You are on page 1of 11

Expert Review of Clinical Pharmacology

ISSN: 1751-2433 (Print) 1751-2441 (Online) Journal homepage: http://www.tandfonline.com/loi/ierj20

Pharmacological treatment options for cryopyrin-


associated periodic syndromes

Emmanuelle C. Landmann & Ulrich A. Walker

To cite this article: Emmanuelle C. Landmann & Ulrich A. Walker (2017) Pharmacological
treatment options for cryopyrin-associated periodic syndromes, Expert Review of Clinical
Pharmacology, 10:8, 855-864, DOI: 10.1080/17512433.2017.1338946

To link to this article: http://dx.doi.org/10.1080/17512433.2017.1338946

Accepted author version posted online: 06


Jun 2017.
Published online: 20 Jun 2017.

Submit your article to this journal

Article views: 114

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=ierj20

Download by: [University of New England] Date: 08 November 2017, At: 03:08
EXPERT REVIEW OF CLINICAL PHARMACOLOGY, 2017
VOL. 10, NO. 8, 855–864
https://doi.org/10.1080/17512433.2017.1338946

REVIEW

Pharmacological treatment options for cryopyrin-associated periodic syndromes


Emmanuelle C. Landmann and Ulrich A. Walker
Department of Rheumatology, University Hospital Basel, Basel, Switzerland

ABSTRACT ARTICLE HISTORY


Introduction: Cryopyrin-associated periodic syndromes (CAPS) are rare monogenic autoinflammatory dis- Received 3 April 2017
eases, comprising a spectrum of phenotypes of varying severity. CAPS are associated with gain-of-function Accepted 2 June 2017
mutations in the NLRP3 inflammasome, a multiprotein complex critical for the activation of IL-1ß, and are KEYWORDS
characterized by episodes of fever, urticaria-like rash, musculoskeletal, ocular, and neurological symptoms. Anakinra; autoinflammatory;
Areas covered: Accounting for the pivotal role of IL-1ß in the pathogenesis of CAPS, three therapeutic canakinumab;
options, all blocking the action of IL-1ß, are currently approved: anakinra, a recombinant IL-1 receptor Cryopyrin-associated
antagonist, the IL-1 trap rilonacept and canakinumab, a monoclonal anti-IL-1ß antibody. All agents reduce periodic syndrome; Familial
Downloaded by [University of New England] at 03:08 08 November 2017

or even resolve clinical symptoms, biochemical activity markers and improve quality of life in CAPS. This Cold Autoinflammatory
review also covers pharmacokinetic, pharmacodynamic and safety aspects of the approved drugs and the Syndrome; interleukin-1;
Muckle-Wells Syndrome;
potential utility of IL-1β blockers in a wide range of other conditions with an autoinflammatory component. Neonatal Onset Multisystem
Expert commentary: Due to the success story of current pharmaceutics, the therapeutic options in Inflammatory Disease;
CAPS are not expected to expand in the near future. Prospective observational studies are needed to NLRP3 inflammasome;
confirm long-term efficacy and sustained benefit. New IL-1ß blockers will likely address unmet clinical rilonacept
needs in other autoinflammatory conditions.

1. Introduction along with the precursors of a number of other IL-1 cytokine family
members [2]. The NLRP3 inflammasome is activated by various
Cryopyrin-associated periodic syndromes (CAPS) are a spectrum of
pathogen-associated molecules, but also by danger signals
rare monogenic autoinflammatory syndromes arising from an
released by stressed and damaged cells [3,4].
overactive NLRP3 inflammasome in innate immune cells and are
characterized by periodic fever episodes and systemic inflamma-
tion. Autoantibodies or autoreactive effectors of the acquired 3. Cryopyrin-associated periodic syndromes
immune system have no role in the pathogenesis of CAPS [1].
CAPS are a group of autoinflammatory diseases, which com-
Current therapeutic approaches of CAPS target IL-1ß with a highly
prises familial cold autoinflammatory syndrome (FCAS),
successful outcome, emphasizing the crucial role of IL-1ß in the
Muckle–Wells syndrome (MWS), and chronic infantile neurolo-
pathogenesis of CAPS. This review aims to provide an overview of
gical cutaneous and articular syndrome (CINCA), also called
the currently licensed drugs for CAPS and to discuss future
neonatal onset multisystem inflammatory disease (NOMID).
developments.
The spectrum of disease severity ranges from FCAS being
the mildest presentation, to MWS as an intermediate, and
2. NLRP3 inflammasome CINCA as the most severe form [5,6].
At the molecular level, CAPS are characterized by missense
IL-1ß is a potent proinflammatory cytokine, mainly originating
single-nucleotide substitutions in the NLRP3-gene on the long
from cells of the innate immune system [2]. Its inactive precursor
arm of chromosome 1 leading to a gain-of-function mutation in
pro-IL-1ß is cleaved intracellularly by the nucleotide-binding oli-
cryopyrin, the NLRP3-gene product [7–10]. The majority of the 181
gomerization domain-like receptor family, pyrin domain contain-
[11] so far identified sequence variants are located in exon 3, which
ing 3 (NLRP3) inflammasome, a multiprotein complex. NLRP3 (also
encodes the NACHT-domain, and its flanking regions [7,9–11]. In
referred to as cryopyrin) is a member of the nucleotide-binding
more than half of the cases, the mutations are inherited in an
oligomerization domain (NOD)-like receptor (NLR) family, and is
autosomal-dominant fashion but somatic mutations are described
composed of a nucleotide-binding NACHT domain, which is
in around 20–35% of patients, particularly in CINCA [7–9,12].
flanked by a leucine-rich repeat (LRR)-sequence on the C’-terminus
Moreover, a number of patients are mutation negative, and may
and a pyrin (PYD)-domain on the N’-Terminus. In order to form the
harbor currently unknown mutation variants, or feature low-level
NLRP3-inflammasome, cryopyrin binds to the intracellular protein
somatic mosaicism [12–15]. Despite the common molecular back-
apoptosis-associated speck-like protein (ASC), that in turn links to
ground, the diagnosis of CAPS is however based on clinical char-
caspase-1 [3]. The latter then cleaves IL-1ß into its active form,
acteristics and the verification of a NLRP3 mutation is not

CONTACT Ulrich A. Walker ulrich.walker@usb.ch Department of Rheumatology, University Hospital Basel, Petersgraben 4, Basel CH-4031, Switzerland
© 2017 Informa UK Limited, trading as Taylor & Francis Group
856 E. C. LANDMANN AND U. A. WALKER

mandatory [16]. Due to the identification of the NLRP3 mutations, 4. Therapeutic options for CAPS
CAPS are now understood as a continuum of phenotypes rather
CAPS flares tremendously impair the quality of life in both,
than three distinct entities.
its physical and psychological determinants [35–37]. Thus,
The cryopyrin variants elicit systemic inflammation
treatment is indicated as soon as CAPS has been diagnosed,
through a constitutive gain of activity of cryopyrin and the
also because of the favorable benefit–risk profile of the
inflammasome, although details are still elusive. In vitro
currently available agents.
studies demonstrate that the mutated cryopyrin confers an
To date, the European Medicines Agency (EMA) and US
excessive and dysregulated secretion of active IL-1ß in the
FDA approved three different medications in the treatment
innate immune system. Monocytes of patients with FCAS
for CAPS, all of which block the action of IL-1ß. These drugs
[17], MWS [18], and CINCA [19] excessively secrete IL-1ß
are the short-acting recombinant IL-1 receptor antagonist
[18–22] spontaneously, after stimulation with lipopolysac-
anakinra, the IL-1ß trap rilonacept and canakinumab, a mono-
charide, or at mild hypothermia. Similarly, CAPS-patients
clonal antibody against IL-1ß [38]. The efficacy and safety of
feature approximately five-fold higher serum levels of IL-1ß
these agents in CAPS were investigated in a number of stu-
than healthy controls [23].
dies. Table 1 provides an overview of the approved indica-
The prevalence of CAPS may be higher in Caucasians than
tions, dosage, and preliminary use in other conditions.
in other ethnicities, but generally is very low, with an estimate
Prior to the advent of the IL-1 antagonists, the traditional
of 1 case in every 360ʹ000 in France [5,7]. Consistent with the
Downloaded by [University of New England] at 03:08 08 November 2017

treatment of CAPS consisted of various immunosuppressive


autosomal-dominant inheritance, there are no gender differ-
agents. An anecdotal report describes a benefit with cyclospor-
ences in the prevalence of CAPS [5,16,24].
ine A, high-dose corticosteroids, mycophenolate, and thalido-
The disease onset of most CAPS is typically in the neo-
mide [61]. Methotrexate was also reported to provide partial
natal period or at early childhood [5,7,24] with recurrent
relief [62,63]. Tocilizumab, an anti-IL-6 receptor monoclonal anti-
episodes of fever and an urticaria-like rash. Aside from
body [64] and TNFα-blockers [65] failed to reliably suppress
fever, the urticaria-like rash is considered the key symptom
symptoms and to normalize laboratory signs of activity.
and generally the first manifestation in all phenotypes [25].
Nonsteroidal antirheumatic drugs are however frequently used
CAPS are also accompanied by signs of IL-1ß-triggered sys-
to assist in alleviating symptoms [61–63].
temic inflammation, such as fatigue, elevated neutrophil
counts, and acute-phase reactants (C-reactive protein
(CRP), erythrocyte sedimentation rate (ESR), and serum amy-
loid A (SAA)) [2,25]. Musculoskeletal symptoms are also
4.1. Anakinra (Kineret ) ®
common and tend to increase in severity over time [26]. Anakinra was the first IL-1 targeting drug on the market. It was
Ocular, auditory, and neurological manifestations may also studied in rheumatoid arthritis since 1993 [66,67], and in CAPS
be observed in some phenotypes [16,26]. since 2003 [68]. Anakinra has been approved by the FDA for
FCAS is characterized by recurrent, short episodes (hours CINCA in 2012 [69] and by the EMA in 2013 for all subtypes of
to a few days) of fever, rash, and polyarthralgia, that are CAPS in persons of at least 8 months of age and a body weight of
triggered by exposure to cold temperatures and may be at least 10 kg [70]. Anakinra is a recombinant, nonglycosylated
associated with conjunctivitis and headache. The intensity version of the physiological human IL-1 receptor antagonist (IL-
of the cold trigger correlates with symptom severity, but 1RA) that competitively inhibits the binding of IL-1ß and IL-1α to
symptoms follow a circadian rhythm as well, being worse in the IL-1 receptor, thereby blocking downstream signaling [70].
the evening [25,27,28]. Anakinra is produced by recombinant DNA technology in
MWS presents either with recurrent flares or virtually per- Escherichia coli. The amino-acid structure of anakinra is identical
manent symptoms of rash, arthralgia, occasional joint effu- to that of the native IL-1RA, except for an additional methionine
sions, and conjunctivitis [29]. Again, fever attacks can be residue on its N’-terminus [70]. The half-life of anakinra ranges
triggered by cold temperature [29] but also be absent [25]. from 4 to 6 h [70], therefore anakinra is administered daily by
Late complications consist of progressive bilateral sensori- subcutaneous injection with a recommended dose of 1–2 mg/kg
neural hearing loss, and AA amyloidosis with gradual renal in children [69,70], and 100 mg in adults [61,71,72]. In severe
failure in approximately one-fourth of the patients [29–31]. cases stepwise dose escalation to 8 mg/kg [69,70], or even to
CINCA, the most severe phenotype is typically associated 10 mg/kg [73] is suggested. Since the drug is principally elimi-
with fever, rash, musculoskeletal, ocular and central nervous nated by the kidney [74], the dose interval should be doubled in
manifestations. The musculoskeletal complications are more severe renal failure or end-stage renal disease (creatinine clear-
severe in CINCA than in the other phenotypes and range from ance below 30 ml/min) [69,70]. Although numerous individuals
arthritis in the majority of the cases, to striking skeletal dys- with CAPS have developed antibodies against anakinra, no effect
morphia with cartilage anomalies, and overgrowth of the on efficacy or safety was reported [70].
patella and the epiphyses of long bones [25,26,32,33]. Owing to its early approval for rheumatoid arthritis, anakinra
Central nervous manifestations consist of aseptic meningitis was the first drug studied in CAPS. In 2003, Hawkins et al. [68]
and increased intracranial pressure leading to morning head- demonstrated a remarkable response to anakinra in MWS. Over
ache, papilledema, optic disc atrophy [33], ocular disability the years, further studies revealed rapid responses to anakinra, not
[34], and cognitive impairment [33]. Anterior uveitis is also only in MWS [29,71,72], but also in FCAS [28], CINCA [65,73,75] and
common [34]. Similar to MWS, perceptive deafness [33] and overlap phenotypes such as CINCA/MWS [36], and MWS/FCAS
amyloidosis [25] develop with disease progression. [76]. Clinical improvement was observed as early as four hours
EXPERT REVIEW OF CLINICAL PHARMACOLOGY 857

Table 1. Approved drugs that target IL-1 in CAPS and trials outside licensed indications.
Anakinra Rilonacept Canakinumab
Approved indications FDA: FDA: FDA:
and year of approval - RA ≥18 years (in 2001) - FCAS, MWS ≥12 years (in 2008) - FCAS, MWS ≥4 years (in 2009)
- CINCA (in 2012) EMA: - SJIA ≥2 years (in 2013)
EMA: - CAPS (in 2009, withdrawal in 2012) EMA:
- RA in combination with methotrexate (in - periodic fever syndromes ≥2 years:
2002) CAPS; TRAPS, HIDS/MKD, FMF (in 2009 and
- all subtypes of CAPS ≥8 months or 2016, respectively)
≥10 kg (in 2013) - Still’s disease (in 2013)
- Gout with at least 3 flares per year (in 2013)
Dose CAPS: 1–2 mg/kg s.c. daily CAPS: weekly CAPS: every 8 weeks
RA: 100 mg s.c. daily ≥18 years: loading dose 320 mg s.c., >40 kg 150 mg s.c.
maintenance dose 160 mg s.c. ≥15 kg and ≤40 kg 2 mg/kg s.c.
≥12 and ≤17 years: loading dose ≥7.5 kg and <15 kg 4 mg/kg s.c.
4.4 mg/kg s.c., maintenance dose 2 to <4 years and ≥7.5 kg 4 mg/kg s.c.
2.2 mg/kg s.c. TRAPS, HIDS/MKD, FMF:
every 4 weeks
>40 kg 150 mg s.c.
≥7.5kg and ≤40 kg 2 mg/kg s.c.
Still’s disease: every 4 weeks
Downloaded by [University of New England] at 03:08 08 November 2017

≥7.5 kg 4 mg/kg s.c.


Gout: 150 mg s.c. during attack
Studies outside licensed FMF [39] FMF [50] Schnitzler’s syndrome [55]
indications Still’s disease [40] SJIA [51] Behçet’s disease [56]
Behçet’s disease [41] Schnitzler’s syndrome [52] Polymyalgia rheumatica [57]
Kawasaki disease [42] Gout [53] Diabetes mellitus type 1 [46]
Gout [43], CPPD [44] Atherosclerosis and chronic kidney Diabetes mellitus type 2 [58]
Diabetes mellitus type 1 [45,46] disease [54] Atherosclerosis [59]
Diabetes mellitus type 2 [47] COPD [60]
Myocardial infarction [48]
Pericarditis [49]
CAPS, cryopyrin-associated periodic syndrome; CINCA, chronic infantile neurological cutaneous and articular syndrome; COPD, chronic obstructive pulmonary disease;
CPPD, calcium pyrophosphate deposition disease; EMA, European Medicines Agency; FCAS, familial cold autoinflammatory syndrome; FDA, Food and Drug
Administration; FMF, familial Mediterranean fever; HIDS, hyper-IgD-syndrome; MKD, mevalonate kinase deficiency; MWS, Muckle Wells-syndrome; RA, rheumatoid
arthritis; s.c., subcutaneous; SJIA, systemic juvenile idiopathic arthritis; TRAPS, TNF receptor-associated periodic syndrome.

after injection [29]. Anakinra ameliorated constitutional (fever, this tremendous improvement of clinical symptoms, serological
fatigue), musculoskeletal, und cutaneous symptoms markers of inflammation (CRP, ESR, SAA) decreased within hours
[36,61,65,71–73]. The neurological manifestations of the severe after the administration of anakinra [72,75].
phenotypes also showed improvement with continued adminis- The most common adverse event of anakinra is injection
tration. In a number of trials, chronic meningitis, and intracranial site reaction [36,61,71–73,75,79]. The majority of injection-site
pressure improved and papilledema stabilized [36] or regressed events (65%) occurred within the first month, whereas
[73,75]. In line with these findings, headache ameliorated or even approximately three-quarter of the cases (73%) resolved
resolved [36,65,73,75] and visual acuity improved in the majority within one week [79]. An increased rate of infections has
of cases [75]. From a pharmacodynamic perspective, anakinra has also been observed, particularly of the upper respiratory
been found to dose dependently cross the blood–brain barrier in tract [72,75,79]. An elevated rate of severe and invasive infec-
nonhuman primates [77], hence a direct anti-inflammatory action tions with Group A Streptococcus was associated with anakinra
in the brain is conceivable. Some neurological symptoms may, treatment, leading to necrotizing fasciitis, sepsis or tissue
however, also benefit from a resolution of systemic inflammation. abscess [80]. To a lesser extend, headache, arthralgia [69,79],
No improvement of cognitive function was noted, probably due and gastrointestinal symptoms [36,69] have been observed,
to irreversible organ damage seen in magnetic resonance imaging however these adverse events may also reflect uncontrolled
(MRI) [36,73,75]. Regarding long-term CAPS complications, the activity of the underlying CAPS. In a long-term cohort study,
progression of perceptive hearing loss is halted in the majority the highest number of adverse events was reported in the first
of patients. Both, improvement [65,71,73] and aggravation [71,75] year of treatment with a gradual decrease over time [79].
of auditory symptoms have been reported. Amyloidosis improved Anakinra passes the placenta and enters breast milk, this is
in a number of patients [71,73] or even resolved [61]. With regard however also observed physiologically with respect to the nat-
to the arthropathy, progression was not unusual [73,75], although ural occurring IL-1RA [81]. A small observational study analyzed
one case of positive development was reported [78]. the outcome of women with CAPS during pregnancy treated
The positive clinical effects were not only maintained on a with anakinra [82]. No complications or negative consequences
short-term, but also after several months of continuous applica- on maternal or fetal health were reported, except for one case of
tion with the vast majority of patients having sustained disease renal agenesis [82,83]. Although no worsening of CAPS-symp-
control [29,72]. Furthermore, continuous treatment with ana- toms was observed during pregnancy, a dose increase is recom-
kinra improved the quality of life globally and with regard to its mended if the weight gain exceeds 10kg [82,83]. Another
physical and psychological components [36,75]. In addition to observational report [83] described no development
858 E. C. LANDMANN AND U. A. WALKER

peculiarities in exposed infants, except for the occurrence of one patients treated with rilonacept experienced an infection,
renal agenesis as well, but the long-term safety of drug exposure compared to 17% in the placebo group [89]. In another
to the fetus and infant needs to be investigated further. patient population, however, no significant increase in infec-
tions was reported [88]. Moreover, weight gain [92], generally
decreasing over time [89] and headache [89] was noted.
4.2. Rilonacept (Arcalyst ) ® Importantly, the occurrence of adverse reactions was indepen-
Rilonacept has been approved by the FDA in 2008 for adults and dent of the dose administered [86]. Based on animal experi-
children with FCAS and MWS above the age of 12 years [84]. In ments, rilonacept may cause an increase of stillbirths and
2009, the EMA has also granted approval for the treatment of osseous abnormalities but the fertility was not altered [84].
CAPS. Due to limited use in the European market, the approval of As there are no controlled data in human pregnancy, rilona-
Rilonacept has however been withdrawn in 2012 [85]. cept should only be given during pregnancy when benefits
Rilonacept is an engineered construct of the extracellular outweigh risks.
IL-1 receptor domains (IL-1 receptor 1 (IL-1R1) and IL-receptor
accessory protein (IL-1RAP)) fused to a humanized IgG1-Fc
portion. Rilonacept captures IL-1ß, IL-1α, and IL-1RA as a
4.3. Canakinumab (Ilaris ) ®
decoy receptor. Compared with the natural IL-1 receptor, the Canakinumab was granted approval by the FDA in 2009 in adults
affinity of rilonacept to IL-1ß is 100-fold higher [86,87]. and children above 4 years of age with FCAS and MWS [93], and by
Downloaded by [University of New England] at 03:08 08 November 2017

Rilonacept is administered weekly, owing to its approximate the EMA in 2009 for all CAPS phenotypes in children above 2 years
terminal half-life of 7 days. In adults, the initial subcutaneous of age [94]. Canakinumab is a recombinant fully humanized mono-
loading dose is 320 mg, followed by weekly injections of 160 mg clonal antibody of the IgG1 isotype, and neutralizes serum IL-1ß
[84]. In pediatric patients below 17 years of age, the loading dose with high selectivity and affinity [95]. Thus, canakinumab blocks
is 4.4 mg/kg (to a maximum of 320 mg per injection) with a the interaction of IL-1ß with its receptors and inhibits its down-
maintenance dosage of 2.2 mg/kg (to a maximum of 160 mg per stream signals [95]. Canakinumab has a mean terminal half-life of
week) [84]. In patients with renal impairment, dose adjustment is 26 days [23], permitting subcutaneous injections every 8 weeks.
not needed, however no controlled study has been performed The recommended dose is 150 mg in patients above 40 kg of
[86,88]. Approximately one-third of the patients developed anti- body weight, 2 mg/kg in patients with a body weight between
drug antibodies but no influence on the pharmacokinetics or 15 kg and 40 kg, and 4 mg/kg in patients with a weight of below
safety of rilonacept was observed [89,90] 15 kg and above 7.5 kg. In infants between 2 and 4 years of age,
After first trials with rilonacept had shown a positive effect the recommended dose is 4 mg/kg [72,93,94]. In renal or hepatic
in patients with rheumatoid arthritis [91], a benefit was also impairment, no dose adjustment is needed, however, the experi-
noted in FCAS [89,92] and MWS [89]. In comparison to pla- ence in these populations is limited [93,94,96]. No immunogenicity
cebo, a clinical and serological response was described within is reported in most trials [96–99].
24 h after drug administration [89,92]. Disease activity and key The efficacy of canakinumab was first described in rheuma-
symptoms of CAPS (fever, rash, eye redness/pain, fatigue, toid arthritis [100]. In CAPS, clinical and serological improve-
arthralgia) decreased significantly after the initiation of treat- ment was noted as early as 24 h after drug administration
ment [89]. If remission was not achieved, a successful result [97,98]. After one week about 89% of patients with CAPS [97]
was noted after a dose escalation to the maximum of 320 mg were in complete remission. The short-term response rates
per week [92]. varied slightly [97–99], possibly due to differences in the
Rilonacept has also sustained benefit on the long term demographic characteristics and CAPS phenotypes included.
[90,92]. After 18 months of consecutive treatment, rilonacept The effects were sustained and complete response rates aug-
improved key symptoms by half in the vast majority (87%) of mented to even 97% after 48 weeks of treatment in one trial
patients and by 70% in three-quarters of patients [90]. The [97], although lower rates (72%) were also reported [101].
patient’s as well as the physician’s assessment of disease Canakinumab was also shown to prevent disease flares in a
activity and daily impairment improved substantially [89,90]. placebo-controlled trial [99], with 80% of patients in the pla-
Additionally, rilonacept efficiently prevented disease flares in cebo group experiencing recurrence of inflammation after
comparison to placebo [89]. In line with clinical improvement, drug discontinuation [97]. Apart from the clinical responses,
rilonacept also induced a significant decline of serological a prompt and long-lasting normalization of acute-phase reac-
markers of CAPS activity (particularly CRP, and SAA) [89,92]. tants (CRP and SAA) was shown with continuous administra-
To the author’s knowledge, there are no published data with tion of canakinumab [98,99].
regard to the brain penetration of rilonacept. Although rilona- With regard to the more severe clinical CAPS presentations,
cept is unlikely to directly cross the blood–brain barrier due to neurological complications also improved [102], in one trial even
its high-molecular weight (251 kDa) [88], neurological symp- in virtually 50% of patients over a period of 2 years [99]; cognitive
toms do improve. During flares of meningitis, leaky meninges impairment however remained [102]. Similarly, visual acuity,
may permit drug entry into the brain. Control of systemic sensorineural hearing loss, and renal amyloidosis improved or
inflammation may also contribute to the resolution of cerebral did not progress [99,102,103]. It is however important to note
inflammation [88]. that pediatric patients as well as those with the more severe
The most common adverse events of rilonacept consist of CAPS phenotypes, particularly with CINCA, may require higher
injection-site reactions [84,89] and upper respiratory tract doses or shorter dosing intervals in order to achieve complete
infections [84,89,92]. In one trial, approximately half (48%) of clinical and serological remission [99,101,102,104]. The necessity
EXPERT REVIEW OF CLINICAL PHARMACOLOGY 859

of dose adjustments in some patients is also underlined by or type of gene defect [36,65,75,99]. Although one report
considerably lower complete response rates in patients with noted an insufficient response in patients without clear
more severe CAPS phenotypes (14%) compared to mild and NLRP3 mutations [62]; in the beta-confident registry CAPS
moderate subtypes (76% and 85%, respectively) [101], perhaps activity was predominantly absent in patients without detect-
owing to the negligible penetrance of the monoclonal antibody able NLRP3 mutations receiving canakinumab [105].
into brain tissue. Therefore, a more aggressive treatment regi- The improved outcome of patients with CAPS treated with
men is proposed in these severe cases with 4 mg/kg every IL-1ß blockers emphasizes the pivotal role of IL-1ß in these
4 weeks [104]. This regimen is also recommended for canakinu- syndromes. IL-1ß blockade is not a cure of CAPS and therefore
mab in the treatment of systemic juvenile idiopathic arthritis [93]. disease flares occur after drug discontinuation [23,98]. Up to
Similar to other immunosuppressive medications, the most date, long-term observations are sparse, however, it is
common adverse event of canakinumab is infections [101], assumed that early and life-long treatment is essential for a
particularly of the upper respiratory tract, although gastroen- good prognosis, also because persistent inflammation leads to
teritis and urinary tract infections were noted as well [97,99]. In progressive and irreversible organ damage.
the beta-confident registry, a registry that prospectively fol- During infections withdrawal of IL-1 blockers should be con-
lows 288 patients treated with canakinumab in 13 countries, sidered carefully due to the risk of CAPS flares triggered by the
the incidence rate of serious infections was 4.1 per 100 patient infectious agent. Unlike the situation with biologics in autoim-
years [105]. A few cases of opportunistic infections (for mune diseases, it is therefore suggested to rather increase the
Downloaded by [University of New England] at 03:08 08 November 2017

instance aspergillosis, herpes zoster, and atypical mycobac- dosage of the IL-1 inhibitor during infection and surgery [75,79].
teria) were observed [94]. Injection site reactions [98,99,101],
as well as headache and vertigo [93,94,98,99] were also
reported. Remarkably, no additional adverse events appeared 5. Conclusion
with increased dosage [101].
The treatment of CAPS has evolved tremendously. Three
Monoclonal antibodies are transferred via the placenta in
approved IL-1ß targeting drugs induce a prompt and sus-
linear manner, and therefore a fetal exposure in the 2nd and
tained clinical and serological response in CAPS of varying
3rd trimester is likely. A delay in fetal ossification was observed
phenotype. The outcome of CAPS has improved significantly,
in animal models [93]. In pregnancy, limited data are available,
also with regard to quality of life. Generally, the agents are
abnormalities were not reported [83]. Women of reproductive
safe and well tolerated by the patients.
age are advised to use contraceptives for up to 3 months after
the last injection of canakinumab [94]. It is not established if
canakinumab is present in breast milk, although IgG is known
6. Expert commentary
to appear physiologically in breast milk [93].
Although the treatment of CAPS with drugs targeting IL-1ß is
a success story, long-term prospective studies are important to
4.4. General remarks with regard to the approved IL-1β
ensure efficacy and safety over decades. Future clinical trials
targeting agents
should also address the association between CAPS pheno-
Up to date, no head-to-head trial has directly compared the types, the vast number of genetic variants and response to
three anti-IL-1 drugs. A cohort study however compared the treatment. This may allow a more targeted dose regimen.
efficacy of anakinra with that of canakinumab [72]. Both More work is also needed to better delineate drug safety in
agents showed good efficacy in clinical and serological disease pregnancy, neonates and infants, and to better characterize
control. A greater sustained remission rate was noted in the evolution of late complications with early treatment.
patients treated with canakinumab (93% versus 75% in Clearly a multinational platform is necessary to generate
patients treated with anakinra), however, variations in disease robust data in a disease of such low prevalence.
severity were present among the two cohorts prior to treat- At present, there is no clear unmet need for new IL-1
ment [72]. In patients who experienced secondary treatment inhibitors in the particular group of patients with CAPS. The
failure with anakinra, despite anakinra dose escalation and only new IL-1β blocker developed for CAPS is HL2351 [106]. It
excellent treatment adherence, switching to canakinumab is a recombinant IL-1 receptor antagonist fused to a hybrid Fc
induced a good response [72,98]. fragment consisting of IgD and IgG4. The hybrid fusion tech-
A few clinical and serological changes observed in treated nology enables a prolonged half-life with a weekly subcuta-
patients, apply to all agents that target IL-1ß, owing to the neous administration. A phase II study in individuals with
resolution of the chronic inflammatory state. Various authors CAPS was however terminated due to recruitment difficulties
reported a considerable growth and weight gain in treated [107]. To our knowledge, no results regarding drug safety or
patients, particularly in children, likely as a consequence of the efficacy are available.
control of the catabolic state of inflammation [73,75,98]. It is particularly important to better understand the role of
Similarly, hematological changes were noted, for instance a IL-1 blockers in promoting bacterial infections, in particular
rise in hemoglobin, red blood cells, as well as a decrease in pneumococci and group A streptococci [80]. Interestingly,
neutrophil granulocytes and platelets [89,90,93,94,98], and a pneumococcal vaccinations were recently found to provoke
change in lipid profiles [84,92]. substantial local and systemic inflammation in CAPS, even in
Also noteworthy is the good clinical and serological those patients who were treated with canakinumab [108,109].
response to IL-1ß blocking drugs, regardless of the presence The role of IL-1 blockers in preventing such inflammatory
860 E. C. LANDMANN AND U. A. WALKER

episodes triggered by bacterial antigens, and their require- principle of IL-1 and NLRP3 inflammasome blockade will likely
ment during surgery need to be better characterized. be developed for numerous other autoinflammatory condi-
The activated inflammasome is also known to cleave proIL- tions. In CAPS future research will focus on a better under-
18 cytokine to the active IL-18 [2]. In vitro studies have demon- standing of genotype phenotype correlations, what triggers
strated an enhanced IL-18 secretion from leukocytes from CAPS flares, and to monitor long term drug efficacy and safety.
patients with CAPS [19,110]. Although it was hypothesized
that IL-18 may drive early disease processes and skin pathol-
ogy in CAPS [110], the extraordinarily rapid and sustained Key issues
resolution of most clinical activity by IL-1ß blockers argues ● CAPS are a group of rare monogenic autoinflammatory
against such prominent role of IL-18 and rather underlines disorders with a constitutively activated NLRP3-
the predominant role of IL-1ß. The role of IL-18 in CAPS inflammasome.
needs to be elucidated in more detail, perhaps with the help ● There are three approved agents that target IL-1β in CAPS:
of antibodies that target IL-18 [111]. anakinra, a recombinant IL-1 receptor antagonist, the IL-1 trap
rilonacept and canakinumab, a monoclonal anti-IL-1ß antibody.
7. Five-year view ● IL-1 blockers induce prompt resolution of clinical and bio-
chemical CAPS activity but may require dose escalation,
The IL-1 blockers currently approved for CAPS are also studied particularly in children and in severe phenotypes.
Downloaded by [University of New England] at 03:08 08 November 2017

in numerous other autoimmune, metabolic and inflammatory ● IL-1β targeting drugs are mostly well-tolerated in indivi-
disorders (Table 1). For instance, atherosclerosis is now recog- duals with CAPS. The rate of severe infections is about 4.1
nized as a disorder with a strong autoinflammatory compo- per 100 person years.
nent [112]. For this reason a number of preliminary trials ● Early and life-long treatment is mostly required to maintain
investigate anti-IL-1β agents for the primary and secondary the goal of sustained remission.
prophylaxis of atherosclerosis [59,113]. The potential utility of
IL-1β blockers in a wide range of conditions with an autoin-
flammatory component justifies the great efforts made to Funding
develop novel agents targeting IL-1ß and its intracellular This paper was not funded.
synthesis.
Gevokizumab (also referred as Xoma 052) is an engineered
humanized allosteric ligand-modifying antibody of the IgG2 iso- Declaration of interest
type [114] which binds IL-1ß with a high affinity [115] and speci-
UA Walker has served for Novartis in the steering committee of the Beta-
ficity [114,115]. Gevokizumab modulates the affinity kinetics of IL- confident (canakinumab) registry. The authors have no other relevant
1ß to its receptor IL-1R1 [114–116] without suppressing negative affiliations or financial involvement with any organization or entity with
regulators of IL-1ß in the intent to restore the physiological pro- a financial interest in or financial conflict with the subject matter or
and anti-inflammatory balance [116]. Gevokizumab has a half-life materials discussed in the manuscript apart from those disclosed.
of approximately 23 days [117]. Gevokizumab had promising
results in preliminary studies of Behçet’s disease [118], psoriasis
[119], and diabetes mellitus type 2 [120]. Although gevokizumab References
failed to meet the primary end point in some phase III studies Papers of special note have been highlighted as either of interest (•) or of
[118], its clinical development will likely continue. considerable interest (••) to readers.
Other antibodies that neutralize IL-1ß are not yet in clinical 1. Dinarello CA. Interleukin-1 in the pathogenesis and treatment of
use but will likely enter further development. One example is inflammatory diseases. Blood. 2011;117(14):3720–3732.
2. Dinarello CA. Immunological and inflammatory functions of the
P2D7KK, a high-affinity anti-IL-1ß-antibody with a 11-fold interleukin-1 family. Annu Rev Immunol. 2009;27:519–550.
higher potency to neutralize IL-1ß than canakinumab, but 3. Martinon F, Mayor A, Tschopp J. The inflammasomes: guardians of
with similar affinity and efficacy to block IL-1R1 [121]. the body. Annu Rev Immunol. 2009;27:229–265.
Direct NLRP3 inflammasome inhibitors are interesting, as 4. Lamkanfi M, Dixit VM. Inflammasomes and their roles in health and
these agents do not only block the formation of IL-1, IL-18, disease. Annu Rev Cell Dev Biol. 2012;28:137–161.
5. Levy R, Gérard L, Kuemmerle-Deschner J, et al. Phenotypic and
and other caspase-1-dependent molecules, but also target genotypic characteristics of cryopyrin-associated periodic syn-
other mechanisms of potential pathophysiological relevance, drome: a series of 136 patients from the Eurofever Registry. Ann
such as pyroptosis, a particular mechanism of cell death Rheum Dis. 2015;74(11):2043–2049.
mediated by inflammasome activation. One of the endogenous 6. Aksentijevich I, D Putnam C, Remmers EF, et al. The clinical con-
tinuum of cryopyrinopathies: novel CIAS1 mutations in North
inhibitors of the NLRP3 inflammasome is pyrin domain-only
American patients and a new cryopyrin model. Arthritis Rheum.
protein 1 (POP1). POP1 prevents inflammasome assembly, thus 2007;56(4):1273–1285.
abating caspase-1 activation and release of IL-1ß and IL-18 [122]. 7. Cuisset L, Jeru I, Dumont B, et al. Mutations in the autoinflamma-
Other small molecules such as MCC950, a NLRP3 inhibitor that tory cryopyrin-associated periodic syndrome gene: epidemiological
interferes with inflammasome assembly [123], and VX-765, an study and lessons from eight years of genetic analysis in France.
Ann Rheum Dis. 2011;70(3):495–499.
orally absorbed prodrug of a competitive caspase-1 inhibitor
8. Aksentijevich I, Nowak M, Mallah M, et al. De novo CIAS1 muta-
[124,125], are currently in experimental use only. tions, cytokine activation, and evidence for genetic heterogeneity
In summary, the therapeutic armamentarium for CAPS can- in patients with neonatal-onset multisystem inflammatory disease
not be expected to expand within the next few years but the (NOMID): a new member of the expanding family of pyrin-
EXPERT REVIEW OF CLINICAL PHARMACOLOGY 861

associated autoinflammatory diseases. Arthritis Rheum. 2002;46 28. Hoffman HM, Rosengren S, Boyle DL, et al. Prevention of cold-
(12):3340–3348. associated acute inflammation in familial cold autoinflammatory
9. Feldmann J, Prieur A-M, Quartier P, et al. Chronic infantile neuro- syndrome by interleukin-1 receptor antagonist. Lancet. 2004;364
logical cutaneous and articular syndrome is caused by mutations in (9447):1779–1785.
CIAS1, a gene highly expressed in polymorphonuclear cells and 29. Hawkins PN, Lachmann HJ, Aganna E, et al. Spectrum of clinical
chondrocytes. Am J Hum Genet. 2002;71(1):198–203. features in Muckle-Wells syndrome and response to anakinra.
10. Hoffman HM, Mueller JL, Broide DH, et al. Mutation of a new gene Arthritis Rheum. 2004;50(2):607–612.
encoding a putative pyrin-like protein causes familial cold autoin- 30. Aganna E, Martinon F, Hawkins PN, et al. Association of mutations
flammatory syndrome and Muckle-Wells syndrome. Nat Genet. in the NALP3/CIAS1/PYPAF1 gene with a broad phenotype includ-
2001;29(3):301–305. ing recurrent fever, cold sensitivity, sensorineural deafness, and AA
• First description of the gene mutated in patients with FCAS amyloidosis. Arthritis Rheum. 2002;46(9):2445–2452.
and MWS. 31. Lachmann HJ, Goodman HJB, Gilbertson JA, et al. Natural history
11. Touitou I. The registry of Hereditary auto-inflammatory Disorders and outcome in systemic AA amyloidosis. N Engl J Med. 2007;356
Mutations – infevers. 2017 [cited 2017 Mar 12]. Available from: (23):2361–2371.
http://fmf.igh.cnrs.fr/ISSAID/infevers/ 32. Hill SC, Namde M, Dwyer A, et al. Arthropathy of neonatal onset
12. Tanaka N, Izawa K, Saito MK, et al. High incidence of NLRP3 somatic multisystem inflammatory disease (NOMID/CINCA). Pediatr Radiol.
mosaicism in patients with chronic infantile neurologic, cutaneous, 2007;37(2):145–152.
articular syndrome: results of an International Multicenter 33. Prieur AM. A recently recognised chronic inflammatory disease of
Collaborative Study. Arthritis Rheum. 2011;63(11):3625–3632. early onset characterised by the triad of rash, central nervous
13. Saito M, Nishikomori R, Kambe N, et al. Disease-associated CIAS1 system involvement and arthropathy. Clin Exp Rheumatol.
Downloaded by [University of New England] at 03:08 08 November 2017

mutations induce monocyte death, revealing low-level mosaicism 2001;19(1):103–106.


in mutation-negative cryopyrin-associated periodic syndrome 34. Dollfus H, Häfner R, Hofmann HM, et al. Chronic infantile neurolo-
patients. Blood. 2008;111(4):2132–2141. gical cutaneous and articular/neonatal onset multisystem inflam-
14. Nakagawa K, Gonzalez-Roca E, Souto A, et al. Somatic NLRP3 matory disease syndrome: ocular manifestations in a recently
mosaicism in Muckle-Wells syndrome. A genetic mechanism shared recognized chronic inflammatory disease of childhood. Arch
by different phenotypes of cryopyrin-associated periodic syn- Ophthalmol. 2000;118(10):1386–1392.
dromes. Ann Rheum Dis. 2015;74(3):603–610. 35. Hoffman HM, Wolfe F, Belomestnov P, et al. Cryopyrin-associated
15. Zhou Q, Aksentijevich I, Wood GM, et al. Brief report: cryopyrin- periodic syndromes: development of a patient-reported outcomes
associated periodic syndrome caused by a myeloid-restricted instrument to assess the pattern and severity of clinical disease
somatic NLRP3 mutation. Arthritis Rheum. 2015;67(9):2482–2486. activity. Curr Med Res Opin. 2008;24(9):2531–2543.
16. Kuemmerle-Deschner JB, Ozen S, Tyrrell PN, et al. Diagnostic cri- 36. Lepore L, Paloni G, Caorsi R, et al. Follow-up and quality of life of
teria for cryopyrin-associated periodic syndrome (CAPS). Ann patients with cryopyrin-associated periodic syndromes treated with
Rheum Dis. 2017;76:942–947. Anakinra. J Pediatr. 2010;157(2):310–315.e1.
17. Rosengren S, Mueller JL, Anderson JP, et al. Monocytes from famil- 37. Koné-Paut I, Lachmann HJ, Kuemmerle-Deschner JB, et al.
ial cold autoinflammatory syndrome patients are activated by mild Sustained remission of symptoms and improved health-related
hypothermia. J Allergy Clin Immunol. 2007;119(4):991–996. quality of life in patients with cryopyrin-associated periodic syn-
18. Agostini L, Martinon F, Burns K, et al. NALP3 forms an IL-1beta- drome treated with canakinumab: results of a double-blind pla-
processing inflammasome with increased activity in Muckle-Wells cebo-controlled randomized withdrawal study. Arthritis Res Ther.
autoinflammatory disorder. Immunity. 2004;20(3):319–325. 2011;13(6):R202.
19. Janssen R, Verhard E, Lankester A, et al. Enhanced interleukin-1beta 38. Ter Haar NM, Oswald M, Jeyaratnam J, et al. Recommendations for
and interleukin-18 release in a patient with chronic infantile neu- the management of autoinflammatory diseases. Ann Rheum Dis.
rologic, cutaneous, articular syndrome. Arthritis Rheum. 2004;50 2015;74(9):1636–1644.
(10):3329–3333. 39. Ben-Zvi I, Kukuy O, Giat E, et al. Anakinra for colchicine resistant
20. Haverkamp MH, Van de Vosse E, Goldbach-Mansky R, et al. familial Mediterranean fever – A randomized, double blind, pla-
Impaired cytokine responses in patients with cryopyrin-associated cebo-controlled trial. Arthritis Rheumatol. 2016;69(4):854–862.
periodic syndrome (CAPS). Clin Exp Immunol. 2014;177(3):720–731. 40. Laskari K, Tzioufas AG, Moutsopoulos HM. Efficacy and long-term
21. Gattorno M, Tassi S, Carta S, et al. Pattern of interleukin-1beta follow-up of IL-1R inhibitor anakinra in adults with Still’s disease: a
secretion in response to lipopolysaccharide and ATP before and case-series study. Arthritis Res Ther. 2011;13(3):R91.
after interleukin-1 blockade in patients with CIAS1 mutations. 41. Cantarini L, Vitale A, Scalini P, et al. Anakinra treatment in drug-
Arthritis Rheum. 2007;56(9):3138–3148. resistant Behcet’s disease: a case series. Clin Rheumatol. 2015;34
22. Tassi S, Carta S, Delfino L, et al. Altered redox state of monocytes (7):1293–1301.
from cryopyrin-associated periodic syndromes causes accelerated 42. Tremoulet AH, Jain S, Kim S, et al. Rationale and study design for a
IL-1beta secretion. Proc Natl Acad Sci U S A. 2010;107(21):9789– phase I/IIa trial of anakinra in children with Kawasaki disease and
9794. early coronary artery abnormalities (the ANAKID trial). Contemp
23. Lachmann HJ, Lowe P, Felix SD, et al. In vivo regulation of inter- Clin Trials. 2016;48:70–75.
leukin 1beta in patients with cryopyrin-associated periodic syn- 43. Ottaviani S, Moltó A, Ea H-K, et al. Efficacy of anakinra in gouty
dromes. J Exp Med. 2009;206(5):1029–1036. arthritis: a retrospective study of 40 cases. Arthritis Res Ther.
24. Ter Haar N, Lachmann H, Özen S, et al. Treatment of autoinflam- 2013;15(5):R123.
matory diseases: results from the Eurofever Registry and a literature 44. Ottaviani S, Brunier L, Sibilia J, et al. Efficacy of anakinra in calcium
review. Ann Rheum Dis. 2013;72(5):678–685. pyrophosphate crystal-induced arthritis: a report of 16 cases and
25. Neven B, Prieur A-M, Quartier Dit Maire P. Cryopyrinopathies: review of the literature. Joint Bone Spine. 2013;80(2):178–182.
update on pathogenesis and treatment. Nat Clin Pract 45. Van Asseldonk EJ, Van Poppel PCM, Ballak DB, et al. One week
Rheumatol. 2008;4(9):481–489. treatment with the IL-1 receptor antagonist anakinra leads to a
26. Houx L, Hachulla E, Kone-Paut I, et al. Musculoskeletal symptoms in sustained improvement in insulin sensitivity in insulin resistant
patients with cryopyrin-associated periodic syndromes: a large patients with type 1 diabetes mellitus. Clin Immunol. 2015;160
database study. Arthritis Rheum. 2015;67(11):3027–3036. (2):155–162.
27. Hoffman HM, Wanderer AA, Broide DH. Familial cold autoinflam- 46. Moran A, Bundy B, Becker DJ, et al. Interleukin-1 antagonism in
matory syndrome: phenotype and genotype of an autosomal type 1 diabetes of recent onset: two multicentre, randomised,
dominant periodic fever. J Allergy Clin Immunol. 2001;108(4):615– double-blind, placebo-controlled trials. Lancet. 2013;381
620. (9881):1905–1915.
862 E. C. LANDMANN AND U. A. WALKER

47. Larsen CM, Faulenbach M, Vaag A, et al. Interleukin-1-receptor 66. Bachove I, Chang C. Anakinra and related drugs targeting inter-
antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356 leukin-1 in the treatment of cryopyrin-associated periodic syn-
(15):1517–1526. dromes. Open Access Rheumatol. 2014;6:15–25.
48. Abbate A, Kontos MC, Abouzaki NA, et al. Comparative safety of 67. Bresnihan B, Alvaro-Gracia JM, Cobby M, et al. Treatment of rheu-
interleukin-1 blockade with anakinra in patients with ST-segment matoid arthritis with recombinant human interleukin-1 receptor
elevation acute myocardial infarction (from the VCU-ART and VCU- antagonist. Arthritis Rheum. 1998;41(12):2196–2204.
ART2 pilot studies). Am J Cardiol. 2015;115(3):288–292. 68. Hawkins PN, Lachmann HJ, McDermott MF. Interleukin-1-receptor
49. Lazaros G, Imazio M, Brucato A, et al. Anakinra: an emerging option for antagonist in the Muckle-Wells syndrome. N Engl J Med. 2003;348
refractory idiopathic recurrent pericarditis: a systematic review of (25):2583–2584.
published evidence. J Cardiovasc Med (Hagerstown). 2016;17 •• Observational study showing for the first time a prompt and
(4):256–262. effective response to anakinra in two patients with MWS.
50. Hashkes PJ, Spalding SJ, Hajj-Ali R, et al. The effect of rilonacept 69. FDA. Kineret. Product information. [cited 2017 Mar 14]. Available
versus placebo on health-related quality of life in patients with from: www.accessdata.fda.gov/drugsatfda_docs/label/2012/
poorly controlled familial Mediterranean fever. Biomed Res Int. 103950s5136lbl.pdf
2014;2014:1–8. 70. EMA. Kineret. Product information. [cited 2017 Mar 15]. Available
51. Ilowite NT, Prather K, Lokhnygina Y, et al. Randomized, double- from: http://www.ema.europa.eu/ema/index.jsp?curl=pages/medi
blind, placebo-controlled trial of the efficacy and safety of rilona- cines/human/medicines/000363/human_med_000874.jsp&mid=
cept in the treatment of systemic juvenile idiopathic arthritis. WC0b01ac058001d124
Arthritis Rheum. 2014;66(9):2570–2579. 71. Kuemmerle-Deschner JB, Tyrrell PN, Koetter I, et al. Efficacy and
52. Krause K, Weller K, Stefaniak R, et al. Efficacy and safety of the safety of anakinra therapy in pediatric and adult patients with the
Downloaded by [University of New England] at 03:08 08 November 2017

interleukin-1 antagonist rilonacept in Schnitzler syndrome: an autoinflammatory Muckle-Wells syndrome. Arthritis Rheum.
open-label study. Allergy. 2012;67(7):943–950. 2011;63(3):840–849.
53. Sundy JS, Schumacher HR, Kivitz A, et al. Rilonacept for gout flare 72. Kuemmerle-Deschner JB, Wittkowski H, Tyrrell PN, et al. Treatment
prevention in patients receiving uric acid-lowering therapy: results of Muckle-Wells syndrome: analysis of two IL-1-blocking regimens.
of RESURGE, a phase III, international safety study. J Rheumatol. Arthritis Res Ther. 2013;15(3):R64.
2014;41(8):1703–1711. • Observational trial comparing a cohort of patients with CAPS
54. clinicaltrials.gov. Inflammation in chronic kidney disease and cardi- treated with anakinra or canakinumab.
ovascular disease – the role of genetics and interleukin-1 receptor 73. Neven B, Marvillet I, Terrada C, et al. Long-term efficacy of the
antagonist (IL-1ra). 2017 [cited 2017 Mar 24]. Available from: interleukin-1 receptor antagonist anakinra in ten patients with
https://clinicaltrials.gov/ct2/show/NCT00897715 neonatal-onset multisystem inflammatory disease/chronic infantile
55. Krause, K, Tsianakas, A, Wagner, N, et al. Efficacy and safety of neurologic, cutaneous, articular syndrome. Arthritis Rheum.
canakinumab in Schnitzler syndrome: a multicenter randomized 2010;62(1):258–267.
placebo-controlled study. J Allergy Clin Immunol. 2016;139 74. Yang B-B, Baughman S, Sullivan JT. Pharmacokinetics of anakinra in
(4):1311–1320. subjects with different levels of renal function. Clin Pharmacol Ther.
56. Emmi G, Talarico R, Lopalco G, et al. Efficacy and safety profile of 2003;74(1):85–94.
anti-interleukin-1 treatment in Behçet’s disease: a multicenter ret- 75. Sibley CH, Plass N, Snow J, et al. Sustained response and preven-
rospective study. Clin Rheumatol. 2016;35(5):1281–1286. tion of damage progression in patients with neonatal-onset multi-
57. clinicaltrials.gov. A 3-arm proof of concept study of AIN457, system inflammatory disease treated with anakinra: a cohort study
ACZ885 or corticosteroids in patients with polymyalgia rheumatica. to determine three- and five-year outcomes. Arthritis Rheum.
2015 [cited 2017 Mar 24.]. Available from: https://clinicaltrials.gov/ 2012;64(7):2375–2386.
ct2/show/NCT01364389 76. Maksimovic L, Stirnemann J, Caux F, et al. New CIAS1 mutation and
58. Hensen J, Howard CP, Walter V, et al. Impact of interleukin-1β anti- anakinra efficacy in overlapping of Muckle-Wells and familial cold
body (canakinumab) on glycaemic indicators in patients with type 2 autoinflammatory syndromes. Rheumatology (Oxford). 2008;47
diabetes mellitus: results of secondary endpoints from a randomized, (3):309–310.
placebo-controlled trial. Diabetes Metab. 2013;39(6):524–531. 77. Fox E, Jayaprakash N, Pham T-H, et al. The serum and cerebrosp-
59. Ridker PM, Thuren T, Zalewski A, et al. Interleukin-1β inhibition and inal fluid pharmacokinetics of anakinra after intravenous admin-
the prevention of recurrent cardiovascular events: rationale and istration to non-human primates. J Neuroimmunol. 2010;223(1–
design of the Canakinumab Anti-inflammatory Thrombosis 2):138–140.
Outcomes Study (CANTOS). Am Heart J. 2011;162(4):597–605. 78. Miyamae T, Inaba Y, Nishimura G, et al. Effect of anakinra on
60. Rogliani P, Calzetta L, Ora J, et al. Canakinumab for the treatment arthropathy in CINCA/NOMID syndrome. Pediatr Rheumatol
of chronic obstructive pulmonary disease. Pulm Pharmacol Ther. Online J. 2010;8:9.
2015;31:15–27. 79. Kullenberg T, Löfqvist M, Leinonen M, et al. Long-term safety
61. Leslie KS, Lachmann HJ, Bruning E, et al. Phenotype, genotype, and profile of anakinra in patients with severe cryopyrin-associated
sustained response to anakinra in 22 patients with autoinflamma- periodic syndromes. Rheumatology (Oxford). 2016;55(8):1499–
tory disease associated with CIAS-1/NALP3 mutations. Arch 1506.
Dermatol. 2006;142(12):1591–1597. • Prospective trial showing the efficacy and safety of anakinra
62. Kuemmerle-Deschner JB, Haug I. Canakinumab in patients with treatment in patients with CAPS over 5 years.
cryopyrin-associated periodic syndrome: an update for clinicians. 80. LaRock CN, Todd J, LaRock DL, et al. IL-1beta is an innate immune
Ther Adv Musculoskelet Dis. 2013;5(6):315–329. sensor of microbial proteolysis. Sci Immunol. 2016 Aug;1(2). pii:
63. Cantarini L, Lucherini OM, Frediani B, et al. Bridging the gap eaah3539. doi: 10.1126/sciimmunol.aah3539.
between the clinician and the patient with cryopyrin-associated 81. Buescher ES, Malinowska I. Soluble receptors and cytokine antago-
periodic syndromes. Int J Immunopathol Pharmacol. 2011;24 nists in human milk. Pediatr Res. 1996;40(6):839–844.
(4):827–836. 82. Chang Z, Spong CY, Jesus AA, et al. Anakinra use during pregnancy
64. Snegireva LS, Kostik MM, Caroli F, et al. Failure of tocilizumab in patients with cryopyrin-associated periodic syndromes (CAPS).
treatment in a CINCA patient: clinical and pathogenic implications. Arthritis Rheumatol. 2014;66(11):3227–3232.
Rheumatology (Oxford). 2013;52(9):1731–1732. 83. Youngstein T, Hoffmann P, Lane T, et al. International experience
65. Goldbach-Mansky R, Dailey NJ, Canna SW, et al. Neonatal-onset of pregnancy outcomes in auto-inflammatory syndromes treated
multisystem inflammatory disease responsive to interleukin-1beta with Interleukin-1 inhibitors. Pediatr Rheumatol. 2015;13(Suppl
inhibition. N Engl J Med. 2006;355(6):581–592. 1):O67.
EXPERT REVIEW OF CLINICAL PHARMACOLOGY 863

84. FDA. Rilonacept. Product information. 2012 [cited 2017 Mar 14]. 102. Sibley CH, Chioato A, Felix S, et al. A 24-month open-label study of
Available from: www.accessdata.fda.gov/drugsatfda_docs/label/ canakinumab in neonatal-onset multisystem inflammatory disease.
2008/125249lbl.pdf Ann Rheum Dis. 2015;74(9):1714–1719.
85. EMA. Rilonacept. Product information. 2012 [cited 2017 Mar 14]. 103. Russo RA, Melo-Gomes S, Lachmann HJ, et al. Efficacy and safety of
Available from: http://www.ema.europa.eu/ema/index.jsp?curl= canakinumab therapy in paediatric patients with cryopyrin-asso-
pages/medicines/human/medicines/001047/human_med_000653. ciated periodic syndrome: a single-centre, real-world experience.
jsp&mid=WC0b01ac058001d124 Rheumatology (Oxford). 2014;53(4):665–670.
86. Gillespie J, Mathews R, McDermott MF. Rilonacept in the manage- 104. Caorsi R, Lepore L, Zulian F, et al. The schedule of administration of
ment of cryopyrin-associated periodic syndromes (CAPS). J Inflamm canakinumab in cryopyrin associated periodic syndrome is driven
Res. 2010;3:1–8. by the phenotype severity rather than the age. Arthritis Res Ther.
87. Economides AN, Carpenter LR, Rudge JS, et al. Cytokine traps: 2013;15(1):R33.
multi-component, high-affinity blockers of cytokine action. Nat • Prospective trial indicating an increased response rate with a
Med. 2003;9(1):47–52. treat-to-target approach.
88. Hoffman HM. Rilonacept for the treatment of cryopyrin-associated 105. Kuemmerle-Deschner JB, Hawkins PN, Van der Poll T, et al. Safety
periodic syndromes (CAPS). Expert Opin Biol Ther. 2009;9(4):519–531. and efficacy of canakinumab in patients with CAPS: interim results
89. Hoffman HM, Throne ML, Amar NJ, et al. Efficacy and safety of from the beta-confident registry [abstract]. Arthritis Rheumatol.
rilonacept (interleukin-1 Trap) in patients with cryopyrin-associated 2015;67(suppl 10):2015.
periodic syndromes: results from two sequential placebo-con- 106. Handok.co.kr. New bio drug, HL2351. 2015 [cited 2017 Mar 22].
trolled studies. Arthritis Rheum. 2008;58(8):2443–2452. Available from: https://www.handok.co.kr/english/media/news_
•• Two consecutive placebo-controlled studies showing effective view.asp?seq=764
Downloaded by [University of New England] at 03:08 08 November 2017

and prompt response to rilonacept in patients with FCAS and 107. clinicaltrials.gov. HL2351 CAPS phase II study. 2016 [cited 2017 Mar
MWS. 22]. Available from: https://clinicaltrials.gov/ct2/show/
90. Hoffman HM, Throne ML, Amar NJ, et al. Long-term efficacy and NCT02853084
safety profile of rilonacept in the treatment of cryopryin-associated 108. Walker UA, Hoffman HM, Williams R, et al. Brief report: severe
periodic syndromes: results of a 72-week open-label extension inflammation following vaccination against streptococcus pneu-
study. Clin Ther. 2012;34(10):2091–2103. moniae in patients with cryopyrin-associated periodic syndromes.
91. Dinarello CA. Setting the cytokine trap for autoimmunity. Nat Med. Arthritis Rheumatol. 2016;68(2):516–520.
2003;9(1):20–22. 109. Jaeger VK, Hoffman HM, Van der Poll T, et al. Safety of vaccinations
92. Goldbach-Mansky R, Shroff SD, Wilson M, et al. A pilot study to in patients with cryopyrin-associated periodic syndromes: a pro-
evaluate the safety and efficacy of the long-acting interleukin-1 spective registry based study. Rheumatology. 2017. in press.
inhibitor rilonacept (interleukin-1 Trap) in patients with familial 110. Brydges SD, Broderick L, McGeough MD, et al. Divergence of IL-1,
cold autoinflammatory syndrome. Arthritis Rheum. 2008;58 IL-18, and cell death in NLRP3 inflammasomopathies. J Clin Invest.
(8):2432–2442. 2013;123(11):4695–4705.
93. FDA. Canakinumab product information. 2016 [cited 2017 Mar 14]. 111. McKie EA, Reid JL, Mistry PC, et al. A study to investigate the
Available from: www.accessdata.fda.gov/drugsatfda_docs/label/ efficacy and safety of an anti-interleukin-18 monoclonal antibody
2016/125319s084lbl.pdf in the treatment of type 2 diabetes mellitus. PLoS One. 2016;11(3):
94. EMA. Canakinumab. Product information. 2017 [cited 2017 Mar 14]. e0150018.
Available from: http://www.ema.europa.eu/docs/en_GB/docu 112. Masters SL, Simon A, Aksentijevich I, et al. Horror autoinflammati-
ment_library/EPAR_-_Product_Information/human/001109/ cus: the molecular pathophysiology of autoinflammatory disease
WC500031680.pdf (*). Annu Rev Immunol. 2009;27:621–668.
95. Rondeau J-M, Ramage P, Zurini M, et al. The molecular mode of 113. Ridker PM, Howard CP, Walter V, et al. Effects of interleukin-1β
action and species specificity of canakinumab, a human monoclo- inhibition with canakinumab on hemoglobin A1c, lipids,
nal antibody neutralizing IL-1β. MAbs. 2015;7(6):1151–1160. C-reactive protein, interleukin-6, and fibrinogen: a phase IIb rando-
96. Chakraborty A, Tannenbaum S, Rordorf C, et al. Pharmacokinetic and mized, placebo-controlled trial. Circulation. 2012;126(23):2739–
pharmacodynamic properties of canakinumab, a human anti-interleu- 2748.
kin-1β monoclonal antibody. Clin Pharmacokinet. 2012;51(6):e1–18. 114. Roell MK, Issafras H, Bauer RJ, et al. Kinetic approach to pathway
97. Lachmann HJ, Kone-Paut I, Kuemmerle-Deschner JB, et al. Use of attenuation using XOMA 052, a regulatory therapeutic antibody
canakinumab in the cryopyrin-associated periodic syndrome. N that modulates interleukin-1beta activity. J Biol Chem. 2010;285
Engl J Med. 2009;360(23):2416–2425. (27):20607–20614.
•• Placebo-controlled, randomized trial demonstrating prompt 115. Blech M, Peter D, Fischer P, et al. One target-two different binding
remission to canakinumab in the vast majority of patients modes: structural insights into gevokizumab and canakinumab
with CAPS. interactions to interleukin-1β. J Mol Biol. 2013;425(1):94–111.
98. Kuemmerle-Deschner JB, Ramos E, Blank N, et al. Canakinumab 116. Issafras H, Corbin JA, Goldfine ID, et al. Detailed mechanistic ana-
(ACZ885, a fully human IgG1 anti-IL-1β mAb) induces sustained lysis of gevokizumab, an allosteric anti-IL-1β antibody with differ-
remission in pediatric patients with cryopyrin-associated periodic ential receptor-modulating properties. J Pharmacol Exp Ther.
syndrome (CAPS). Arthritis Res Ther. 2011;13(1):R34. 2014;348(1):202–215.
• Prospective study showing efficacy of canakinumab in patients 117. Owyang AM, Issafras H, Corbin J, et al. XOMA 052, a potent, high-
with CAPS treated over 2 years. affinity monoclonal antibody for the treatment of IL-1β-mediated
99. Kuemmerle-Deschner JB, Hachulla E, Cartwright R, et al. Two-year diseases. MAbs. 2011;3(1):49–60.
results from an open-label, multicentre, phase III study evaluating 118. Hatemi G, Seyahi E, Fresko I, et al. One year in review 2016: Behçet’s
the safety and efficacy of canakinumab in patients with cryopyrin- syndrome. Clin Exp Rheumatol. 2016;34(6 Suppl 102):10–22.
associated periodic syndrome across different severity phenotypes. 119. Mansouri B, Richards L, Menter A. Treatment of two patients with
Ann Rheum Dis. 2011;70(12):2095–2102. generalized pustular psoriasis with the interleukin-1β inhibitor
100. Alten R, Gram H, Joosten LA, et al. The human anti-IL-1β mono- gevokizumab. Br J Dermatol. 2015;173(1):239–241.
clonal antibody ACZ885 is effective in joint inflammation models in 120. Cavelti-Weder C, Babians-Brunner A, Keller C, et al. Effects of gevo-
mice and in a proof-of-concept study in patients with rheumatoid kizumab on glycemia and inflammatory markers in type 2 diabetes.
arthritis. Arthritis Res Ther. 2008;10(3):R67. Diabetes Care. 2012;35(8):1654–1662.
101. Kuemmerle-Deschner JB, Hofer F, Endres T, et al. Real-life effective- 121. Goh AX, Bertin-Maghit S, Ping Yeo S, et al. A novel human anti-
ness of canakinumab in cryopyrin-associated periodic syndrome. interleukin-1β neutralizing monoclonal antibody showing in vivo
Rheumatology (Oxford). 2016;55(4):689–696. efficacy. MAbs. 2014;6(3):765–773.
864 E. C. LANDMANN AND U. A. WALKER

122. De Almeida L, Khare S, Misharin AV, et al. The PYRIN domain-only autoinflammatory syndrome patients. J Immunol. 2005;175
protein POP1 inhibits inflammasome assembly and ameliorates (4):2630–2634.
inflammatory disease. Immunity. 2015;43(2):264–276. 125. Wannamaker W, Davies R, Namchuk M, et al. (S)-1-((S)-2-{[1-(4-
123. Coll RC, Robertson AAB, Chae JJ, et al. A small-molecule inhibitor of amino-3-chloro-phenyl)-methanoyl]-amino}-3,3-dimethyl-butanoy
the NLRP3 inflammasome for the treatment of inflammatory dis- l)-pyrrolidine-2-carboxylic acid ((2R,3S)-2-ethoxy-5-oxo-tetrahydro-
eases. Nat Med. 2015;21(3):248–255. furan-3-yl)-amide (VX-765), an orally available selective interleukin
124. Stack JH, Beaumont K, Larsen PD, et al. IL-converting enzyme/ (IL)-converting enzyme/caspase-1 inhibitor, exhibits potent anti-
caspase-1 inhibitor VX-765 blocks the hypersensitive response inflammatory activities by inhibiting the release of IL-1beta and
to an inflammatory stimulus in monocytes from familial cold IL-18. J Pharmacol Exp Ther. 2007;321(2):509–516.
Downloaded by [University of New England] at 03:08 08 November 2017

You might also like