Elsevier

Seminars in Immunology

Volume 25, Issue 6, 15 December 2013, Pages 469-484
Seminars in Immunology

Review
Treating inflammation by blocking interleukin-1 in humans

https://doi.org/10.1016/j.smim.2013.10.008Get rights and content

Abstract

IL-1 is a master cytokine of local and systemic inflammation. With the availability of specific IL-1 targeting therapies, a broadening list of diseases has revealed the pathologic role of IL-1-mediated inflammation. Although IL-1, either IL-1α or IL-1β, was administered to patients in order to improve bone marrow function or increase host immune responses to cancer, these patients experienced unacceptable toxicity with fever, anorexia, myalgias, arthralgias, fatigue, gastrointestinal upset and sleep disturbances; frank hypotension occurred. Thus it was not unexpected that specific pharmacological blockade of IL-1 activity in inflammatory diseases would be beneficial. Monotherapy blocking IL-1 activity in a broad spectrum of inflammatory syndromes results in a rapid and sustained reduction in disease severity. In common conditions such as heart failure and gout arthritis, IL-1 blockade can be effective therapy. Three IL-1blockers have been approved: the IL-1 receptor antagonist, anakinra, blocks the IL-1 receptor and therefore reduces the activity of IL-1α and IL-1β. A soluble decoy receptor, rilonacept, and a neutralizing monoclonal anti-interleukin-1β antibody, canakinumab, are also approved. A monoclonal antibody directed against the IL-1 receptor and a neutralizing anti-IL-1α are in clinical trials. By specifically blocking IL-1, we have learned a great deal about the role of this cytokine in inflammation but equally important, reducing IL-1 activity has lifted the burden of disease for many patients.

Introduction

The importance of IL-1 as a master cytokine in inflammation comes from infants born with a loss of function mutation in the naturally occurring endogenous IL-1 receptor antagonist (IL-1Ra). IL-1Ra blocks the IL-1 receptor type 1 (IL-1R1), which is on all cells; therefore, systemic inflammation may come for either IL-1α or IL-1β. These infants succumb early in life with overwhelming sterile inflammation of the skin, joints and bone with large numbers of infiltrating neutrophils and high levels of interleukin-17 [1], [2]. The condition is called deficiency of interleukin-1 antagonist (DIRA) and daily treatment with anakinra rapidly reverses the inflammation and prevents a fatal outcome. Mice deficient in IL-1Ra are similarly affected in that these mice develop spontaneous inflammation such as a rheumatoid arthritis-like disease and can succumb to lethal arteritis. The best evidence for a role for either IL-1α or IL-1β in disease comes from specific blockade, as correlations of circulating levels and disease severity are not informative and do not establish causality. Even in the most severe IL-1β-mediated autoinflammatory diseases, IL-1β levels in the circulation increase only by factor of five [3].

There are two IL-1's. IL-1α is expressed as a precursor and is constitutively present in most cells of healthy subjects. The cytokine is found in normal keratinocytes of the skin, the epithelial cells of mucosal membranes throughout the body and the cells of organs such as the liver, lung and kidney. Platelets also contain IL-1α. The entire endothelium of the vasculature contains the IL-1α precursor and in membrane fragments from the endothelium termed “apoptotic bodies” [4]. These membrane fragments are active in inducing neutrophil infiltration [4] in several inflammatory conditions of the blood vessels termed vasculitis [5]. During ischemia, however, cell death by necrosis takes place and the IL-1α precursor is released [6], [7].

In contrast, IL-1β is not present in health or at levels not detected by standard assays. IL-1β is a product of blood monocytes, tissue macrophages and dendritic cells. The rate-limiting step in the production of IL-1β is transcription, but IL-1β mRNA requires an additional signal for synthesis. The stimulus can be a microbial product but cytokines, such as TNFα, IL-18, IL-1α or IL-1β itself induce IL-1β [8]. In fact, IL-1 induction of itself is part of the mechanism of “autoinflammation”. IL-1β is first synthesized as an inactive precursor but the precursor requires cleavage by caspase-1, an intracellular cysteine protease. Caspase-1 itself requires activation in order to process IL-1β into an active cytokine. The activation of caspase-1 proceeds following the oligomerization of a complex of intracellular proteins termed the “inflammasome” by the late Tschopp [9], [10]. With activation of caspase-1, the N-terminal amino acids are cut and mature IL-1β is readily secreted as an active cytokine. One of the components of the inflammasome termed “cryopyrin” (also termed NLRP3) plays a critical role in the secretion of IL-1β. A single amino acid mutation in cryopyrin [11] results in enhanced caspase-1 activity and greater secretion of IL-1β⋅ Increased production and secretion of IL-1β from blood monocytes is characteristic of a group of autoinflammatory diseases termed “cryopyrin associated periodic syndrome (CAPS)”.

Section snippets

Autoinflammatory diseases are different from autoimmune diseases

Autoinflammatory diseases are chronic, debilitating syndromes [12]. As stated above, some autoinflammatory conditions are due to mutations in the intracellular proteins that control caspase-1, the enzyme that converts IL-1β to an active cytokine prior to release from the cell. Although these conditions are rare, the inflammatory manifestations are common to many diseases. Blood monocytes from patients with autoinflammatory diseases release more IL-1β compared to cells from healthy persons [13],

Treating a broad spectrum of inflammatory conditions with anakinra

The IL-1 receptor is expressed in nearly all tissues and thus anakinra provides an optimal therapy, as the antagonist prevents the binding of either IL-1α or IL-1β. However, how much of the efficacy of anakinra is due to blocking IL-1α and how much is due to blocking IL-1β? The amount of IL-1β that circulates in IL-1-mediated inflammatory conditions is in the low nanogram range [3]; IL-1α is rarely found in the circulation. Therefore, the use of anakinra has provided the data on the role of

IL-1 and acute onset diseases

IL-1-mediated inflammation contributes to catastrophic events such acute lung injury, myocardial infarction, acute kidney failure and stroke with end organ failure. There is no dearth of animal studies demonstrating an essential role for IL-1 following an ischemic injury of the heart [21], lung [22], liver [23], kidney [24] and brain [25] (Fig. 1). Inflammation following an ischemic event is characterized by infiltration of neutrophils and myeloid precursors into the surrounding ischemic area,

Post-infarction cardiac remodeling

Based on a large number of animal studies, blocking IL-1 in humans with heart disease has entered clinical medicine [27]. Anakinra has been used successfully in patients with ST-elevation myocardial infarction (STEMI) [28] and repeated in a second trial [29]. Both trials were randomized and placebo controlled. STEMI has a high risk of death but patients who survive the acute event often progress to chronic heart failure due to loss of viable myocardium. In the first randomized,

Type-1 diabetes

In 1986, the Danish scientists Mandrup-Poulsen and colleagues published their findings that picomolar concentrations of IL-1β were selectively toxic for the insulin-producing pancreatic beta-cell (reviewed in [35]). These studies resulted in a paradigm change for the pathogenesis of Type-1 diabetes in that a macrophage product rather than a cytotoxic T-cell became the target for salvaging the beta-cell. In the non-obese diabetic mouse strain, the sine qua non model for Type-1 diabetes, IL-1

Rheumatoid arthritis

Anakinra has been studied in several controlled studies in patients with rheumatoid arthritis with and without methotrexate (reviewed in [61]) (Table 1). Overall, each study revealed a statistically significant reduction in disease severity, improvement in quality of life and a decrease in radiographic evidence of joint space narrowing. Because of the half-life of 6 h, daily injections of anakinra are required. The subcutaneous injections can produce injection site reactions, although these

Diseases of Mendelian inheritance

A group of rare genetic disorders, which have a common phenotype of recurrent fevers, debilitating fatigue, myalgia, arthralgia, gastrointestinal symptoms and skin rashes, are treated with IL-1 blocking therapies. These include FMF, CAPS, TRAPS, HIDS, MKD and PAPA. Neutrophilia, elevated hepatic acute phase proteins and increased erythrocyte sedimentation rates are characteristic during the episodes. A common feature to nearly all hereditary autoinflammatory syndromes is the rapid and sustained

Chronic inflammatory diseases

A growing number of chronic inflammatory disorders without a known genetic basis respond to reducing IL-1 activity. For example, treating idiopathic recurrent pericarditis with immunosuppressive agents, anti-TNFα antibodies, non-steroidal anti-inflammatory agents, intravenous gamma globulin or high dose glucocorticoids often results partial remissions whereas anakinra (Table 3) provides complete remission in all cases reported [121], [122], [123], [124]. Although most chronic inflammatory

Mental impairment and hearing loss

CAPS patients exhibit various neurologic abnormalities such as aseptic lepo-meningitis and thus reflecting IL-1-mediated inflammation in the brain. In a study of CAPS patients, 92% had headache with features of migraine, 54% had sensorineural deafness and 46% had papilledema [110]. Treatment with either anakinra or canakinumab leads to complete resolution of symptoms [110], [113], [151]. Children with severe CAPS show manifestations of elevated intracranial pressure and are believed to be

Anti-IL-1α

The monoclonal antibody against IL-1α is being tested in Type 2 diabetes, cancer, cancer cachexia, leukemia, psoriasis, vascular disease and scarring acne vulgaris. Indeed, inflammation resulting from ischemic damage starts with the release of the IL-1α precursor from dying cells. The IL-1α is active (Fig. 2) but as the inflammation progresses, IL-1β becomes the dominant cytokine the process becomes an IL-1β-mediated process. Due to its specificity and long half-life, neutralization of IL-1β is

Smoldering/indolent myeloma

In the microenvironment of the bone marrow, IL-1β produced by myeloma precursor plasma cells stimulates the stromal cells to release large amounts of IL-6, which in turn promotes the survival and expansion of the pre-myeloma cells [175]. It was reasoned that in the indolent stages of multiple myeloma, blocking IL-1β would reduce IL-6 activity [176]. Patients with smoldering or indolent myeloma at high risk for progression to multiple myeloma were selected with the clinical objective of slowing

IL-1 and host defense against infection

Since its introduction in 2002, anakinra has had a remarkable record of safety [61], [177]. It is estimated that over 150,000 patients have received anakinra, some treated daily for over 10 years. Anakinra has been administered to patients with active infections [178], [179]. Following the introduction of anti-TNFα blocking therapies, a wide-spectrum of opportunistic infections were reported, similar to those observed in immunosuppressed persons. Host defense against opportunistic organisms as

Acknowledgements

The author thanks A. Abbate, M. Donath, T. Mandrup-Poulsen, M. G. Netea, B, Pilstrom, and Anna Simon for their assistance with this review. This work is supported by NIH Grants AI-15614, AR-45584 (to CAD).

References (354)

  • S. Gosavi et al.

    Topological frustration and the folding of interleukin-1 beta

    J Mol Biol

    (2006)
  • S. Reddy et al.

    An autoinflammatory disease due to homozygous deletion of the IL1RN locus

    N Engl J Med

    (2009)
  • I. Aksentijevich et al.

    An autoinflammatory disease with deficiency of the interleukin-1-receptor antagonist

    N Engl J Med

    (2009)
  • H.J. Lachmann et al.

    In vivo regulation of interleukin 1beta in patients with cryopyrin-associated periodic syndromes

    J Exp Med

    (2009)
  • Y. Berda-Haddad et al.

    Sterile inflammation of endothelial cell-derived apoptotic bodies is mediated by interleukin-1alpha

    Proc Natl Acad Sci U S A

    (2011)
  • C. Beyer et al.

    The role of microparticles in the pathogenesis of rheumatic diseases

    Nat Rev Rheumatol

    (2011)
  • P. Rider et al.

    IL-1α and IL-1β recruit different myeloid cells and promote different stages of sterile inflammation

    J Immunol

    (2011)
  • I. Cohen et al.

    Differential release of chromatin-bound IL-1alpha discriminates between necrotic and apoptotic cell death by the ability to induce sterile inflammation

    Proc Natl Acad Sci U S A

    (2010)
  • C.A. Dinarello et al.

    Interleukin 1 induces interleukin 1. I. Induction of circulating interleukin 1 in rabbits in vivo and in human mononuclear cells in vitro

    J Immunol

    (1987)
  • F. Martinon et al.

    The inflammasomes: guardians of the body

    Annu Rev Immunol

    (2009)
  • S.L. Masters et al.

    Horror autoinflammaticus: the molecular pathophysiology of autoinflammatory disease

    Annu Rev Immunol

    (2009)
  • M. Gattorno et al.

    The pattern of response to anti-interleukin-1 treatment distinguishes two subsets of patients with systemic-onset juvenile idiopathic arthritis

    Arthritis Rheum

    (2008)
  • M. Gattorno et al.

    Pattern of interleukin-1beta secretion in response to lipopolysaccharide and ATP before and after interleukin-1 blockade in patients with CIAS1 mutations

    Arthritis Rheum

    (2007)
  • R. Goldbach-Mansky et al.

    Neonatal-onset multisystem inflammatory disease responsive to interleukin-1beta inhibition

    N Engl J Med

    (2006)
  • V. Pascual et al.

    Role of interleukin-1 (IL-1) in the pathogenesis of systemic onset juvenile idiopathic arthritis and clinical response to IL-1 blockade

    J Exp Med

    (2005)
  • M. Colina et al.

    Dysregulation of P2X7 receptor-inflammasome axis in SAPHO syndrome: successful treatment with anakinra

    Rheumatology (Oxford)

    (2010)
  • C.A. Dinarello

    How interleukin-1beta induces gouty arthritis

    Arthritis Rheum

    (2010)
  • M.Y. Donath et al.

    Type 2 diabetes as an inflammatory disease

    Nat Rev Immunol

    (2011)
  • A. Abbate et al.

    Anakinra, a recombinant human interleukin-1 receptor antagonist, inhibits apoptosis in experimental acute myocardial infarction

    Circulation

    (2008)
  • E. Abraham et al.

    Effects of therapy with interleukin-1 receptor antagonist on pulmonary cytokine expression following hemorrhage and resuscitation

    Lymphokine Cytokine Res

    (1994)
  • C.M. Calkins et al.

    IL-1 regulates in vivo C-X-C chemokine induction and neutrophil sequestration following endotoxemia

    J Endotoxin Res

    (2002)
  • K. Rusai et al.

    Administration of interleukin-1 receptor antagonist ameliorates renal ischemia-reperfusion injury

    Transpl Int

    (2008)
  • H. Boutin et al.

    Role of IL-1alpha and IL-1beta in ischemic brain damage

    J Neurosci

    (2001)
  • H.C. Emsley et al.

    A randomised phase II study of interleukin-1 receptor antagonist in acute stroke patients

    J Neurol Neurosurg Psychiatry

    (2005)
  • A. Abbate et al.

    Blocking interleukin-1 as a novel therapeutic strategy for secondary prevention of cardiovascular events

    BioDrugs

    (2012)
  • A. Abbate et al.

    Interleukin-1beta modulation using a genetically engineered antibody prevents adverse cardiac remodelling following acute myocardial infarction in the mouse

    Eur J Heart Fail

    (2010)
  • B.W. Van Tassell et al.

    Enhanced interleukin-1 activity contributes to exercise intolerance in patients with systolic heart failure

    PLOS One

    (2012)
  • I. Ikonomidis et al.

    Inhibition of interleukin-1 by anakinra improves vascular and left ventricular function in patients with rheumatoid arthritis

    Circulation

    (2008)
  • B.S. Cain et al.

    Tumor necrosis factor-α and interleukin-1β synergistically depress human myocardial function

    Crit Care Med

    (1999)
  • B.J. Pomerantz et al.

    Inhibition of caspase 1 reduces human myocardial ischemic dysfunction via inhibition of IL-18 and IL-1beta

    Proc Natl Acad Sci U S A

    (2001)
  • B.W. Van Tassell et al.

    Effects of interleukin-1 blockade with anakinra on aerobic exercise capacity in patients with heart failure with preserved ejection fraction

    Am Heart J

    (2013)
  • T. Mandrup-Poulsen et al.

    Blockade of interleukin 1 in type 1 diabetes mellitus

    Nat Rev Endocrinol

    (2010)
  • M.D. Dayer-Metroz et al.

    IL-1ra delays the spontaenous autoimmune diabetes in the BB rat

    Eur J Clin Invest

    (1992)
  • K.M. Sumpter et al.

    Preliminary studies related to anti-interleukin-1beta therapy in children with newly diagnosed type 1 diabetes

    Pediatr Diabetes

    (2011)
  • N. Ruperto et al.

    Evaluation of safety and preliminary efficacy of canakinumab in children with systemic onset juvenile idiopaththic artritis

    Arthr Rheumat.

    (2009)
  • C.H. Sibley et al.

    Sustained response and prevention of damage progression in patients with neonatal-onset multisystem inflammatory disease treated with anakinra: a cohort study to determine three- and five-year outcomes

    Arthritis Rheum.

    (2012)
  • J.F. Bach et al.

    A historical view from thirty eventful years of immunotherapy in autoimmune diabetes

    Semin Immunol

    (2012)
  • J.F. Bach et al.

    Factors predictive of cyclosporine-induced nephrotoxicity: the role of cyclosporine blood levels

    Transplant Proc

    (1990)
  • K.C. Herold et al.

    Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus

    N Engl J Med

    (2002)
  • M.D. Pescovitz et al.

    Rituximab, B-lymphocyte depletion, and preservation of beta-cell function

    N Engl J Med

    (2009)
  • Cited by (443)

    View all citing articles on Scopus
    View full text