1,650
Views
1
CrossRef citations to date
0
Altmetric
Editorial

Rationale for IL-37 as a novel therapeutic agent in inflammation

ORCID Icon & ORCID Icon
Pages 1203-1206 | Received 14 Mar 2022, Accepted 29 Jul 2022, Published online: 16 Aug 2022

1. Introduction

An estimated 5–8% of the world’s population is afflicted by severe inflammatory diseases that are among the leading causes of death, particularly in women younger than 65 years in case of autoimmune etiologies. Therefore, the unmet medical need is urgent – and interleukin (IL-) 37 holds substantial promise as a new therapeutic in this field.

IL-37 is an IL-1 family cytokine with powerful anti-inflammatory functions that govern principal pathways of innate and adaptive immunity and inflammation [Citation1,Citation2], including signaling by pattern recognition receptors such as Toll-like receptors (TLRs), cytokines such as IL-1 and tumor necrosis factor (TNF), and type 1-, 2-, and 3-polarized adaptive immune responses. Such broad actions are rare in cytokine biology; only IL-10 is known to have comparable scope and potency. Due to these powerful functions, IL-37 has aptly been called a ‘peacemaker’ in a Faculty of 1000 review.

Of the five IL37 mRNA isoforms a-e,IL37c and e lack exon 4, which encodes the first three β-strands of the IL-37 protein [Citation3]. IL-37b, comprising exons 1, 2 and 4–6 is the most abundantly expressed isoform in blood cells [Citation4], and by far the most studied. Extracellular IL-37 attenuates inflammation at picomolar concentrations [Citation3] utilizing its receptor IL-1R8:IL-18Rα [Citation5]. Data on expression of this heterodimer, particularly at protein level, are sparse.

In addition, IL-37 employs an intracellular pathway involving the transcriptional modulator Smad3 [Citation1], and via both mechanisms works intracellular switches to dampen inflammation, including transcription factors such as NF-κB and AP-1, second messengers such as STATs and MAP-kinases, and the metabolic checkpoint kinases mTOR and AMPK [Citation1,Citation5,Citation6]. By inhibiting mTOR and augmenting AMPK, IL-37 induces a pseudo-starvational state, which constitutes an additional anti-inflammatory mechanism aimed at achieving and maintaining metabolic homeostasis. Moreover, IL-37 suppresses IL-1β and IL-18 processing by the NLRP3 and AIM2 inflammasomes [Citation7]. Notably, many of the mediators reined in by IL-37 are themselves targets of inhibitory drugs in clinical use (e.g. IL-17 in psoriasis; TNF and mTOR in autoimmune disease). Although there is no murine homologue of IL-37, recombinant (rec) and transgenically expressed human IL-37 effectively protect mice from various inflammatory diseases [Citation1,Citation3,Citation8–10]. Given the clinical focus on several targets downstream of IL-37, its compelling characteristics such as multifaceted mechanisms of action and its success in preclinical models, IL-37-based drugs hold great potential.

2. Generating IL-37-based drugs

As a powerful roadblock of inflammatory responses, IL-37 function must be tightly regulated to avoid excessive suppression of innate and adaptive immunity. For example, in mice transgenic for IL-37 (IL-37tg), only little IL-37 protein is detectable under steady-state conditions despite the transgene’s constitutively active CMV promoter; abundance only increases upon inflammatory challenge [Citation1,Citation5,Citation8]. In addition to an mRNA instability element, such regulation is achieved by homodimerization. Cytokine multimerization is common, but in most cases enables signaling. In contrast, the bioactivity of the head-to-head homodimer that is readily assembled by IL-37 molecules even at low concentrations is substantially lower than that of IL-37 monomers [Citation3,Citation11]. This rare structure–function relationship can be exploited by mutating tyrosine in position 85 to alanine, thus preventing dimerization and generating recIL-37 with considerably enhanced bioactivity [Citation3]. To further improve drug-like properties, this IL-37 variant can be fused with an Fc-protein [Citation12].

3. IL-37 in disease

3.1. Interpreting the findings

Since the discovery of its function and its renaming in 2010 [Citation1], the number of reports on IL-37 has skyrocketed from less than 10 to more than 550 today. Notably, many of these publications focus on measuring IL-37 in various settings, while surprisingly few elaborate on mechanisms of action or directly explore therapeutic potential in vivo (). Interpreting changes in serum or tissue abundance is not straightforward for an anti-inflammatory mediator; in fact, we are often asked what to make of correlations between IL-37 and activity of a disease. Isn’t it counterintuitive to suggest treating an illness with IL-37 when its abundance increases with worsening of that illness?

Table 1. Data on IL-37 in diseases.

To justify clinical trials, which provide top-level evidence, relevance of a mediator to disease pathogenesis can be demonstrated using preclinical in vivo blocking experiments (second-level evidence). Evidence that can be considered third-level includes ex vivo-blockade in cultured cells as in [Citation1], or treatment with a mediator (e.g. recIL-37 [Citation5,Citation13] or via transgenic expression [Citation1,Citation8]). The absence of IL-37 in mice renders in vivo blockade challenging; thus, most available mechanistic and translational evidence on this cytokine falls in the last category. But what if not even this third level of evidence is available, and measurements of abundance is all we have?

Inflammation is vital to combat pathogens; but to regain and maintain healthy homeostasis, subsequent activation of counter-regulatory mechanisms is usually essential. Such activation can be compartmentalized, hence cytokine abundance may differ between compartments within the same individual. To combat the myriad of pathogens, specialized responses have evolved, which in turn require specific pathways and mediators for counter-balance. Besides IL-37, examples for such mediators include IL-1 receptor antagonist, soluble TNF-receptors, IL-10, and IL-18-binding protein. Therefore, it is likely that disease-associated local and/or systemic increases in IL-37 [Citation14,Citation15] are part of a targeted attempt to curtail a specific type of inflammation; however, this attempt is insufficient, at least at the time the disease was investigated. Albeit always speculatively, one might infer that augmenting IL-37 in this scenario could be useful.

The absence of change unfortunately commonly remains unreported (an exception being [Citation16]), which is disappointing because such absence is likely reasonable evidence that a mediator is not involved in the disease process, at least at the time and location of sampling. Local or systemic decreases in IL-37 abundance, occurring for example in necrotizing enterocolitis [Citation17], also require cautious, context-dependent interpretation, as they may indicate targeted dampening of its balancing activities, or a deficiency in IL-37 or the pathways involved in its activation.

3.2. Data in diseases

An overview of data on IL-37 in selected diseases is provided in .

3.3. Immuno-oncology (IO) – activating or neutralizing IL-37?

The immunosuppressive functions of IL-37 are not dissimilar from those of PD-1 and CTLA-4, both checkpoint inhibitors whose successful targeting has revolutionized medical care of hard-to-treat neoplasms. It is therefore not far-fetched to hypothesize that targeting IL-37 could be an attractive IO-strategy; and both augmenting or blocking its functions represent conceivable approaches in the vast field of cancer, in which every disease type and often even stage represents its own challenge with specific immunological circumstances that require tightly controlled manipulation. Therefore, a lot more research is required before translational decisions can be made.

4. Conclusions and perspectives

We argue that there is strong and robust evidence to support efforts aimed at taking the next translational steps toward deploying the peacemaking capacities of IL-37 in patients suffering from inflammatory or autoimmune disease. The unmet need in these fields is urgent, as existing medications not only have poor efficacy but also cause severe adverse effects and long-term morbidity that rival the damage done by the disease itself. A substantial number of high-quality publications support the conclusion that IL-37 represents an attractive novel treatment option. Stand-alone or combination strategies can be considered.

Having said this, one key question remains: Which clinical scenarios should be prioritized in such translational efforts? Considering the available evidence from the balcony perspective, we observe that the strongest data supporting therapeutic benefits of IL-37 arise from models of acute disease. Examples comprise studies on NEC (3 days), endotoxaemia (24 h), gout (4 h), stroke (1–3 days) or pulmonary inflammation in Covid (5 days), whereas data on longer-term models are sparse. The acute treatment concept is also supported physiologically, i.e. by the multiple auto-regulatory mechanisms designed to quickly shut down IL-37 function, and by the fact that IL-37 is an alarmin (a mediator that is stored intracellularly for example in monocytes, and can be released rapidly upon inflammatory stimulation of these cells [Citation4]). In other words, the immune system quickly activates the IL-37 pathway when it is needed to curtail acute inflammation, then terminates its activities just as quickly. In contrast to models of such acute inflammation, there is a paucity of evidence for IL-37 benefit in models of chronic disease (exceptions including asthma, 12 weeks, and murine intestinal inflammation, 7 days to ~2 months). This discrepancy may be due to a number of reasons other than IL-37 lacking activity in chronic disease (for instance barriers to publication of chronic models, such as greater logistical challenges, cost, and biological variability), and we consider continuing the preclinical exploration of IL-37 in chronic disease – and IO, for that matter – well justified. However, it appears sensible to prioritize moving forward with the first clinical trials designed to deliver the immense therapeutic potential of IL-37 to patients in acute inflammatory disease, including acute flares of chronic illnesses.

Declaration of interest

C A. Nold-Petry and M F. Nold hold three patent families on IL-37, namely PCT/AU2016/050495, PCT/AU2017/051443 (Monash University, Hudson Institute) and PCT/EP2020/087031 (Monash University, Hudson Institute, F. Hoffmann-La Roche). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This work was supported by the National Health and Medical Research Council Investigator Grant Leadership 1 APP1173584 to C A Nold-Petry, and by the Fielding Foundation’s Innovation Award and Fellowship to M F Nold.

References

  • Nold MF, Nold-Petry CA, Zepp JA, et al. IL-37 is a fundamental inhibitor of innate immunity. Nat Immunol. 2010 Nov;11(11):1014–1022.
  • Luo Y, Cai X, Liu S, et al. Suppression of antigen-specific adaptive immunity by IL-37 via induction of tolerogenic dendritic cells. PNAS. 2014 Oct 21;111(42):15178–15183.
  • Ellisdon AM, Nold-Petry CA, D’Andrea L, et al. Homodimerization attenuates the anti-inflammatory activity of interleukin-37. Sci Immunol. 2017 Feb 10;2(8). DOI:10.1126/sciimmunol.aaj1548.
  • Rudloff I, Cho SX, Lao JC, et al. Monocytes and dendritic cells are the primary sources of interleukin 37 in human immune cells. J Leukoc Biol. 2017 Apr;101(4):901–911.
  • Nold-Petry CA, Lo CY, Rudloff I, et al. IL-37 requires the receptors IL-18Ralpha and IL-1R8 (SIGIRR) to carry out its multifaceted anti-inflammatory program upon innate signal transduction. Nat Immunol. 2015 Apr;16(4):354–365.
  • Ballak DB, van Diepen JA, Moschen AR, et al. IL-37 protects against obesity-induced inflammation and insulin resistance. Nat Commun. 2014 Sep 3;5(1):4711.
  • Rudloff I, Ung HK, Dowling JK, et al. Parsing the IL-37-mediated suppression of inflammasome function. Cells. 2020 Jan 10;9(1):178.
  • McNamee EN, Masterson JC, Jedlicka P, et al. Interleukin 37 expression protects mice from colitis. PNAS. 2011 Oct 4;108(40):16711–16716.
  • Lunding L, Webering S, Vock C, et al. IL-37 requires IL-18Ralpha and SIGIRR/IL-1R8 to diminish allergic airway inflammation in mice. Allergy. 2015 Apr;70(4):366–373.
  • Bulau AM, Fink M, Maucksch C, et al. In vivo expression of interleukin-37 reduces local and systemic inflammation in concanavalin A-induced hepatitis. ScientificWorldJournal. 2011;11:2480–2490.
  • Eisenmesser EZ, Gottschlich A, Redzic JS, et al. Interleukin-37 monomer is the active form for reducing innate immunity. PNAS. 2019 Mar 19;116(12):5514–5522.
  • Bujotzek A, Tiefenthaler G, Lariviere L, et al. Protein engineering of a stable and potent anti-inflammatory IL-37-Fc fusion with enhanced therapeutic potential. Cell Chem Biol. 2021 Oct 19;29(4):586–596.e4.
  • Cavalli G, Koenders M, Kalabokis V, et al. Treating experimental arthritis with the innate immune inhibitor interleukin-37 reduces joint and systemic inflammation. Rheumatology (Oxford). 2016 Dec;55(12):2220–2229.
  • Cao S, Shi H, Sun G, et al. Serum IL-37 level is associated with rheumatoid arthritis and disease activity: a meta-analysis. Biomed Res Int. 2021;2021:6653439.
  • Ye L, Ji L, Wen Z, et al. IL-37 inhibits the production of inflammatory cytokines in peripheral blood mononuclear cells of patients with systemic lupus erythematosus: its correlation with disease activity. J Transl Med. 2014 Mar 16;12(1):69.
  • Godsell J, Rudloff I, Kandane-Rathnayake R, et al. Clinical associations of IL-10 and IL-37 in systemic lupus erythematosus. Sci Rep. 2016 Oct 6;6(1):34604.
  • Cho SX, Rudloff I, Lao JC, et al. Characterization of the pathoimmunology of necrotizing enterocolitis reveals novel therapeutic opportunities. Nat Commun. 2020 Nov 13;11(1):5794.
  • Imaeda H, Takahashi K, Fujimoto T, et al. Epithelial expression of interleukin-37b in inflammatory bowel disease. Clin Exp Immunol. 2013 Jun;172(3):410–416.
  • Xia L, Shen H, Lu J. Elevated serum and synovial fluid levels of interleukin-37 in patients with rheumatoid arthritis: attenuated the production of inflammatory cytokines. Cytokine. 2015 Dec;76(2):553–557.
  • Kluck V, van Deuren RC, Cavalli G, et al. Rare genetic variants in interleukin-37 link this anti-inflammatory cytokine to the pathogenesis and treatment of gout. Ann Rheum Dis. 2020 Apr;79(4):536–544.
  • Xu J, Chen J, Li W, et al. Additive therapeutic effects of mesenchymal stem cells and IL-37 for systemic lupus erythematosus. J Am Soc Nephrol. 2020 Jan;31(1):54–65.
  • Tawfik MG, Nasef SI, Omar HH, et al. Serum Interleukin-37: a new player in Lupus Nephritis? Int J Rheum Dis. 2017 Aug;20(8):996–1001.
  • Li A, Ling Y, Song Z, et al. Correlation between early plasma interleukin 37 responses with low inflammatory cytokine levels and benign clinical outcomes in severe acute respiratory syndrome coronavirus 2 infection. J Infect Dis. 2021 Feb 24;223(4):568–580.
  • Christie MJ, Irving AT, Forster SC, et al. Of bats and men: immunomodulatory treatment options for COVID-19 guided by the immunopathology of SARS-CoV-2 infection. Sci Immunol. 2021 Sep 17;6(63):eabd0205.
  • Schroder A, Lunding LP, Zissler UM, et al. IL-37 regulates allergic inflammation by counterbalancing pro-inflammatory IL-1 and IL-33. Allergy. 2022 Mar;77(3):856–869.
  • Zhang SR, Nold MF, Tang SC, et al. IL-37 increases in patients after ischemic stroke and protects from inflammatory brain injury, motor impairment and lung infection in mice. Sci Rep. 2019 May 6;9(1):6922.
  • Wu B, Meng K, Ji Q, et al. Interleukin-37 ameliorates myocardial ischaemia/reperfusion injury in mice. Clin Exp Immunol. 2014 Jun;176(3):438–451.
  • Yang Y, Zhang ZX, Lian D, et al. IL-37 inhibits IL-18-induced tubular epithelial cell expression of pro-inflammatory cytokines and renal ischemia-reperfusion injury. Kidney Int. 2015 Feb;87(2):396–408.
  • Ji Q, Meng K, Yu K, et al. Exogenous interleukin 37 ameliorates atherosclerosis via inducing the Treg response in ApoE-deficient mice. Sci Rep. 2017 Jun 12;7(1):3310.
  • Cavalli G, Justice JN, and Boyle KE, et al. Interleukin 37 reverses the metabolic cost of inflammation, increases oxidative respiration, and improves exercise tolerance. PNAS. 2017 Feb 28;114(9):2313–2318.
  • Ballak DB, Li S, Cavalli G, et al. Interleukin-37 treatment of mice with metabolic syndrome improves insulin sensitivity and reduces pro-inflammatory cytokine production in adipose tissue. J Biol Chem. 2018 Sep 14;293(37):14224–14236.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.