Skip to main content
Rheumatology (Oxford, England) logoLink to Rheumatology (Oxford, England)
. 2018 Dec 1;58(6):953–962. doi: 10.1093/rheumatology/key339

Clinical significance of Janus Kinase inhibitor selectivity

Ernest H Choy 1,
PMCID: PMC6532440  PMID: 30508136

Abstract

Cytokines are key drivers of inflammation in RA, and anti-cytokine therapy has improved the outcome of RA. Janus Kinases (JAK) are intracellular tyrosine kinases linked to intracellular domains of many cytokine receptors. There are four JAK isoforms: JAK1, JAK2, JAK3 and TYK2. Different cytokine receptor families utilize specific JAK isoforms for signal transduction. Phosphorylation of JAK when cytokine binds to its cognate receptor leads to phosphorylation of other intracellular molecules that eventually leads to gene transcription. Oral JAK inhibitors (JAKi) have been developed as anti-cytokine therapy in RA. Two JAKi, tofacitinib and baricitinib, have been approved recently for the treatment of RA, and many JAKi are currently in development. JAKi inhibit JAK isoforms with different selectivity. This review discusses the efficacy and safety of JAKi in RA, in particular the potential clinical significance of JAKi selectivity.

Keywords: rheumatoid arthritis, Janus Kinase, treatment, targeted synthetic DMARDs, DMARDs


Rheumatology key messages

  • Janus Kinase inhibitor selectively is relative and not absolute.

  • Current approved Janus Kinase inhibitors and those in development significantly inhibit Janus Kinase 1 isoform.

  • Janus Kinase 1 is an effective target in RA, although zoster reactivation is a class effect.

Background

During the 1990s, when mAbs were trialled in patients with RA, many rheumatologists questioned whether i.v. or s.c. injections could ever be a realistic treatment. At the time, researchers argued that the mAbs were the ideal tools to identify specific therapeutic targets for which oral inhibitors can be developed. This vision was realized when in 2009 the Food and Drug Administration approved the first Janus Kinase inhibitor (JAKi), tofacitinib, for the treatment of RA. In 2017, tofacitinib and another JAKi, baricitinib, received approval in Europe. Janus Kinases (JAKs) are intracellular molecules involved in signal transduction of type I and II cytokine receptors [1] including IL-6 receptor, a proven therapeutic target in RA [2]. JAKi are classified by EULAR as targeted synthetic DMARDs. There are four JAK isoforms: JAK1, JAK2, JAK3 and TYK2, which act in pairs to phosphorylate other intracellular proteins. Other JAKi are being developed for the treatment of RA. Current JAKi that are approved and in development for RA inhibit JAK isoforms with different selectivity. Is JAK selectivity clinically meaningful? This review will discuss the efficacy and safety of JAKi in RA as well as the potential clinical significance of JAK isoform selectivity.

Cytokine signalling and biology of the JAK/STAT pathway

Cytokines can be classified by the structure of their receptors. Type I cytokine receptors have certain conserved motifs in their extracellular amino acid domain. These include common γ chain (such as IL-2), gp130 family (such as IL-6), p40 subunit (IL-12 and IL-23) and common β chain cytokine receptors (haemopoietic cytokines such as GM-CSF). Type II cytokine receptors are members of the IL-10 and IFN families. Both Type I and II cytokine receptor families utilize the JAK for signalling transduction. Among these cytokines and cytokine receptors, IL-6 and IL-6 receptor are established therapeutic targets in RA. The role of other JAK signalling cytokines in RA is less well established. JAKs also have essential homeostatic roles, being responsible for signalling of some hormones including prolactin and growth hormone.

JAKs are associated with the membrane proximal regions of cytokine receptors [3]. Conformational changes in cytokine receptors induced by ligand binding lead to phosphorylation of JAKs through the reciprocal interaction of two juxtapositioned JAKs (Fig. 1). Hence, JAK activation requires two JAK isoforms either as homodimers or heterodimers to auto-phosphosphorylate, which allows recruitment and phosphorylation of various signalling molecules including members of the signal transducer and activator of transcription (STAT) family of DNA binding proteins [4]. Phosphorylation of STATs promotes their translocation to the nucleus and gene transcription.

Fig. 1.

Fig. 1

Cytokine signalling via JAK isoforms and their inhibitors

JAK: Janus Kinase; p: phosphate.

Since type I and II cytokine receptor families include a large number of cytokines, growth factors and hormones, JAKs are critical to immune function and homeostasis. Consequently, JAKs are highly conserved and JAK isoforms are non-redundant. JAK isoform knock-out animals have severe clinical phenotypes: JAK1-deficient mice die perinatally [5] while JAK2 knockout animals are embryonic lethal due to defective erythropoiesis [6]. JAK3-deficient mice suffer from severe immunodeficiency syndrome and reduced survival [7]. In humans, mutations in JAK3 cause severe combined immune deficiency syndrome [8, 9]. TYK2-deficient mice are viable but are susceptible to viral infection due to reduced IFN response [10].

Therapeutic principle of JAK inhibition

Evidence from animals and patients with JAK isoform deficiency showed that complete blockade of JAK isoforms is undesirable as it will lead to severe immunodeficiency and abnormal homeostasis. Therefore, the principle of JAK inhibition differs from cytokine inhibition using biologics in that the objective is not to specifically block the JAK pathway completely but to reversibly reduce the activity of one or more JAK isoforms, akin to turning down a thermostat. One potential clinical advantage of such a mode of action is that inhibition can be rapidly reversed with ‘fast-on’ and ‘fast-off’ effects.

JAKi

Two JAKi, tofacitinib and baricitinib, in combination with MTX or as monotherapy, have been approved for the treatment of RA. Both tofacitinib and baricitinib have been examined in large phase III and IV randomized control trials (RCT) in a range of RA patients from conventional synthetic DMARD (csDMARD)-naïve early patients, csDMARD inadequate responders to biologic inadequate responders. The ACR responses in these studies are summarized in Table 1. Tofacitinib is selective for JAK1 for JAK3, while baricitinib is selective for JAK1 and JAK2. In addition, four JAKi are currently in development: the JAK1 selective upadacitinib and filgotinib, the JAK1 and JAK2 inhibitor peficitinib, and the JAK3 selective inhibitor decernotinib.

Table 1.

ACR responses at primary endpoints in tofacitinib and baricitinib RCTs

Controls JAKi Active comparator
ACR20 ACR50 ACR70 ACR20 ACR50 ACR70 ACR20 ACR50 ACR70
MTX inadequate responders
    Oral-STANDARD [11] Tofacitinib (5 mg bid) 26 7 2 61 34 12 56 24 9
    Oral-STANDARD [11] Tofacitinib (10 mg bid) 59 28 15
    RA-BEAM [12] Barcitinib (4 mg qd) 40 17 5 70 45 19 61 35 13
cDMARD inadequate responders
    Oral-SYNC [13] Tofacitinib (5 mg bid) 27 10 2 56 27 8
    Oral-SYNC [13] Tofacitinib (10 mg bid) 65 34 14
    RA-BUILD [14] Barcitinib (2 mg qd) 39 13 3 66 34 18
    RA-BUILD [14] Barcitinib (4 mg qd) 62 33 18
bDMARD inadequate responders
    Oral-STEP [15] Tofacitinib (5 mg bid) 24 8 2 42 27 14
    Oral-STEP [15] Tofacitinib (10 mg bid) 48 28 11
    RA-BEACON [16] Barcitinib (2 mg qd) 27 8 2 49 20 13
    RA-BEACON [16] Barcitinib (4 mg qd) 55 28 11
MTX naïve
    Oral-START [17] Tofacitinib (5 mg bid) 51 (MTX as control) 27 (MTX as control) 12 (MTX as control) 71 47 26
    Oral-START [17] Tofacitinib (10 mg bid) 76 56 38
    RA-BEGIN [18] Barcitinib (4 mg qd)+MTX 62 (MTX as control) 43 (MTX as control) 21 (MTX as control) 78 63 40
    Monotherapy
    Oral-SOLO [19] Tofacitinib (5 mg bid) 27 13 6 60 31 15
    Oral-SOLO [19] Tofacitinib (10 mg bid) 66 37 20
    RA-BEGIN [18] Barcitinib (4 mg qd) 62 (MTX as control) 43 (MTX as control) 21 (MTX as control) 77 60 42

Numbers are percentages. JAKi: Janus Kinase inhibitor; RCT: randomized control trial; bDMARD: biological DMARD.

In MTX inadequate responders, tofacitinib [5 and 10 mg bis in die (bid)] in Oral-STANDARD [11] and baricitinib [4 mg once daily (od)] in RA-BEAM [12] added to MTX showed superior ACR responses when compared with placebo. Tofacitinib showed a similar response to adalimumab while baricitinib in RA-BEAM achieved statistically significant higher ACR20 responses than adalimumab (70 vs 61%). However, the difference was small (<10%) and the sample size was larger in RA-BEAM than in Oral-STANDARD. Similar to MTX inadequate responders, in csDMARD inadequate responders, adding tofacitinib in Oral-SYNC [13] and baricitinib in RA-BUILD [14] achieved higher ACR responses than placebo. In biologic inadequate responders, tofacitinib (5 and 10 mg bid) in Oral-Step [15] and baricitinib (2 and 4 mg od) in RA-BEACON [16] in combination with MTX achieved higher ACR responses than placebo.

Radiographic damage

In ORAL-SCAN [20], radiographic damage was statistically significantly less in patients treated with tofacitinib 10 mg when compared with placebo-treated patients. Tofacitinib 5 mg-treated patients had less radiographic damage than placebo-treated patients but this did not achieve statistical significance. Baricitinib has also been shown to reduce radiographic damage in RA-BUILD [14], RA BEAM [12] and RA-BEGIN [18]. In RA-BUILD, baricitinib, both 2 and 4 mg in combination with MTX statistically significantly reduced radiographic progression when compared with placebo. In RA-BEGIN, baricitinib 4 mg monotherapy-treated patients had less radiographic progression than placebo but the difference was not statistically significant.

Monotherapy vs combination therapy with MTX

Since JAKi are not biological DMARDs, they do not incite an anti-drug antibody response so theoretically concomitant treatment with MTX should be unnecessary. Tofacitinib monotherapy was assessed in Oral-SOLO [19] and Oral-START [17], while baricitinib monotherapy was assessed in RA-BEGIN [18]. Tofacitinib (5 and 10 mg) and baricitinib 4 mg monotherapy were superior to MTX. Barcitinib monotherapy produced a similar therapeutic response to 4 mg plus MTX. However, the sample size of the study was not powered to compare difference between monotherapy vs combination therapy. Indeed, the sample size of the monotherapy was smaller (N = 159) than the MTX plus baricitinib group (N = 215). Furthermore, both Oral-START and RA-BEGIN were trials of patients with early RA while in routine clinical practice, JAKi are used in patients with established disease. These studies showed than JAKi monotherapy is effective, but it is unclear whether monotherapy is as effective as combination therapy. For tofacitinib, this was assessed in ORAL-STRATEGY [21], a 1-year, double-blind, head-to-head, non-inferiority, RCT comparing tofacitinib (5 mg bid) monotherapy, tofacitinib (5 mg bid) plus MTX, and subcutaneous adalimumab (40 mg fortnightly) plus MTX in MTX inadequate responder patients. The primary endpoint was ACR50 response at month 6. This was met by 38, 46 and 44% of patients in tofacitinib monotherapy, tofacitinib plus MTX and adalimumab plus MTX, respectively. Tofacitinib plus MTX was non-inferior to adalimumab plus MTX but non-inferiority was not demonstrated in the tofacitinib monotherapy group, suggesting that in patients who can tolerate MTX, combining tofacitinib with MTX is better than switching to monotherapy.

JAKi in development

Phase II RCT data of upadacitinib [22, 23], filgotinib [24, 25], peficitinib [26, 27] and decernotinib [28, 29] are summarized in Table 2. Overall, these JAKi demonstrated superior ACR responses than placebo-treated group. Recently, phase III trials of upadacitinib in csDMARD inadequate responders (SELECT Next) [30] and biologic inadequate responder (SELECT Beyond) [31] patients have been published that confirmed the efficacy of updacitinib (15 and 30 mg od).

Table 2.

Results of phase II RCT of JAKi in development

Controls JAKi
ACR20 ACR50 ACR70 ACR20 ACR50 ACR70
JAK1 selective
    FilgotinibDARWIN 1 [24] (+ MTX) 50 mg qd 44 15 8 56 33 16
100 mg qd 64 38 21
200 mg qd 69 43 24
    FilgotinibDARWIN 2 [25] (monotherapy) 50 mg qd 29 11 4 67 35 8
100 mg qd 67 36 19
200 mg qd 72 43 13
    UpadacitinibBALANCE 2 [23] (MTX inadequate responders) 6 mg bid 46 18 6 68 46 28
12 mg bid 80 50 16
18 mg bid 64 40 26
    UpadacitinibBALANCE 1 [22] (TNF inadequate responders) 6 mg bid 58 36 26
12 mg bid 71 42 22
18 mg bid 67 38 22
Moderate JAK3 selective
    Dercernotinib [28] 100 mg qd 18 7 3 47 23 10
150 mg qd 67 39 11
200 mg qd 57 35 10
100 mg bid 68 39 22
    Dercernotinibmonotherapy [27] 25 mg bid 29 7 2 39 17 7
50 mg bid 61 32 12
100 mg bid 65 38 18
150 mg bid 66 49 22
    Peficitinib [27] 25 mg qd 44 26 11 44 18 9
50 mg qd 62 33 15
100 mg qd 46 33 17
150 mg qd 57 37 19
    Peficitinib [26] 25 mg qd 29 10 8 22 15 7
50 mg qd 37 25 16
100 mg qd 48 28 19
150 mg qd 56 28 11

Numbers are percentages. bid: bis in die (twice per day); JAK: Janus Kinase; JAKi: Janus Kinase inhibitor; RCT: randomized control trial; qd: quaque die (once per day).

Safety profile of JAKi

Safety data on JAKi are mostly based on RCT and extension studies. Limited real-world data are available for tofacitinib [32]. Caution should be exercised when comparing the incidence rate (IR) of adverse events with biologic agents, in which IR from real-world data [33] are available. In pooled analyses, the IR of serious adverse events was 9.4/100 patient years (95% CI 9.0–0.9) for tofacitinib [34]. For baricitinib, the IR of severe adverse events has not been reported but the IR of specific adverse events are available. The key adverse event data on tofacitinib and baricitinib are summarized in Table 3.

Table 3.

Safety of tofacitinib and baricitinib

Tofacitinib (JAK, JAK3) Baricitinib (JAK1, JAK2) Peficitinib (JAK1, JAK2) Fligotinib (JAK1) Upadacitinib (JAK1) Decernotinib (JAK3)
Serious infection 2.7 (2.5–3.9) 2.9 (2.5–3.4)
Herpes zoster 3–4
Malignancies 0.9(0.8–1) 0.8 (0.6–1.0)
Lymphoma 0.1 (0.1–0.2) 0.09 (0.03–0.19)
Non-melanoma skin cancer 0.6 (0.5–0.7) 0.4 (0.2–0.5)
Gastrointestinal perforation 0.11 (0.07–0.17) 0.05 (0.01–0.13)
DVT/PE NR 0.5 (0.3–0.7)
Changes in laboratory tests (mean + s.d.)
    Haemoglobin (g/dl) +0.47 + 0.05 (5 mg), +0.28 + 0.05 (10 mg) −0.17 Decrease Increase Decreases at higher doses Increase
    Neutrophil (×103/mm3) −1.09 + 0.1 (5 mg), −1.49 + 0.1 (10 mg) −1.08 +0.07 Decrease Decrease Decrease Decrease in high dose
    Lymphocyte count (×103/mm3) −0.24 + 0.03 (5 mg), −0.36 + 0.03 (10 mg) −0.01 (2 mg) −0.05 (4 mg) Decrease No change; some patients developed lymphopaenia Decrease Decrease
    Platelets −30% Increase Decrease Decrease NR NR
    Liver transaminase Increase Increase Increase in high dose group Increase Increase Increase
    Cholesterol Increase Increase Increase Increase Increase Increase
    Creatinine Increase Increase Increase Increase Increase Increase
    Creatinine phosphokinase Increase Increase Increase NR Increase Increase

Safety data are incidence rate per 100 patient years with 95% CI. Laboratory results are mean ± s.d. unless stated otherwise. JAK: Janus Kinase; NR: not reported; DVT/PE: deep vein thrombosis and pulmonary embolus.

Infections

The IR of serious infections was 2.7/100 patient years (95% CI 2.5–3.9) [29] and 2.9/100 patient years (95% CI 2.5–3.4) [35] for tofacitinib and baricitinib, respectively. Both tofacitinib and baricitinib were associated with increased incidence of reactivation of herpes zoster (3–4/100 patient years). This was higher than placebo and exceeded those expected with biologic agents. Risk was highest in Japan and Korea [36]. Concomitant glucocorticoid was an additional risk factor. Reactivation of herpes zoster appears to be a class effect and may be due to inhibition of IFN and IL-15, which are key anti-viral cytokines that signal through JAK1, JAK2 and JAK3. Response to zoster vaccine prior to JAKi treatment has been evaluated in an RCT [37]. Zoster vaccine was given to 112 patients with active RA taking MTX for 2 weeks before receiving either placebo or tofacitinib for 12 weeks, after which placebo-treated patients received tofacitinib in an open-label extension study. Both humoral and cellular immune responses to the zoster vaccine were similar in placebo- and tofacitnib-treated patients. In the open-label extension phase, five patients developed reactivation of zoster. All these patients had suboptimal vaccine immune response to the zoster vaccine.

Malignancies

IR of malignancies excluding non-melanoma skin cancer for tofacitinib [29] and baricitinib [30] were 0.9/100 patient years (95% CI 0.8–1) and 0.8/100 patient years (95% CI 0.6–1.0), respectively. IR for lymphoma was 0.1/100 patient years (95% CI 0.1–0.2) for tofacitinib and 0.09/100 patient years (95% CI 0.03–0.19) for baricitinib. Non-melanoma skin cancer occurred in 0.6/100 patient years (95% CI 0.5–0.7) for tofacitinib and 0.4/100 patient years (95% CI 0.2–0.5) for baricitinib. However, long-term real-world data will be needed to assess accurately the risk of malignancies.

Gastrointestinal perforation

Gastrointestinal perforation is associated with IL-6 inhibition. IL-6 signals via JAK1, JAK2 and TYK2. Therefore, inhibiting IL-6 signalling by JAKi may be associated with gastrointestinal perforation. IR of gastrointestinal perforation was 0.11/100 patient years (95% CI 0.07–0.17) for tofacitinib and 0.05/100 patient years (95% CI 0.01–0.13) for baricitinib. These were numerically lower than that was observed with tocilizumab reported in German biologic registry, which was 0.27/100 patient years [38]. Indeed, a real-world study also suggested the same. Lower gastrointestinal tract perforation was slightly less frequent in tofacitinib-treated than in tocilizumab-treated patients, although the risk was higher than in patients treated by TNF inhibitors [39].

Deep vein thrombosis and pulmonary embolus

Five cases of deep vein thrombosis and pulmonary embolus (DVT/PE) were observed in baricitinib- (IR 1.2/100 patient years) but none in the placebo-treated patients during RCTs [40]. The overall IR of DVT/PE was 0.5/100 patient years (95% CI 0.3–0.7). There was no association between platelet count and the occurrence of DVT/PE. The IR of DVT and PE associated with tofacitinib has not been reported. However, as these adverse events are uncommon, data from registries will be needed to evaluate the association between DVT/PE and JAKi.

Laboratory abnormalities

Haemoglobin

One of the extra-articular features of active RA is anaemia of chronic diseases, which is mediated by hepcidin as part of the acute-phase response [2]. Hence, effective suppression of inflammation should increase haemoglobin (Hb), which has been seen with biologic treatment [41, 42]. However, haemopoietic cytokines including erythropoietin signal via JAK2 [43]. A consequence of JAK2 inhibition is reduced erythropoiesis. This is reflected by Hb changes associated with baricitinib [44], in which a statistically significant greater reduction (P = 0.02) in Hb occurred in patients treated with baricitinib (−0.17 ± 0.02) when compared with placebo-treated patients (−0.12 ± 0.02). Anaemia occurred in 29% of baricitinib-treated vs 26% of placebo-treatment patients. In contrast, a small increase in Hb was observed in a pooled analysis of tofacitinib, which has less inhibitory effect on JAK2: 0.47 g/dl and 0.28 g/dl with 5 and 10 mg, respectively [45]. The likely reason for a smaller increase in Hb with tofacitinib 10 mg is dose-associated inhibition of JAK2, i.e. at low dose (5 mg) tofacitinib is selective for JAK1 and JAK3 but at 10 mg, this selectivity is diminished and JAK2 is inhibited. Compared with MTX, both doses of tofacitinib were associated with a slightly higher incidence of anaemia, although in total <1% of patients experienced major decrease in Hb as defined by decrease from baseline of ⩾3 g/dl or an absolute haemoglobin level of ⩽7 g/dl. Nevertheless, the Summary of Product Characteristics recommends that tofacitinib [46] and baricitinib [47] should not be used in patients who are anaemic (Hb <8g/dl) and treatment should be interrupted when Hb drops below 8 g/dl.

Neutrophil

Decrease in neutrophil count with occasional cases of neutropaenia (Table 3) has been observed with all JAKi. The Summary of Product Characteristics recommends monitoring of neutrophil count in patients taking JAKi [46, 47].

Lymphocyte

Lymphopaenia can occur in patients treated with JAKi. For tofacitinib, lymphopaenia <500 cells/ml occurred in 8.3/100 patient years (95% CI 3.0–18.1) [39]. Multivariate analysis suggested that lymphocyte count <500 cells/ml was associated with increased risk of serious infection [29]. Lymphopaenia was uncommon (<1% of patients) but has also been observed after treatment with baricitinib [48]. The presence of lymphopaenia was associated with a slightly higher overall infection rate, but not for serious infections; however, the lack of association with serious infection may be due to insufficient event and patient number. The Summary of Product Characteristics recommends interrupting JAKi treatment when lymphocyte count is <500 cells/ml [46, 47].

Platelet

Thrombocytosis is a feature of active disease in RA. Suppression of inflammation should reduce platelet number. Surprisingly, treatment by baricitinib was associated with thrombocytosis [41]. Increases in platelet counts above 600×109 cells/l occurred in 2.0% of patients compared with 1.1% of placebo-treated patients [47]. There are no published data on change in platelet count after treatment with tofacitinib.

Biochemistry

Lipids

Increase in both high-density lipoprotein and low-density lipoprotein cholesterol occurred after treatment with both tofacitinib [49] and baricitinib [50], although there was no change in high-density lipoprotein/low-density lipoprotein ratio. IR of major adverse cardiovascular events was 0.58/100 patient years (95% CI 0.39–0.88) for tofacitinib [51] and 0.5/100 patient years (95% CI 0.4–0.7] for baricitinib [40]. A mechanistic study examined the effect of tofacitinib on cholesterol and lipoprotein transport showed that high-density lipoprotein, low-density lipoprotein and total cholesterol levels were lower in

RA patients than in healthy volunteers, while the cholesterol ester fractional catabolic rate was higher in RA patients [52], suggesting cholesterol catabolism is increased. Treatment with tofacitinib reduced inflammation and restored cholesterol catabolism.

Liver transaminases

Increases in liver transaminases (>3× upper limit of normal) occurred in up to 2% of patients receiving tofacitinib [53] and 1.4% of patients treated with baricitinib [47]. Most cases were transient and asymptomatic. Liver function tests should be monitored in patient taking JAKi and when liver transaminases increase significantly, treatment should be temporarily discontinued.

Increase in creatinine levels

A small increase in serum creatinine by 2–4 µmol/l was observed in RCTs when compared with placebo for tofacitinib [54] and baricitinib [55]. This plateaued after 3 months and was not associated with significant renal side effects.

Increase in creatine phosphokinase

Increase in creatine phosphokinase (>5× the upper limit of normal) occurred in up to 1% of patients treated with either tofacitinib [46] or baricitinib [47]. In most cases, creatine phosphokinase elevation was transient, asymptomatic and did not require treatment discontinuation.

Safety of JAKi in development

Exposure is too low to assess major clinical safety such as serious infection. For laboratory measures, changes in biochemistry measures have been observed with all JAKi, suggesting that these are class effects (Table 3).

For haematology measures, decreases in neutrophil and lymphocyte count, including neutropaenia and lymphopaenia, have been observed with all JAKi, suggesting that these are also class effects. However, JAKi have different effects on Hb and platelet counts. Filgotinib and decernotinib increased Hb but peficitinib and upadacitinib, especially at high doses, decreased Hb. Therefore, filgotinib and decernotinib are similar to tofacitinib, while peficitinib and upadacitinib are more akin to baricitinib. The most likely explanation for these differences is inhibition of JAK2, which is expected from the in vitro profile of baricitinib and peficitinib. Another potential differentiating feature among JAKi is the effect on platelet count: both filgotinib and peficitinib decrease platelet numbers while baricitinib increases platelet number. The effect of upadacitinib and decernotinib on platelet number was not reported.

JAK isoform selectivity

Structurally, JAKs are composed of seven homologous regions, JH1–7. JH5–7 are critical for the association of the JAKs with their cognate receptors while JH1 kinase is the active catalytic domain and the main target for JAKi. Because the JH1 domains of the JAK isoforms exhibit a high degree of homology, JAKi are selective but not specific. JAKi selectivity is assessed by in vitro assay of cytokine release and/or pSTAT activation. It is important to note that results from these assays depend on the assay substrates/cell lines and cytokines measured. Furthermore, JAK1, JAK2 and TYK2 are ubiquitously expressed, but JAK3 is predominantly expressed by haematopoietic cells and is highly regulated with cell development and activation.

JAK isoform selectivity is dose dependent

Table 4 shows the concentration needed to inhibit 50% of activation (IC50) of different JAKi. A low IC50 value implies higher potency. JAK isoform selectivity is determined by ratio and difference between IC50s for different JAK isoforms. Table 4 shows not only the IC50 but also the ratio of IC50s. One may consider the ratio as the JAK selectivity dose window’. Conventional pharmacology will select compounds with high potency; however, in the case of JAKi, potency affects the JAK selectivity dose window. Highly potent compounds will have narrow windows while low potency compounds will have a larger selectivity window. Therefore, the clinical impact of JAK isoform selectivity is dependent on dose, cell type, tissue penetration and the genetics of the individual patient. This is probably best illustrated by the change in Hb in tofacitinib 5 mg-treated patients, with less Hb increase in the 10 mg group.

Table 4.

In vitro JAK isoform selectivity

Enzyme essay IC50 (nM)
Compound JAK1 JAK2 JAK3 TYK2 JAK2:JAK1 JAK3:JAK1 TYK2:JAK1
Tofacitinib 15.1 77.4 55.0 489 5.1 3.6 32.4
Baricitinib 4.0 6.6 787 61 1.5 196.8 15.3
Filgotinib 363 2400 >10 000 2600 6.6 >27.5 7.2
Upadacitinib 8 600 139 NA 75 17.4 NA
Peficitinib 3.9 5.0 0.7 4.8 1.3 0.2 1.2
Decernotinib 112 619 74.4 >10 000 5.5 0.67 >89

JAK: Janus kinase; IC50: half maximal inhibitory concentration; TYK2: Non-receptor Tyrosine-protein Kinase 2.

JAK isoforms as therapeutic targets in RA

Since JAK isoforms are important in the signal transduction of multiple cytokines, different JAKi might inhibit different cytokines, which could be important in tailoring treatment to patients. All current JAKi approved and in development have a significant effect on JAK1. Even for the JAK3-selective decernotinib, the ratio between JAK3 and JAK1 inhibition is only 0.6. JAK1 is involved in the signalling transduction of IL-6, IFN and the common γ -chain cytokines including IL-2 and IL-15. Among these, IL-6 is a proven therapeutic target in RA. Some of the side effects associated with IL-6 inhibition, such as increase in liver transaminases and lipids, neutropaenia as well as gastrointestinal perforation, are also associated with JAKi, suggesting that inhibiting IL-6 is a significant mode of action. However, JAKi differ from IL-6 inhibitors in that CRP, while reduced, did not normalize, suggesting that inhibition of IL-6 activity is incomplete [39]. A phase II RCT with very small patient numbers suggested that anti-IFN-γ may be effective in RA [56]. Whether IFN suppression is an important contributor to the therapeutic benefit of JAK1 inhibitors is unclear. However, IFN-γ is important in anti-viral immunity, which is a potential explanation for herpes zoster reactivation. Nevertheless, current evidence suggests that JAK1 is an important therapeutic target in RA.

Whether JAK2 is a good therapeutic target in RA is less clear. Pro-inflammatory cytokines such as GM-CSF signal via JAK2. A phase II RCT of anti-GM-CSF receptor α mAb showed benefit in RA [57]. A head-to-head trial suggested of anti-GM-CSFα mAb was as efficacious as TNF inhibitor [58]. Therefore, inhibiting both JAK1 and JAK2 may produce a greater anti-inflammatory effect by inhibiting GM-CSF. The disadvantage of inhibiting JAK2 is potential side effects associated with inhibiting haematopoetic cytokines including erythropoietin, IL-3 and IL-5, and hormones such as prolactin and growth hormone. The clinical significance of long-term JAK2 inhibition remains unknown. It will be prudent to investigate whether JAK2 inhibition leads to increase in platelet count and the rare cases of DVT/PE. In vitro experimentation has shown that JAK2 is involved in platelet activation. JAK2 inhibitor attenuated collagen-induced platelet aggregation in a dose-dependent manner [59], so an association between thrombosis and JAK2 via effect on platelet seems counterintuitive. Aside from the hitherto unknown effect of JAK2 inhibition and thrombosis, small molecule inhibitors, unlike mAbs, are more likely to have off-target side effects that may not be associated with JAK inhibition but are compound specific.

Based on available data, it is unclear whether JAK3 and TYK2 are desirable therapeutic targets in RA.

Conclusion

JAK isoform selectively of JAKi is relative and not absolute. It is dose and tissue dependent.

Current approved JAKi and those in development significantly inhibit JAK1, which is an effective therapeutic target in RA, although herpes zoster reactivation is probably a class effect of JAK1 inhibitors. The balance of benefit and risk of inhibiting JAK2, JAK3 and TYK2 is uncertain and should be evaluated in the future. The exact role of JAKi in the treatment pathway of RA should be further assessed by head-to-head and strategy trials. The manufacturing cost of JAKi is substantially less than biologics. With the potential of generic JAKi in the future, widening access to more effective treatment for RA is becoming tangible.

Acknowledgement

The CREATE Centre was funded by Arthritis Research UK and Health and Care Research Wales.

Funding: This manuscript has no funding supporting.

Disclosure statement: E.H.C. has received research grants and/or served as member of advisory boards and speaker bureaus of Abbvie, Allergan, Amgen, AstraZeneca, Bio-Cancer, Biogen, BMS, Boehringer Ingelheim, Celgene, Chugai Pharma, Daiichi Sankyo, Eli Lilly, Ferring Pharmaceutical, GSK, Hospira, ISIS, Jazz Pharmaceuticals, Janssen, MedImmune, Merrimack Pharmaceutical, MSD, Napp, Novimmune, Novartis, ObsEva, Pfizer, Regeneron, Roche, R-Pharm, Sanofi, SynAct Pharma, Synovate, Tonix and UCB.

References

  • 1. Yamaoka K, Saharinen P, Pesu M. et al. The Janus kinases (Jaks). Genome Biol 2004;5:253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Dayer JM, Choy E.. Therapeutic targets in rheumatoid arthritis: the interleukin-6 receptor. Rheumatology (Oxford) 2010;49:15–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Rawlings JS, Rosler KM, Harrison DA.. The JAK/STAT signaling pathway. J Cell Sci 2004;117:1281–3. [DOI] [PubMed] [Google Scholar]
  • 4. Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM.. JAK-STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects. Drugs 2017;77:521–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Rodig SJ, Meraz MA, White JM.. Disruption of the Jak1 gene demonstrates obligatory and nonredundant roles of the Jaks in cytokine-induced biologic responses. Cell 1998;93:373–83. [DOI] [PubMed] [Google Scholar]
  • 6. Parganas E, Wang D, Stravopodis D. et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell 1998;93:385–95. [DOI] [PubMed] [Google Scholar]
  • 7. Thomis DC, Gurniak CB, Tivol E, Sharpe AH, Berg LJ.. Defects in B lymphocyte maturation and T lymphocyte activation in mice lacking Jak3. Science 1995;270:794–7. [DOI] [PubMed] [Google Scholar]
  • 8. Macchi P, Villa A, Giliani S. et al. Mutations of Jak-3 gene in patients with autosomal severe combined immune deficiency (SCID). Nature 1995;377:65–8. [DOI] [PubMed] [Google Scholar]
  • 9. Russell SM, Tayebi N, Nakajima H. et al. Mutation of Jak3 in a patient with SCID: essential role of Jak3 in lymphoid development. Science 1995;270:797–800. [DOI] [PubMed] [Google Scholar]
  • 10. Karaghiosoff M, Neubauer H, Lassnig C. et al. Partial impairment of cytokine responses in Tyk2-deficient mice. Immunity 2000;13:549–60. [DOI] [PubMed] [Google Scholar]
  • 11. van Vollenhoven RF, Fleischmann R, Cohen S. et al. Tofacitinib or adalimumab versus placebo in rheumatoid arthritis. N Engl J Med 2012;367:508–19. [DOI] [PubMed] [Google Scholar]
  • 12. Taylor PC, Keystone EC, van der Heijde D. et al. Baricitinib versus placebo or adalimumab in rheumatoid arthritis. N Engl J Med 2017;376:652–62. [DOI] [PubMed] [Google Scholar]
  • 13. Kremer J, Li ZG, Hall S. et al. Tofacitinib in combination with nonbiologic disease‐modifying antirheumatic drugs in patients with active rheumatoid arthritis: a randomized trial. Ann Intern Med 2013;159:253–61. [DOI] [PubMed] [Google Scholar]
  • 14. Dougados M, van der Heijde D, Chen YC. et al. Baricitinib in patients with inadequate response or intolerance to conventional synthetic DMARDs: results from the RA-BUILD study. Ann Rheum Dis 2017;76:88–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Burmester GR, Blanco R, Charles-Schoeman C. et al. Tofacitinib (CP-690, 550) in combination with methotrexate in patients with active rheumatoid arthritis with an inadequate response to tumour necrosis factor inhibitors: a randomised phase 3 trial. Lancet 2013;381:451–60. [DOI] [PubMed] [Google Scholar]
  • 16. Genovese MC, Kremer J, Zamani O. et al. Baricitinib in patients with refractory rheumatoid arthritis. N Engl J Med 2016;374:1243–52. [DOI] [PubMed] [Google Scholar]
  • 17. Lee EB, Fleischmann R, Hall S. et al. Tofacitinib versus methotrexate in rheumatoid arthritis. N Engl J Med 2014;370:2377–86. [DOI] [PubMed] [Google Scholar]
  • 18. Fleischmann R, Schiff M, van der Heijde D. et al. Baricitinib, methotrexate, or combination in patients with rheumatoid arthritis and no or limited prior disease-modifying antirheumatic drug treatment. Arthritis Rheumatol 2017;69:506–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Fleischmann R, Kremer J, Cush J. et al. Placebo-controlled trial of tofacitinib monotherapy in rheumatoid arthritis. N Engl J Med 2012;367:495–507. [DOI] [PubMed] [Google Scholar]
  • 20. van der Heijde D, Tanaka Y, Fleischmann R. et al. Tofacitinib (CP-690, 550) in patients with rheumatoid arthritis receiving methotrexate: twelve-month data from a twenty-four-month phase III randomized radiographic study. Arthritis Rheum 2013;65:559–70. [DOI] [PubMed] [Google Scholar]
  • 21. Fleischmann R, Mysler E, Hall S. et al. Efficacy and safety of tofacitinib monotherapy, tofacitinib with methotrexate, and adalimumab with methotrexate in patients with rheumatoid arthritis (ORAL Strategy): a phase 3b/4, double-blind, head-to-head, randomised controlled trial. Lancet 2017;390:457–68. [DOI] [PubMed] [Google Scholar]
  • 22. Kremer JM, Emery P, Camp HS. et al. A phase IIb study of ABT-494, a selective JAK-1 inhibitor, in patients with rheumatoid arthritis and an inadequate response to anti-tumor necrosis factor therapy. Arthritis Rheumatol 2016;68:2867–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Genovese MC, Smolen JS, Weinblatt ME. et al. Efficacy and safety of ABT-494, a selective JAK-1 inhibitor, in a phase IIb study in patients with rheumatoid arthritis and an inadequate response to methotrexate. Arthritis Rheumatol 2016;68:2857–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Westhovens R, Taylor PC, Alten R. et al. Filgotinib (GLPG0634/GS-6034), an oral JAK1 selective inhibitor, is effective in combination with methotrexate (MTX) in patients with active rheumatoid arthritis and insufficient response to MTX: results from a randomised, dose-finding study (DARWIN 1). Ann Rheum Dis 2017;76:998–1008. [DOI] [PubMed] [Google Scholar]
  • 25. Kavanaugh A, Kremer J, Ponce L. et al. Filgotinib (GLPG0634/GS-6034), an oral selective JAK1 inhibitor, is effective as monotherapy in patients with active rheumatoid arthritis: results from a randomised, dose-finding study (DARWIN 2). Ann Rheum Dis 2017;76:1009–19. [DOI] [PubMed] [Google Scholar]
  • 26. Genovese MC, Greenwald M, Codding C. et al. Peficitinib, a JAK inhibitor, in combination with limited conventional synthetic disease-modifying antirheumatic drugs in the treatment of moderate-to-severe rheumatoid arthritis. Arthritis Rheumatol 2017;69:932–42. [DOI] [PubMed] [Google Scholar]
  • 27. Kivitz AJ, Gutierrez-Ureña SR, Poiley J. et al. Peficitinib, a JAK inhibitor, in the treatment of moderate-to-severe rheumatoid arthritis in patients with an inadequate response to methotrexate. Arthritis Rheumatol 2017;69:709–19. [DOI] [PubMed] [Google Scholar]
  • 28. Fleischmann RM, Damjanov NS, Kivitz AJ. et al. A randomized, double-blind, placebo-controlled, twelve-week, dose-ranging study of decernotinib, an oral selective JAK-3 inhibitor, as monotherapy in patients with active rheumatoid arthritis. Arthritis Rheumatol 2015;67:334–43. [DOI] [PubMed] [Google Scholar]
  • 29. Genovese MC, van Vollenhoven RF, Pacheco-Tena C, Zhang Y, Kinnman N.. VX-509 (decernotinib), an oral selective JAK-3 inhibitor, in combination with methotrexate in patients with rheumatoid arthritis. Arthritis Rheumatol 2016;68:46–55. [DOI] [PubMed] [Google Scholar]
  • 30. Burmester GR, Kremer JM, Van den Bosch F. et al. Safety and efficacy of upadacitinib in patients with rheumatoid arthritis and inadequate response to conventional synthetic disease-modifying anti-rheumatic drugs (SELECT-NEXT): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2018;391:2503–12. [DOI] [PubMed] [Google Scholar]
  • 31. Genovese MC, Fleischmann R, Combe B. et al. Safety and efficacy of upadacitinib in patients with active rheumatoid arthritis refractory to biologic disease-modifying anti-rheumatic drugs (SELECT-BEYOND): a double-blind, randomised controlled phase 3 trial. Lancet 2018;391:2513–24. [DOI] [PubMed] [Google Scholar]
  • 32. Cohen S, Curtis JR, DeMasi R. et al. Worldwide, 3-year, post-marketing surveillance experience with tofacitinib in rheumatoid arthritis. Rheumatol Ther 2018;5:283–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Codreanu C, Damjanov N.. Safety of biologics in rheumatoid arthritis: data from randomized controlled trials and registries. Biologics 2015;9:1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Cohen SB, Tanaka Y, Mariette X. et al. Long-term safety of tofacitinib for the treatment of rheumatoid arthritis up to 8.5 years: integrated analysis of data from the global clinical trials. Ann Rheum Dis 2017;76:1253–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Genovese MC, Smolen JS, Takeuchi T. et al. Safety profile of baricitinib for the treatment of rheumatoid arthritis up to 5.5 years: an updated integrated safety analysis. Presented at the ACR/ARHP Annual Meeting, San Diego, CA, USA, 3–8 November 2017; Abstr. 511. [Google Scholar]
  • 36. Winthrop KL, Curtis JR, Lindsey S. et al. Herpes zoster and tofacitinib. Clinical outcomes and the risk of concomitant therapy. Arthritis Rheumatol 2017;69:1960–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Winthrop KL, Wouters AG, Choy EH. et al. The safety and immunogenicity of live zoster vaccination in patients with rheumatoid arthritis before starting tofacitinib: a randomized phase II trial. Arthritis Rheumatol 2017;69:1969–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Strangfeld A, Richter A, Siegmund B. et al. Risk for lower intestinal perforations in patients with rheumatoid arthritis treated with tocilizumab in comparison to treatment with other biologic or conventional synthetic DMARDs. Ann Rheum Dis 2017;76:504–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Xie F, Yun H, Bernatsky S, Curtis JR.. Brief report: risk of gastrointestinal perforation among rheumatoid arthritis patients receiving tofacitinib, tocilizumab, or other biologic treatments. Arthritis Rheumatol 2016;68:2612–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Weinblatt M, Taylor PC, Burmester G. et al. Cardiovascular safety during treatment with baricitinib in patients with rheumatoid arthritis. Presented at the ACR/ARHP Annual Meeting, San Diego, CA, USA, 3–8 November 2017; Abstr. 499.
  • 41. Doyle MK, Rahman MU, Han C. et al. Treatment with infliximab plus methotrexate improves anemia in patients with rheumatoid arthritis independent of improvement in other clinical outcome measures-a pooled analysis from three large, multicenter, double-blind, randomized clinical trials. Semin Arthritis Rheum 2009;39:123–31. [DOI] [PubMed] [Google Scholar]
  • 42. Paul SK, Montvida O, Best JH. et al. Effectiveness of biologic and non-biologic antirheumatic drugs on anaemia markers in 153,788 patients with rheumatoid arthritis: new evidence from real-world data. Semin Arthritis Rheum 2018;47:478–84. [DOI] [PubMed] [Google Scholar]
  • 43. Kuhrt D, Wojchowski DM.. Emerging EPO and EPO receptor regulators and signal transducers. Blood 2015;125:3536–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Smolen JS, Genovese MC, Takeuchi T. Safety profile of baricitinib in patients with active rheumatoid arthritis: an integrated analysis. Presented at European League Against Rheumatism (EULAR), London, UK, 8–11 June 2016; Poster THU0166.
  • 45. Schulze-Koops H, Strand V, Nduaka C. et al. Analysis of haematological changes in tofacitinib-treated patients with rheumatoid arthritis across phase 3 and long-term extension studies. Rheumatology (Oxford) 2017;56:46–57. [DOI] [PubMed] [Google Scholar]
  • 46.Tofacitinib. Summary of product characteristics. London, UK: European Medicine Agency, 2017.
  • 47.Baricitinib. Summary of product characteristics. London, UK: European Medicine Agency, 2017.
  • 48. Kremer JM, Checn L, Saifan CG. et al. Analysis of neutrophils, lymphocytes, and platelets in pooled phase 2 and phase 3 studies of baricitinib for rheumatoid arthritis. Presented at European League Against Rheumatism (EULAR), Madrid, Spain, 14–17 June 2017; Poster 1325.
  • 49. Charles-Schoeman C, Gonzalez-Gay MA, Kaplan I. et al. Effects of tofacitinib and other DMARDs on lipid profiles in rheumatoid arthritis: implications for the rheumatologist. Semin Arthritis Rheum 2016;46:71–80. [DOI] [PubMed] [Google Scholar]
  • 50. McInnes IB, Kremer J, Emery P. et al. Lipid profile and effect of statin treatment in pooled phase 2 and phase 3 baricitinib studies. In: 2016ACR/ARHP Annual Meeting. Abstract No. 3023.
  • 51. Charles-Schoeman C, Wicker P, Gonzalez-Gay MA. et al. Cardiovascular safety findings in patients with rheumatoid arthritis treated with tofacitinib, an oral Janus kinase inhibitor. Semin Arthritis Rheum 2016;46:261–71. [DOI] [PubMed] [Google Scholar]
  • 52. Charles-Schoeman C, Fleischmann R, Davignon J. et al. Potential mechanisms leading to the abnormal lipid profile in patients with rheumatoid arthritis versus healthy volunteers and reversal by tofacitinib. Arthritis Rheumatol 2015;67:616–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Fleischmann R, Wollenhaupt J, Takiya L. et al. Safety and maintenance of response for tofacitinib monotherapy and combination therapy in rheumatoid arthritis: an analysis of pooled data from open-label long-term extension studies. RMD Open 2017;3:e000491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Isaacs JD, Zuckerman A, Krishnaswami S. et al. Changes in serum creatinine in patients with active rheumatoid arthritis treated with tofacitinib: results from clinical trials. Arthritis Res Ther 2014;16:R158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Keystone EC, Taylor PC, Drescher E. et al. Safety and efficacy of baricitinib at 24 weeks in patients with rheumatoid arthritis who have had an inadequate response to methotrexate. Ann Rheum Dis 2015;74:333–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Sigidin YA, Loukina GV, Skurkovich B, Skurkovich S.. Randomized, double-blind trial of anti-interferon-gamma antibodies in rheumatoid arthritis. Scand J Rheumatol 2001;30:203–7. [DOI] [PubMed] [Google Scholar]
  • 57. Burmester GR, McInnes IB, Kremer J. et al. A randomised phase IIb study of mavrilimumab, a novel GM-CSF receptor alpha monoclonal antibody, in the treatment of rheumatoid arthritis. Ann Rheum Dis 2017;76:1020–30. [DOI] [PubMed] [Google Scholar]
  • 58. Weinblatt ME, McInnes IB, Kremer JM. et al. A randomized phase IIb study of mavrilimumab and golimumab in rheumatoid arthritis. Arthritis Rheumatol 2018;70:49–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Lu WJ, Lin KC, Huang SY. et al. Role of a Janus kinase 2-dependent signaling pathway in platelet activation. Thromb Res 2014;133:1088–96. [DOI] [PubMed] [Google Scholar]

Articles from Rheumatology (Oxford, England) are provided here courtesy of Oxford University Press

RESOURCES