcutaneous ustekinumab (STELARA, Janssen Biotech Inc, Horsham, PA) in patients with uveitis began enrollment. In the STELABEC trial, ustekinumab will be tested in 30 patients with Behçet’s disease with active posterior and panuveitis (clinicaltrials.gov NCT02648581). Treatment will be administered at weeks 0, 4, and 16, with end point assessment at week 24. In the STELARA for the Treatment of Active Sight-Threatening Uveitis trial, 7 patients with active, sight-threatening, noninfectious, intermediate, posterior, or panuveitis uveitis will be treated at weeks 0, 4, and 8, with end point assessment at 16 weeks (clinicaltrials.gov NCT02911116). The results of these studies are anticipated for release in 2019 or 2020.

In addition to ustekinumab, newer IL-23 specific agents have been developed that target the p19 subunit. Phase 3 trials for the human monoclonal antibodies tildrakizumab and guselkumab in patients with psoriasis were both successful, and these agents are now Food and Drug Administration approved for the treatment of moderate to severe disease.6871 Other p19-specific agents that are in development include brazikumab for Crohn’s disease (clinicaltrials.gov NCT01714726)72 and risankizumab for psoriasis,73 Crohn’s disease,74 and psoriatic arthritis (clinicaltrials.gov NCT02986373). In anti-IL-23 studies, a common adverse event was nasopharyngitis, but serious adverse events were not significantly different when compared with placebo. However, as with many other immune-modifying medications, there may be a need for dermatologic monitoring because nonmelanoma skin cancers were found in 14 patients in the PHOENIX trial for ustekinumab.75 Overall, the rate of antibodies against the anti‒IL-23 biologics was variable and relatively rare.

Despite the growing body of literature supporting IL-23 as a good therapeutic target for the treatment of immune- mediated uveitis, how to target IL-23 or its receptor for best treatment efficacy in uveitis remains unknown. Keino et al76 discovered that an oral IL-12/IL-23 small molecule inhibitor, STA-5326, that targets expression of the p40 subunit through c-Rel (a member of the nuclear factor-kb family of transcription factors) was found to reduce the severity of uveitis in EAU mice at 2 different doses, 5 mg/kg and 20 mg/kg.76 The STELARA for the Treatment of Active Sight-Threatening Uveitis and STELABAC trials both deliver anti-IL-23 therapy via the subcutaneous route, which was effective for treatment of patients with Crohn’s disease, psoriasis, and psoriatic arthritis but was not effective in a phase 2 clinical trial in patients with relapsing-remitting multiple sclerosis.77 The route of drug delivery was only 1 possible reason cited for failure, along with inclusion of patients with late-stage disease who were unlikely to benefit from treatment and inability of ustekinumab to cross the bloodebrain barrier.78 Because the pending uveitis trials of ustekinumab are relatively small, they may be susceptible to some of the pitfalls encountered in the multiple sclerosis trial. However, the prior anecdotal support for efficacy of this route still provides optimism that these trials will provide the first clear evidence for use of anti-IL-23 therapy as the next option for biologic therapy in patients with uveitis.

In conclusion, both IL-17 and IL-23 appear to play a crucial role in noninfectious uveitis. However, although targeting IL-23 and IL-17 still holds significant promise, the complex effects that result from modification of this immune axis will likely require refinement in patient selection, timing of therapy, and route of drug administration to determine the true efficacy.

Acknowledgments

Financial Disclosure(s):

The author(s) made the following disclosures: K.P.: Support Research to Prevent Blindness Career Development Award (New York, NY), The Alcon Research Institute Young Investigators Grant (Fort Worth, TX), and the National Institute of Health, NEI K08EY023998.

P.L.: Supported National Eye Institute Grant K08 EY022948, a Collins Medical Trust Grant, Research to Prevent Blindness Career Development Award. The manuscript is supported by Core Grants P30 EY010572 and P30 EY01730 from the National Institute of Health (Bethesda, MD) and by unrestricted departmental funding from Research to Prevent Blindness (New York, NY).

Abbreviations and Acronyms:

EAU

experimental autoimmune uveitis

IL

interleukin

IL-23R

interleukin 23 receptor

SNP

single nucleotide polymorphism

Th

T-helper

References

  • 1.Leopold IH, Purnell JE, Cannon EJ, et al. Local and systemic cortisone in ocular disease. Am J Ophthalmol 1951;34:361–371. [DOI] [PubMed] [Google Scholar]
  • 2.Jabs DA, Rosenbaum JT, Foster CS, et al. Guidelines for the use of immunosuppressive drugs in patients with ocular inflammatory disorders: recommendations of an expert panel. Am J Ophthalmol 2000;130:492–513. [DOI] [PubMed] [Google Scholar]
  • 3.Jaffe GJ, Dick AD, Brezin AP, et al. Adalimumab in patients with active noninfectious uveitis. N Engl J Med 2016;375: 932–943. [DOI] [PubMed] [Google Scholar]
  • 4.Nguyen QD, Merrill PT, Jaffe GJ, et al. Adalimumab for prevention of uveitic flare in patients with inactive non- infectious uveitis controlled by corticosteroids (VISUAL II): a multicentre, double-masked, randomised, placebo-controlled phase 3 trial. Lancet 2016;388:1183–1192. [DOI] [PubMed] [Google Scholar]
  • 5.Mesquida M, Molins B, Llorenc V, et al. Long-term effects of tocilizumab therapy for refractory uveitis-related macular edema. Ophthalmology 2014;121:2380–2386. [DOI] [PubMed] [Google Scholar]
  • 6.Suhler EB, Lim LL, Beardsley RM, et al. Rituximab therapy for refractory scleritis: results of a phase I/II dose-ranging, randomized, clinical trial. Ophthalmology 2014;121: 1885–1891. [DOI] [PubMed] [Google Scholar]
  • 7.Seok J, Warren HS, Cuenca AG, et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci U S A 2013;110:3507–3512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Gaffen SL, Jain R, Garg AV, Cua DJ. The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat Rev Immunol 2014;14:585–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Tesmer LA, Lundy SK, Sarkar S, Fox DA. Th17 cells in human disease. Immunol Rev 2008;223:87–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Amadi-Obi A, Yu CR, Liu X, et al. TH17 cells contribute to uveitis and scleritis and are expanded by IL-2 and inhibited by IL-27/STAT1. Nat Med 2007;13:711–718. [DOI] [PubMed] [Google Scholar]
  • 11.Caspi RR. A look at autoimmunity and inflammation in the eye. J Clin Invest 2010;120:3073–3083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Yazid S, Gardner PJ, Carvalho L, et al. Annexin-A1 restricts Th17 cells and attenuates the severity of autoimmune disease. J Autoimmun 2015;58:1–11. [DOI] [PubMed] [Google Scholar]
  • 13.Zhuang Z, Wang Y, Zhu G, et al. Imbalance of Th17/Treg cells in pathogenesis of patients with human leukocyte antigen B27 associated acute anterior uveitis. Sci Rep 2017;7: 40414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 cells. Annu Rev Immunol 2009;27:485–517. [DOI] [PubMed] [Google Scholar]
  • 15.Peng Y, Han G, Shao H, et al. Characterization of IL-17þ interphotoreceptor retinoid-binding protein-specific T cells in experimental autoimmune uveitis. Invest Ophthalmol Vis Sci 2007;48:4153–4161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Nian H, Liang D, Zuo A, et al. Characterization of autoreactive and bystander IL-17þ T cells induced in immunized C57BL/6mice. Invest Ophthalmol Vis Sci 2012;53:897–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gagliani N, Amezcua Vesely MC, Iseppon A, et al. Th17 cells transdifferentiate into regulatory T cells during resolution of inflammation. Nature 2015;523:221–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Littman DR, Rudensky AY. Th17 and regulatory T cells in mediating and restraining inflammation. Cell 2010;140:845–858. [DOI] [PubMed] [Google Scholar]
  • 19.Lee JS, Tato CM, Joyce-Shaikh B, et al. Interleukin-23-independent IL-17 production regulates intestinal epithelial permeability. Immunity 2015;43:727–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.McDonald DR. TH17 deficiency in human disease. J Allergy Clin Immunol 2012;129:1429–1435. quiz 36–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chi W, Zhu X, Yang P, et al. Upregulated IL-23 and IL-17 in Behcet patients with active uveitis. Invest Ophthalmol Vis Sci 2008;49:3058–3064. [DOI] [PubMed] [Google Scholar]
  • 22.Jawad S, Liu B, Agron E, et al. Elevated serum levels of interleukin-17A in uveitis patients. Ocul Immunol Inflamm 2013;21:434–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Luger D, Silver PB, Tang J, et al. Either a Th17 or a Th1 effector response can drive autoimmunity: conditions of disease induction affect dominant effector category. J Exp Med 2008;205:799–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Dick AD, Tugal-Tutkun I, Foster S, et al. Secukinumab in the treatment of noninfectious uveitis: results of three randomized, controlled clinical trials. Ophthalmology 2013;120: 777–787. [DOI] [PubMed] [Google Scholar]
  • 25.Hueber W, Sands BE, Lewitzky S, et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut 2012;61:1693–1700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ke Y, Liu K, Huang GQ, et al. Anti-inflammatory role of IL- 17 in experimental autoimmune uveitis. J Immunol 2009;182: 3183–3190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Haak S, Croxford AL, Kreymborg K, et al. IL-17A and IL-17F do not contribute vitally to autoimmune neuro-inflammation in mice. J Clin Invest 2009;119:61–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Komiyama Y, Nakae S, Matsuki T, et al. IL-17 plays an important role in the development of experimental autoimmune encephalomyelitis. J Immunol 2006;177:566–573. [DOI] [PubMed] [Google Scholar]
  • 29.Yang XO, Chang SH, Park H, et al. Regulation of inflammatory responses by IL-17F. J Exp Med 2008;205:1063–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Cua DJ, Sherlock J, Chen Y, et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature 2003;421:744–748. [DOI] [PubMed] [Google Scholar]
  • 31.Murphy CA, Langrish CL, Chen Y, et al. Divergent pro- and antiinflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation. J Exp Med 2003;198:1951–1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.McGeachy MJ, Bak-Jensen KS, Chen Y, et al. TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nat Immunol 2007;8:1390–1397. [DOI] [PubMed] [Google Scholar]
  • 33.Aggarwal S, Ghilardi N, Xie MH, et al. Interleukin-23 promotes a distinct CD4 T cell activation state characterized by the production of interleukin-17. J Biol Chem 2003;278:1910–1914. [DOI] [PubMed] [Google Scholar]
  • 34.Kastelein RA, Hunter CA, Cua DJ. Discovery and biology of IL-23 and IL-27: related but functionally distinct regulators of inflammation. Annu Rev Immunol 2007;25:221–242. [DOI] [PubMed] [Google Scholar]
  • 35.Parham C, Chirica M, Timans J, et al. A receptor for the heterodimeric cytokine IL-23 is composed of IL-12Rbeta1 and a novel cytokine receptor subunit, IL-23 R. J Immunol 2002;168:5699–5708. [DOI] [PubMed] [Google Scholar]
  • 36.Sun D, Liang D, Kaplan HJ, Shao H. The role of Th17-associated cytokines in the pathogenesis of experimental autoimmune uveitis (EAU). Cytokine 2015;74:76–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Chi W, Yang P, Li B, et al. IL-23 promotes CD4þ T cells to produce IL-17 in Vogt-Koyanagi-Harada disease. J Allergy Clin Immunol 2007;119:1218–1224. [DOI] [PubMed] [Google Scholar]
  • 38.Jiang S, Liu X, Luo L, et al. Elevated serum IL-23 correlates with intraocular inflammation after cataract surgery in patients with Vogt-Koyanagi-Harada disease. Br J Ophthalmol 2010;94:1078–1082. [DOI] [PubMed] [Google Scholar]
  • 39.Przepiera-Bedzak H, Fischer K, Brzosko M. Extra-articular symptoms in constellation with selected serum cytokines and disease activity in spondyloarthritis. Mediators Inflamm 2016;2016:7617954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Velez G, Roybal CN, Colgan D, et al. Precision medicine: personalized proteomics for the diagnosis and treatment of idiopathic inflammatory disease. JAMA Ophthalmol 2016;134:444–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Yang P, Foster CS. Interleukin 21, interleukin 23, and transforming growth factor beta1 in HLA-A29-associated birdshot retinochoroidopathy. Am J Ophthalmol 2013;156: 400–406.e2. [DOI] [PubMed] [Google Scholar]
  • 42.Duerr RH, Taylor KD, Brant SR, et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 2006;314:1461–1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Jung JH, Song GG, Kim JH, et al. The association between genetic polymorphisms of the interleukin-23 receptor gene and susceptibility to uveitis: a meta-analysis. BMC Ophthalmol 2017;17:81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kim HS, Choi D, Lim LL, et al. Association of interleukin 23 receptor gene with sarcoidosis. Dis Markers 2011;31:17–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Dong H, Li Q, Zhang Y, et al. IL23R gene confers susceptibility to ankylosing spondylitis concomitant with uveitis in a Han Chinese population. PLoS One 2013;8:e67505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hou S, Liao D, Zhang J, et al. Genetic variations of IL17F and IL23A show associations with Behcet’s disease and Vogt-Koyanagi-Harada syndrome. Ophthalmology 2015;122: 518–523. [DOI] [PubMed] [Google Scholar]
  • 47.Farahnik B, Beroukhim K, Zhu TH, et al. Ixekizumab for the treatment of psoriasis: A Review of Phase III Trials. Dermatol Ther (Heidelb) 2016;6(1):25–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Van der Heijde D, Gladman DD, Kishimoto M, et al. Efficacy and safety of ixekizumab in patients with active psoriatic arthritis: 52-week results from a phase III study (SPIRIT-P1). J Rheumatol 2018;45:367–377. [DOI] [PubMed] [Google Scholar]
  • 49.Nash P, Kirkham B, Okada M, et al. Ixekizumab for the treatment of patients with active psoriatic arthritis and an inadequate response to tumour necrosis factor inhibitors: results from the 24-week randomised, double-blind, placebo-controlled period of the SPIRIT-P2 phase 3 trial. Lancet 2017;389(10086):2317–2327. [DOI] [PubMed] [Google Scholar]
  • 50.Blauvelt A, Papp KA, Lebwohl MG, et al. Rapid onset of action in patients with moderate-to-severe psoriasis treated with brodalumab: A pooled analysis of data from two phase 3 randomized clinical trials (AMAGINE-2 and AMAGINE-3). J Am Acad Dermatol 2017;77(2):372–374. [DOI] [PubMed] [Google Scholar]
  • 51.Papp KA, Reich K, Paul C, et al. A prospective phase III, randomized, double-blind, placebo-controlled study of brodalumab in patients with moderate-to-severe plaque psoriasis. Br J Dermatol 2016;175(2):273–286. [DOI] [PubMed] [Google Scholar]
  • 52.Lebwohl M, Strober B, Menter A, et al. Phase 3 studies comparing brodalumab with ustekinumab in psoriasis. N Engl J Med 2015;373(14):1318–1328. [DOI] [PubMed] [Google Scholar]
  • 53.Langley RG, Elewski BE, Lebwohl M, et al. Secukinumab in plaque psoriasiseresults of two phase 3 trials. N Engl J Med 2014;371(4):326–338. [DOI] [PubMed] [Google Scholar]
  • 54.Blauvelt A, Prinz JC, Gottlieb AB, et al. Secukinumab administration by pre-filled syringe: efficacy, safety and us- ability results from a randomized controlled trial in psoriasis (FEATURE). Br J Dermatol 2015;172(2):484–493. [DOI] [PubMed] [Google Scholar]
  • 55.McInnes IB, Mease PJ, Kirkham B, et al. Secukinumab, a human anti-interleukin-17A monoclonal antibody, in patients with psoriatic arthritis (FUTURE 2): a randomised, double- blind, placebo-controlled, phase 3 trial. Lancet 2015;386(9999):1137–1146. [DOI] [PubMed] [Google Scholar]
  • 56.Mease PJ, McInnes IB, Kirkham B, et al. Secukinumab inhibition of interleukin-17A in patients with psoriatic arthritis. N Engl J Med 2015;373(14):1329–1339. [DOI] [PubMed] [Google Scholar]
  • 57.Baeten D, Sieper J, Braun J, et al. Secukinumab, an inter- leukin-17A inhibitor, in ankylosing spondylitis. N Engl J Med 2015;373(26):2534–2548. [DOI] [PubMed] [Google Scholar]
  • 58.Sandborn WJ, Gasink C, Gao LL, et al. Ustekinumab induction and maintenance therapy in refractory Crohn’s disease. N Engl J Med 2012;367(16):1519–1528. [DOI] [PubMed] [Google Scholar]
  • 59.Feagan BG, Sandborn WJ, Gasink C, et al. Ustekinumab as Induction and Maintenance Therapy for Crohn’s Disease. N Engl J Med 2016;375(20):1946–1960. [DOI] [PubMed] [Google Scholar]
  • 60.Leonardi CL, Kimball AB, Papp KA, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal anti-body, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet 2008;371(9625):1665–1674. [DOI] [PubMed] [Google Scholar]
  • 61.Papp KA, Langley RG, Lebwohl M, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal anti- body, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet 2008;371(9625):1675–1684. [DOI] [PubMed] [Google Scholar]
  • 62.Griffiths CE, Strober BE, van de Kerkhof P, et al. Comparison of ustekinumab and etanercept for moderate-to-severe psoriasis. N Engl J Med 2010;362(2):118–128. [DOI] [PubMed] [Google Scholar]
  • 63.McInnes IB, Kavanaugh A, Gottlieb AB, et al. PSUMMIT 1 Study Group. Efficacy and safety of ustekinumab in patients with active psoriatic arthritis: 1 year results of the phase 3, multicentre, double-blind, placebo-controlled PSUMMIT 1 trial. Lancet 2013;382(9894):780–789. [DOI] [PubMed] [Google Scholar]
  • 64.Ritchlin C, Rahman P, Kavanaugh A, et al. Efficacy and safety of the anti-IL-12/23 p40 monoclonal antibody, ustekinumab, in patients with active psoriatic arthritis despite conventional non-biological and biological anti-tumour necrosis factor therapy: 6-month and 1-year results of the phase 3, multi-centre, double-blind, placebo-controlled, randomised PSUM- MIT 2 trial. Ann Rheum Dis 2014;73(6):990–999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Letko E, Yeh S, Foster CS, et al. Efficacy and safety of intravenous secukinumab in noninfectious uveitis requiring steroid-sparing immunosuppressive therapy. Ophthalmology 2015;122:939–948. [DOI] [PubMed] [Google Scholar]
  • 66.Mugheddu C, Atzori L, Del Piano M, et al. Successful ustekinumab treatment of noninfectious uveitis and concomitant severe psoriatic arthritis and plaque psoriasis. Dermatol Ther 2017;30. [DOI] [PubMed] [Google Scholar]
  • 67.Baerveldt EM, Kappen JH, Thio HB, et al. Successful long- term triple disease control by ustekinumab in a patient with Behcet’s disease, psoriasis and hidradenitis suppurativa. Ann Rheum Dis 2013;72:626–627. [DOI] [PubMed] [Google Scholar]
  • 68.Langley RG, Tsai TF, Flavin S, et al. Efficacy and safety of guselkumab in patients with psoriasis who have an inadequate response to ustekinumab: results of the randomized, double-blind, phase III NAVIGATE trial. Br J Dermatol 2018;178:114–123. [DOI] [PubMed] [Google Scholar]
  • 69.Blauvelt A, Papp KA, Griffiths CE, et al. Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the continuous treatment of patients with moderate to severe psoriasis: results from the phase III, double-blinded, placebo- and active comparator-controlled VOYAGE 1 trial. J Am Acad Dermatol 2017;76:405–417. [DOI] [PubMed] [Google Scholar]
  • 70.Reich K, Armstrong AW, Foley P, et al. Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the treatment of patients with moderate to severe psoriasis with randomized withdrawal and retreatment: results from the phase III, double-blind, placebo- and active comparator-controlled VOYAGE 2 trial. J Am Acad Dermatol 2017;76:418–431. [DOI] [PubMed] [Google Scholar]
  • 71.Reich K, Papp KA, Blauvelt A, et al. Tildrakizumab versus placebo or etanercept for chronic plaque psoriasis (reSURFACE 1 and reSURFACE 2): results from two randomised controlled, phase 3 trials. Lancet 2017;390:276–288. [DOI] [PubMed] [Google Scholar]
  • 72.Sands BE, Chen J, Feagan BG, et al. Efficacy and safety of MEDI2070, an antibody against interleukin 23, in patients with moderate to severe Crohn’s disease: a phase 2a study. Gastroenterology 2017;153:77–86.e6. [DOI] [PubMed] [Google Scholar]
  • 73.Papp KA, Blauvelt A, Bukhalo M, et al. Risankizumab versus ustekinumab for moderate-to-severe plaque psoriasis. N Engl J Med 2017;376:1551–1560. [DOI] [PubMed] [Google Scholar]
  • 74.Feagan BG, Sandborn WJ, D’Haens G, et al. Induction therapy with the selective interleukin-23 inhibitor risankizumab in patients with moderate-to-severe Crohn’s disease: a randomised, double-blind, placebo-controlled phase 2 study. Lancet 2017;389:1699–1709. [DOI] [PubMed] [Google Scholar]
  • 75.Chen Z, Gong Y, Shi Y. Novel biologic agents targeting interleukin-23 and interleukin-17 for moderate-to-severe psoriasis. Clin Drug Investig 2017;37:891–899. [DOI] [PubMed] [Google Scholar]
  • 76.Keino H, Watanabe T, Sato Y, et al. Therapeutic effect of the potent IL-12/IL-23 inhibitor STA-5326 on experimental autoimmune uveoretinitis. Arthritis Res Ther 2008;10:R122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Segal BM, Constantinescu CS, Raychaudhuri A, et al. Repeated subcutaneous injections of IL12/23 p40 neutralising antibody, ustekinumab, in patients with relapsing-remitting multiple sclerosis: a phase II, double-blind, placebo- controlled, randomised, dose-ranging study. Lancet Neurol 2008;7:796–804. [DOI] [PubMed] [Google Scholar]
  • 78.Longbrake EE, Racke MK. Why did IL-12/IL-23 antibody therapy fail in multiple sclerosis? Expert Rev Neurother 2009;9:319–321. [DOI] [PubMed] [Google Scholar]

RESOURCES