Skip to main content
Pharmacology Research & Perspectives logoLink to Pharmacology Research & Perspectives
. 2019 Nov 15;7(6):e00537. doi: 10.1002/prp2.537

Janus kinase inhibitors for the treatment of rheumatoid arthritis demonstrate similar profiles of in vitro cytokine receptor inhibition

Martin E Dowty 1,, Tsung H Lin 1, Michael I Jesson 1, Martin Hegen 1, David A Martin 1, Vaibhav Katkade 2, Sujatha Menon 3, Jean‐Baptiste Telliez 1
PMCID: PMC6857076  PMID: 31832202

Abstract

Janus kinase (JAK) inhibitors have emerged as an effective class of therapies for various inflammatory diseases such as rheumatoid arthritis (RA). JAK inhibitors function intracellularly by modulating the catalytic activity of JAKs and disrupting the receptor‐mediated signaling of multiple cytokines and growth factors, including those with pro‐inflammatory activity. Understanding the inhibition profiles of different JAK inhibitors, based on the associated cytokine receptors and downstream inflammatory pathways affected, is important to identify the potential mechanisms for observed differences in efficacy and safety. This study applied an integrated modeling approach, using in vitro whole blood cytokine inhibition potencies and plasma pharmacokinetics, to determine JAK‐dependent cytokine receptor inhibition profiles, in the context of doses estimated to provide a similar clinical response in RA clinical trials. The calculated profiles of cytokine receptor inhibition for the JAK inhibitors tofacitinib, baricitinib, upadacitinib, and filgotinib and its metabolite, were generally similar when clinically efficacious doses for RA were considered. Only minor numerical differences in percentage cytokine receptor inhibition were observed, suggesting limited differentiation of these inhibitors based on JAK pharmacology, with each showing a differential selectivity for JAK1 heterodimer inhibition. Nevertheless, only robust clinical testing involving head‐to‐head studies will ultimately determine whether there are clinically meaningful differences between these JAK inhibitors. Furthermore, ongoing and future research into inhibitors with alternative JAK selectivity remains of clinical importance. Thus, all JAK inhibitors should be characterized via thorough preclinical, metabolic and pharmacological evaluation, adequate long‐term clinical data, and when available, real‐world experience.

Keywords: cytokine receptors, immunopharmacology, inhibition, JAK‐STAT, rheumatoid arthritis


Abbreviations

ACR

American College of Rheumatology

ATP

adenosine triphosphate

BID

twice daily

Cav

average plasma concentration

Cav,u

average plasma concentration, unbound

CD

cluster of differentiation

CI

confidence interval

CRP

C‐reactive protein

EDTA

ethylenediaminetetraacetic acid

EMA

European Medicines Agency

EPO

erythropoietin

FDA

United States Food and Drug Administration

fu

fraction unbound

G‐CSF

granulocyte colony‐stimulating factor

GM‐CSF

granulocyte‐macrophage colony‐stimulating factor

HZ

herpes zoster

IC50

half‐maximal inhibitory concentration

IC50,u

half‐maximal inhibitory concentration, unbound

ICxx

proportion of JAK inhibitory concentration

IFN

interferon

IL

interleukin

IR

incidence rate

JAK

Janus kinase

KM

Michaelis‐Menten constant

LC‐MS/MS

liquid chromatography‐tandem mass spectrometry

NA

not applicable

NK

natural killer

PBS

phosphate‐buffered saline

pSTAT

phosphorylated signal transducer and activator of transcription protein

QD

once daily

RA

rheumatoid arthritis

RFU

relative fluorescence unit

SD

standard deviation

TPO

thrombopoietin

TYK

tyrosine kinase

1. INTRODUCTION

Inflammatory diseases, such as rheumatoid arthritis (RA), are chronic immune‐mediated conditions, which collectively have an estimated prevalence of 5%‐7% in Western society,1 can be highly disabling2 and painful, and cause reduced quality of life.3

Inflammation is a complex immune response involving diverse cell types and feedback loops promoting the production of pro‐inflammatory mediators.1, 4 Notably, cytokines are a group of structurally unrelated protein messengers that, upon binding to and activating their specific receptors on immune cells, transmit signals that regulate the expression of numerous inflammatory genes. Under dysregulated conditions, cytokines play a key role in the pathologic inflammatory response characteristic of immune‐mediated diseases.1, 5, 6

Janus kinases (JAKs) are enzymes that are essential in the signaling pathways of type I and type II cell‐surface cytokine receptors which lack intrinsic kinase catalytic activity.5, 6 There are four different JAK isoforms in humans (JAK1, JAK2, JAK3, and TYK2) which function in pairs to transmit intracellular signals from cytokine‐activated receptors.5 JAK1 pairs with JAK3 to mediate the signaling pathways of common gamma chain (γc) cytokines. JAK1 also pairs with JAK2 and/or TYK2 for signaling through receptors of the IL‐6, IL‐10, and interferon cytokine families. Additionally, JAK2 pairs with TYK2 for signaling through IL‐12 and IL‐23 cytokine receptors, and with itself for signaling from receptors for hormone‐like cytokines such as erythropoietin (EPO).5

Indeed, there are over 50 cytokines that signal through JAK‐mediated type I and II receptors,6 many implicated in inflammatory disease pathophysiology.1, 4 Notably, IL‐6 induces acute‐phase proteins such as C‐reactive protein (CRP) and may be involved in the autoimmune process through B‐cell modulation and T‐helper‐17‐cell differentiation.7 Common γc cytokines play a key role in adaptive immune functions, for example, in T‐cell and natural killer (NK)‐cell differentiation.5 JAKs are therefore an attractive therapeutic target for RA and other inflammatory diseases.6

Biologic drugs for treating inflammatory diseases target extracellular elements of the inflammation pathway such as cytokines or their receptors.1, 4, 5 In contrast, targeted synthetic JAK inhibitors reduce inflammation by directly binding to and modulating the intracellular catalytic activity of JAKs and disrupting the receptor‐mediated signaling of multiple cytokines, including those of pro‐inflammatory pathways.4

Current JAK inhibitor drugs were designed to be selective for certain JAK isoforms.8 Tofacitinib is an oral, small molecule JAK inhibitor for the treatment of RA, psoriatic arthritis, and ulcerative colitis. Tofacitinib is reported to have functional selectivity for heterodimer pairs involving JAK1 and/or JAK3.4, 5 Other JAK inhibitors with ongoing or completed late development RA clinical trials include baricitinib, upadacitinib, and filgotinib. Baricitinib is approved for the treatment of RA, with reported preferential selectivity for JAK1 and JAK2.9 Upadacitinib and filgotinib are under investigation for the treatment of inflammatory diseases including RA; both drugs, as well as filgotinib's active metabolite, are reported to selectively inhibit JAK1.10, 11, 12

Understanding the different inhibition profiles of JAK inhibitors, based on the associated cytokine receptors and downstream inflammatory pathways inhibited, is important to better characterize the impact of JAK inhibition and the potential rationale for differences in clinical efficacy and safety profiles. Although JAK selectivity can be evaluated using enzymatic assays, the selectivity observed using biochemical assays may not necessarily be maintained when evaluated under physiologic cellular conditions.13 Suggested reasons for this discrepancy include differences within the complex intracellular milieu, particularly when assessing activity in primary cells (ie, human whole blood), such as the difference in the adenosine triphosphate (ATP) Michaelis‐Menten constant (KM) of each kinase.13

The objective of this study was to characterize cytokine receptor inhibition profiles of JAK inhibitors for the treatment of RA, in the context of drug doses that provided a similar response in patients in RA clinical trial settings. To achieve this, we applied an integrated modeling approach, using knowledge of both intracellular JAK‐dependent cytokine signaling inhibition potencies and in vivo pharmacokinetics.

2. MATERIALS AND METHODS

2.1. Drug compounds

Tofacitinib and the active metabolite of filgotinib were synthesized by Pfizer Discovery Research. Baricitinib (Catalog No. G‐5743), upadacitinib (Catalog No. M15685), and filgotinib (Catalog No. I‐9794) were purchased from Advanced ChemBlocks Inc. The compounds were prepared as 30 mmol/L stocks in 100% dimethyl sulfoxide (DMSO). An 11‐point dilution series was created in DMSO with a maximum concentration of 10 mmol/L or 30 mmol/L. Further dilution was performed by adding 4 µL of the above compound solutions into 96 µL of phosphate‐buffered saline (PBS) with a maximum concentration of 400 µmol/L or 1200 µmol/L.

2.2. Cytokines

EPO (Catalog No. 287 TC), G‐CSF (Catalog No. 214‐GS), GM‐CSF (Catalog No. 215‐GM), IFNα (Catalog No. 11200‐2), IFNγ (Catalog No. 285‐IF), IL‐2 (Catalog No. 202‐IL), IL‐3 (Catalog No. 203‐IL), IL‐4 (Catalog No. 204‐IL), IL‐6 (Catalog No. 206‐IL), IL‐7 (Catalog No. 207‐IL), IL‐10 (Catalog No. 217‐IL), IL‐12 (Catalog No. 219‐IL), IL‐13 (Catalog No. 213‐IL), IL‐15 (Catalog No. 247‐IL), IL‐23 (Catalog No. 1290‐IL), IL‐27 (Catalog No. 2526‐IL), and TPO (Catalog No. 288‐TP) were obtained from R&D Systems. IL‐21 (Catalog No. AF‐200‐21) was purchased from PeproTech.

2.3. Antibodies

Antibodies specific to phosphorylated signal transducer and activator of transcription proteins (pSTATs) and cluster of differentiation (CD) molecules were supplied by BD Biosciences (San Jose, CA, USA): anti‐pSTAT1‐AF647 (Catalog No. 612597); anti‐pSTAT1‐AF488 (Catalog No. 612596); anti‐pSTAT3‐AF647 (Catalog No. 557815); anti‐pSTAT4‐AF647 (Catalog No. 558137); anti‐pSTAT5‐AF488 (Catalog No. 612598); anti‐pSTAT5‐AF647 (Catalog No. 612599); anti‐CD3‐BV421 (Catalog No. 562426); anti‐CD3‐BV650 (Catalog No. 563852); anti‐CD14‐Pacific Blue (Catalog. No. 558121); anti‐CD14‐AF488 (Catalog No. 557700); anti‐CD19‐BV421 (Catalog No. 562440).

2.4. Cells

Cryopreserved human bone marrow CD34+ cells were purchased from STEMCELL Technologies (Catalog No. 70002.3; Vancouver, Canada). Frozen bone marrow CD34+ cells were thawed, washed once with StemSpan™ SFEM II medium (Catalog No. 09605; STEMCELL Technologies), suspended in StemSpan™ SFEM II medium containing StemSpan™ Erythroid Expansion Supplement (Catalog No. 02692; STEMCELL Technologies), and cultured for 7 days. CD34+ cells were then harvested, washed once with Dulbecco's (D)‐PBS, and suspended at 0.5 x 106 cells/mL in human whole blood to be used in the EPO stimulation assay.

Human whole blood was collected from 13 healthy donors (seven males and six females) via venipuncture into Vacutainer collection tubes containing sodium heparin, in accordance with Pfizer protocols (Protocol No. GOHW RDP‐01) approved by the Shulman Institutional Review Board. Blood was warmed to 37°C prior to use.

2.5. Other materials

Phosflow Lyse/Fix Buffer 5X (Catalog No. 558049) was purchased from BD Biosciences. Fetal bovine serum (Catalog No. A3160601) was purchased from Thermo Fisher Scientific and sodium azide (Catalog No. S8032) was obtained from Sigma Aldrich. D‐PBS (without Ca2+ or Mg2+) was obtained from Invitrogen (Catalog No. 14190). Glutathione S‐transferase (GST)‐tagged recombinant human kinase domains of JAK1, JAK2, and JAK3 were purchased from Thermo Fisher Scientific. His‐tagged recombinant human TYK2 kinase domain was expressed in SF21/baculovirus and purified using a two‐step affinity (Ni‐nitrilotriacetic acid) and size‐exclusion (SEC S200) purification method.

2.6. Enzymatic potency of JAK inhibitors

The potency of tofacitinib, upadacitinib, baricitinib, and filgotinib and its metabolite against the four JAK isoforms, JAK1, JAK2, JAK3, and TYK2, was measured in terms of half‐maximal inhibitory concentration (IC50). Human JAK activity was determined using a microfluidic assay to monitor phosphorylation of a synthetic peptide by the recombinant human kinase domain of each of the JAK isoforms.

Test compounds were solubilized in DMSO to a stock concentration of 30 mmol/L. Compounds were diluted in DMSO to create an 11‐point half‐log dilution series with a maximum concentration of 600 μmol/L. The test compound plate also contained positive control wells containing a proprietary potent inhibitor to define 100% inhibition and negative control wells containing DMSO to define no inhibition. The test compounds were diluted 1:60 in the assay, resulting in a final assay compound concentration range of 10 μmol/L to 100 pmol/L, with a final assay concentration of 1.7% DMSO. Test compounds and controls solubilized in 100% DMSO were added (250 nL) to a 384‐well polypropylene plate (Corning) using a non‐contact acoustic dispenser.

Kinase assays were carried out at room temperature in 15 μL reaction buffer containing 20 mmol/L HEPES (pH 7.4), 10 mmol/L magnesium chloride, 0.01% bovine serum albumin, 0.0005% Tween 20, and 1 mmol/L dithiothreitol. Reaction mixtures contained 1 μmol/L of a fluorescently labeled synthetic peptide (5FAM‐KKSRGDYMTMQID for JAK1 and TYK2, and FITC‐KGGEEEEYFELVKK for JAK2 and JAK3) at a concentration less than the apparent KM. Reaction mixtures contained 1 mmol/L ATP.

Test compound was added to the buffer containing ATP and substrate, and immediately after this step, the enzyme was added to begin the reaction. The assays were stopped with 15 μL of a buffer containing 180 mmol/L HEPES (pH = 7.4), 20 mmol/L ethylenediaminetetraacetic acid (EDTA), and 0.2% coating reagent, resulting in a final concentration of 10 mmol/L EDTA, 0.1% coating reagent, and 100 mmol/L HEPES (pH = 7.4). The assay plates were placed on a Caliper Life Science Lab Chip 3000 (LC3000) instrument or Caliper Life Science EZ Reader instrument and each well was sampled using appropriate separation conditions to determine the level of phosphorylation.

2.7. In vitro analyses

2.7.1. Whole blood potency

Inhibition curves and IC50 values were determined for cytokine signaling of tofacitinib, baricitinib, upadacitinib, and filgotinib and its metabolite. For each cytokine assay, all five compounds were tested side‐by‐side in quadruplicate (ie, using blood from four donors).

A total of 90 µL/well of human whole blood was added to 96‐well polypropylene plates (Catalog No. 10755‐246; VWR), followed by 5 µL test compound solutions prepared per above to give a maximum concentration of 20 µmol/L or 60 µmol/L. The plates were mixed and incubated for 60 minutes at 37ºC. Then, 5 µL of PBS or cytokine was added to each well (5 µL/well; final, 10 U/mL EPO, 100 ng/mL G‐CSF, 10 ng/mL GM‐CSF, 5000 U/mL IFNα, 100 ng/mL IFNγ, 50 ng/mL IL‐2, 50 ng/mL IL‐3, 5 ng/mL IL‐4, 50 ng/mL IL‐6, 30 ng/mL IL‐7, 30 ng/mL IL‐10, 30 ng/mL IL‐12, 30 ng/mL IL‐13, 30 ng/mL IL‐15, 50 ng/mL IL‐21, 25 ng/mL IL‐23, 1000 ng/mL IL‐27) and incubated for 15 minutes. Anti‐cell surface antibodies were added 15 minutes prior to cytokine stimulation; anti‐CD14‐Pacific Blue (0.5 µL/well) to GM‐CSF, IFNγ, and TPO‐treated samples; anti‐CD3‐BV650 (0.5 µL/well) plus anti‐CD14‐Pacific Blue (0.5 µL/well) to IL‐6‐treated samples; and anti‐CD19‐BV421 (0.5 µL/well) plus anti‐CD14‐AF488 (0.5 µL/well) to IL‐13‐treated samples.

The reaction was quenched by adding Lyse/Fix Buffer to all wells at 700 µL/well and incubating for 20 minutes at 37°C; after washing with staining buffer (D‐PBS containing 0.5% fetal bovine serum and 0.1% sodium azide), 350 µL ice‐cold 90% methanol (Catalog No. 4823‐32; RICCA Chemical) was added to each well and incubated at 4ºC for 30 minutes. One more wash was done with staining buffer and all samples were finally suspended in 150 µL/well of the desired anti‐pSTAT antibodies at 1:150 dilution in staining buffer; anti‐pSTAT1‐AF647 in IFNγ‐treated samples; anti‐pSTAT1‐AF488 and anti‐pSTAT3‐AF647 in IFNα‐, IL‐6‐, and IL‐27‐treated samples; anti‐pSTAT3‐AF647 in G‐CSF‐, IL‐10‐, IL‐21‐, and IL‐23‐treated samples; anti‐pSTAT4‐AF647 in IL‐12‐treated samples; anti‐pSTAT5‐AF647 in EPO‐, GM‐CSF‐, IL‐2‐, IL‐3‐, IL‐7‐, IL‐15‐, and TPO‐treated samples; anti‐pSTAT5‐AF488 and anti‐pSTAT6‐AF647 in IL‐4‐treated samples; and anti‐pSTAT6‐AF647 in IL‐13‐treated samples.

After overnight incubation at 4°C, all the samples were transferred into 96‐well polypropylene U‐bottom plates (Catalog No. 072‐00‐745; Thermo Fisher Scientific) and flow cytometric analysis was performed on an LSR Fortessa equipped with a High Throughput Sampler plate loader (BD Biosciences). The lymphocyte population was gated for pSTAT histogram analysis for IFNα‐, IL‐2‐, IL‐3‐, IL‐4‐, IL‐7‐, IL‐10‐, IL‐12‐, IL‐15‐, IL‐21‐, IL‐23‐, and IL‐27‐treated samples; granulocyte population for G‐CSF‐treated samples; CD14+ cells for GM‐CSF‐, IFNγ‐, and TPO‐treated samples; CD3+ cells and CD14+ cells for IL‐6‐treated samples; CD14+ cells and CD19+ cells for IL‐13‐treated samples; all events (entire populations) for EPO‐treated cells. Background fluorescence was defined using unstimulated cells and a gate was placed at the foot of the peak to include ~0.5% gated population.

Histogram statistical analysis was performed using FACSDiva version 8.0.1 (BD Biosciences). Relative fluorescence unit (RFU), which measures the level of pSTAT, was calculated by multiplying the percent positive population and its mean fluorescence. Data from 11 compound concentrations (singlicate at each concentration) were normalized as a percentage of control, as shown in Equation 1:

%ofcontrol=100×ABCB (1)

A, RFU from wells containing compound and cytokine; B, RFU from wells without cytokine (minimum fluorescence); C, RFU from wells containing only cytokine (maximum fluorescence).

Inhibition curves and IC50 values were determined using Prism software (Version 7, GraphPad).

2.7.2. Plasma protein binding

An equilibrium dialysis method was used to determine the plasma fraction unbound (fu) values, as described previously.14 Briefly, dialysis membranes (MWCO 12‐14K) and 96‐well dialysis devices were assembled following the manufacturer's instructions (HTDialysis, LLC, Gales Ferry, CT, USA). Human plasma samples (pooled mixed gender; BioIVT, www.bioivt.com) containing 1 μmol/L test compounds with 1% DMSO were dialyzed against PBS for 6 hours in a humidified incubator (75% relative humidity; 5% CO2/95% air) at 37°C with shaking at 450 RPM. Quadruplicates of binding were measured for each compound. Samples were matrix‐matched and quenched with cold acetonitrile containing internal standard(s). The solutions were centrifuged and the supernatant was analyzed using liquid chromatography‐tandem mass spectrometry (LC‐MS/MS).

2.7.3. Blood‐to‐plasma ratio

Human blood‐to‐plasma ratio was measured by Unilabs York Bioanalytical Solutions. Test compounds were incubated in quadruplicate with fresh human blood (mixed gender, at least 1 sample per gender, Clinical Trials Laboratory Services Ltd, London, UK) at 1 μmol/L in a humidified incubator (95% relative humidity; 5% CO2/95% air) for 1 and 3 hours at 37°C with shaking at 450 RPM. Following incubation, plasma samples were obtained by centrifuging blood samples at 3000g for 7 minutes. Both plasma and blood samples were matrix‐matched with each other and quenched with acetonitrile containing internal standard. The solutions were centrifuged, and the supernatant was analyzed by LC‐MS/MS. Peak area ratios were used to calculate blood‐to‐plasma ratio.

2.8. Data analyses

An integrated modeling approach was applied to determine cytokine receptor inhibition profiles. Whole blood IC50 values were converted to unbound values (IC50,u) using measured blood‐to‐plasma ratios and measured fu values, as shown in Equation 2:

IC50,u=IC50blood:plasma ratio×fu (2)

Human daily average plasma concentrations (Cav) were used as reported or predicted in the literature for tofacitinib, baricitinib, upadacitinib, and filgotinib and its metabolite, for doses that we determined to be clinically meaningful (ie, tofacitinib 5 mg twice daily [BID], baricitinib 4 mg once daily [QD], upadacitinib 15 mg QD, and filgotinib 200 mg QD). Doses were selected on the rationale that they provided a generally comparable proportion of patients meeting American College of Rheumatology (ACR) response criteria in clinical trial settings.15, 16, 17, 18 Cav as a measure of average daily drug plasma concentration was used as it has been shown to be a predictive exposure metric of the efficacy of tofacitinib and filgotinib, rather than Cmax or Cmin.19, 20, 21 Cav values were converted to unbound values (Cav,u) using fu values, as shown in Equation 3:

Cav,u=Cav×fu (3)

Proportions (%) of JAK receptor inhibitory concentrations (ICxx) at clinically meaningful doses were then calculated, as shown in Equation 4:

ICxx=100×Cav,uIC50,u+Cav,u (4)

3. RESULTS

3.1. Enzymatic potency of JAK inhibitors

Table 1 presents IC50 values for the inhibition of the JAK isoforms (JAK1, JAK2, JAK3, and TYK2) by tofacitinib, baricitinib, upadacitinib, and filgotinib, measured using an enzymatic assay. Measurements were performed once for the filgotinib metabolite and showed weak inhibition (therefore data not shown).

Table 1.

Mean IC50 values in enzymatic assay for tofacitinib, baricitinib, upadacitinib, and filgotinib inhibition of JAK1, JAK2, JAK3, and TYK2

  IC50 (nmol/L) [n]a, b
JAK1 JAK2 JAK3 TYK2
Tofacitinib 15 [7] 71 [7] 45 [8] 472 [10]
Baricitinib 0.78 [3] 2 [3] 253 [3] 14 [3]
Upadacitinib 0.76 [3] 19 [3] 224 [3] 118 [3]
Filgotinib 45 [3] 357 [3] 9097 [3] 397 [3]

Abbreviations: ATP, adenosine triphosphate; IC50, half‐maximal inhibitory concentration; JAK, Janus kinase; TYK, tyrosine kinase.

a

IC50 values represent the geometric mean of independent experiments; [n] denotes the number of experiments.

b

All reactions were carried out in the presence of 1 mmol/L ATP.

3.2. In vitro potency of JAK inhibitors

3.2.1. Whole blood potency

Table 2 presents mean IC50 values for inhibition of cytokine receptors by tofacitinib, baricitinib, upadacitinib, and filgotinib and its metabolite, measured in human whole blood side‐by‐side. Measurements were performed for each compound at 11 different concentrations, and using blood from four donors per cytokine. Additionally, Figure S1 presents IC50 curves (one of the four obtained) for representative cytokine receptors from different receptor classes (IFNγ, IFNα, IL‐6, IL‐15, IL‐12, and EPO) for illustrative purposes.

Table 2.

Mean IC50 and IC50,u values in human whole blood from four donors for tofacitinib, baricitinib, upadacitinib, and filgotinib

JAK signaling pair Gated cell population STAT phosphorylation Cytokine receptor Tofacitiniba Baricitinibb Upadacitinibc Filgotinibd Filgotinib metabolitee
Mean (SD) human whole blood, nmol/L
IC50 IC50,u IC50 IC50,u IC50 IC50,u IC50 IC50,u IC50 IC50,u
JAK1/JAK2 CD14+ cells pSTAT1 IFNγ 225 (18) 114 109 (12) 49 109 (11) 53 6953 (568) 2792 >60 000 (NA)  
JAK1/JAK2 Granulocytes pSTAT3 G‐CSF 247 (12) 125 146 (17) 65 78 (11) 38 6108 (900) 2453 >52 300 (NA)  
JAK1/TYK2 Lymphocytes pSTAT1 IFNα 58 (7.8) 29 46 (5.6) 21 21 (1.5) 10 1323 (155) 531 11 525 (826) 4441
JAK1/TYK2 Lymphocytes pSTAT3 IFNα 48 (8.2) 24 38 (5.9) 17 19 (2.2) 9.2 1057 (143) 425 9880 (1260) 3807
JAK1/TYK2 Lymphocytes pSTAT3 IL‐10 126 (62) 64 90 (43) 40 90 (31) 43 2550 (1083) 1024 >42 100 (NA)  
JAK1/JAK2, JAK1/TYK2 CD3+ cells pSTAT1 IL‐6 30 (4.4) 15 21 (1.8) 9.2 12 (0.47) 5.9 671 (131) 270 7230 (1169) 2786
JAK1/JAK2, JAK1/TYK2 CD3+ cells pSTAT3 IL‐6 340 (38) 173 229 (27) 102 152 (26) 73 4938 (425) 1983 >54 700 (NA)  
JAK1/JAK2, JAK1/TYK2 CD14+ cells pSTAT1 IL‐6 48 (9.0) 24 33 (5.3) 15 26 (3.9) 13 810 (207) 325 7583 (1158) 2922
JAK1/JAK2, JAK1/TYK2 CD14+ cells pSTAT6 IL‐13 275 (30) 140 196 (24) 88 126 (16) 61 5143 (709) 2065 >60 000 (NA)  
JAK1/JAK2, JAK1/TYK2 CD19+ cells pSTAT6 IL‐13 119 (30) 61 73 (17) 33 72 (13) 35 2125 (359) 853 40 250 (11 656) 15 509
JAK1/JAK2, JAK1/TYK2 Lymphocytes pSTAT1 IL‐27 73 (9.5) 37 34 (4.2) 15 52 (4.2) 25 2113 (187) 848 21 525 (3351) 8294
JAK1/JAK2, JAK1/TYK2 Lymphocytes pSTAT3 IL‐27 56 (9.3) 29 30 (5.4) 13 44 (6.4) 21 1165 (121) 468 9605 (1113) 3701
JAK1/JAK3 Lymphocytes pSTAT5 IL‐2 54 (6.0) 27 90 (18) 40 50 (8.3) 24 2660 (611) 1068 28 650 (5991) 11 039
JAK1/JAK3 Lymphocytes pSTAT5 IL‐4 11 (2.7) 5.4 14 (3.2) 6.1 8.2 (3.7) 4.0 363 (140) 146 4968 (1891) 1914
JAK1/JAK3 Lymphocytes pSTAT6 IL‐4 84 (23) 42 131 (36) 59 77 (20) 37 3623 (897) 1455 >54 300 (NA)  
JAK1/JAK3 Lymphocytes pSTAT5 IL‐7 72 (23) 36 126 (46) 56 101 (31) 49 3625 (988) 1456 41600 (9523) 16 029
JAK1/JAK3 Lymphocytes pSTAT5 IL‐15 67 (1.9) 34 110 (15) 49 64 (6.2) 31 3378 (980) 1357 31700 (8358) 12215
JAK1/JAK3 Lymphocytes pSTAT3 IL‐21 79 (14) 40 142 (16) 63 51 (5.0) 25 3985 (278) 1601 >48700 (NA)  
JAK2/TYK2 Lymphocytes pSTAT4 IL‐12 265 (93) 134 91 (26) 41 145 (53) 70 8028 (2149) 3224 >5900 (NA)  
JAK2/TYK2 Lymphocytes pSTAT3 IL‐23 408 (135) 207 115 (39) 51 242 (79) 117 13193 (3470) 5299 >58100 (NA)  
JAK2/JAK2 Erythroid progenitor cells pSTAT5 EPO 290 (88) 148 105 (33) 47 174 (66) 84 9895 (3481) 3974 53 550 (3748) 2034
JAK2/JAK2 CD14+ cells pSTAT5 TPO 608 (189) 309 243 (42) 109 297 (66) 143 >17 600 (NA)   >60 000 (NA)  
JAK2/JAK2 Lymphocytes pSTAT5 IL‐3 825 (54) 419 309 (23) 138 326 (33) 158 >20 000 (NA)   >6000 (NA)  
JAK2/JAK2 CD14+ cells pSTAT5 GM‐CSF 1758 (237) 893 637 (110) 285 782 (138) 377 >20 000 (NA)   >60 000 (NA)  

Missing data for filgotinib are due to the IC50 values not being measurable.

Abbreviations: CD, cluster of differentiation; EPO, erythropoietin; fu, fraction unbound; G‐CSF, granulocyte colony‐stimulating factor; GM‐CSF, granulocyte‐macrophage colony‐stimulating factor; IC50, half‐maximal inhibitory concentration; IC50,u, half‐maximal inhibitory concentration, unbound; IFN, interferon; IL, interleukin; JAK, Janus kinase; NA, not applicable; pSTAT, phosphorylated signal transducer and activator of transcription protein; SD, standard deviation; TPO, thrombopoietin; TYK, tyrosine kinase.

a

Blood‐to‐plasma ratio, 1.20; fu, 0.61.

b

Blood‐to‐plasma ratio, 1.32; fu, 0.59.

c

Blood‐to‐plasma ratio, 1.16; fu, 0.56.

d

Blood‐to‐plasma ratio, 1.22; fu, 0.49.

e

Blood‐to‐plasma ratio, 1.09; fu, 0.42.

3.2.2. Plasma protein binding

From quadruplicate measurements, mean fu values were determined to be 0.61 for tofacitinib, 0.59 for baricitinib, 0.56 for upadacitinib, 0.49 for filgotinib, and 0.42 for filgotinib metabolite.

3.2.3. Blood‐to‐plasma ratios

From quadruplicate measurements, mean blood‐to‐plasma ratios were determined to be 1.20 for tofacitinib, 1.32 for baricitinib, 1.16 for upadacitinib, 1.22 for filgotinib, and 1.09 for filgotinib metabolite.

3.2.4. Unbound half‐maximal inhibitory concentrations

Per Equation 2, the measured IC50 values, fu values, and blood‐to‐plasma ratios were used to calculate IC50,u values, which are presented alongside the IC50 values in Table 2.

3.3. Cytokine inhibition

Table 3 presents Cav values obtained from the literature for doses that were determined to be clinically meaningful (based on generally comparable ACR response rates), and calculated Cav,u values (Equation 3) for tofacitinib, baricitinib, upadacitinib, and filgotinib and its metabolite.

Table 3.

Actual and unbound Cav values for clinically meaningful doses of tofacitinib, baricitinib, upadacitinib, and filgotinib and its metabolite

  Dose Cav (nmol/L) Cav,u (nmol/L) fu
Tofacitinib19 5 mg BIDa 68 41.0 0.61
Baricitinib9 4 mg QDb 27 16.0 0.59
Upadacitinib47, 48 15 mg QDc 49 27.4 0.56
Filgotinib11 200 mg QDc 474 232.0 0.49
Filgotinib metabolite11 200 mg QDc 7438 3124.0 0.42

Abbreviations: BID, twice daily; Cav, average plasma concentration; Cav,u, average plasma concentration, unbound; EMA, European Medicines Agency; FDA, United States Food and Drug Administration; fu, fraction unbound; QD, once daily.

a

This dose is approved by both the EMA and the FDA.

b

This dose is approved by the EMA but not approved by the FDA (dosage of 2 mg QD is approved by the FDA).

c

These JAK inhibitors are under investigation.

Calculated ICxx data (Equation 4) are summarized in Figure 1. Cytokine receptor inhibition profiles across a broad range of pathways were generally similar among the JAK inhibitors studied, and generally consistent across each JAK pair. There appeared to be some small numerical differences among the JAK inhibitors analyzed. Relative inhibition of most JAK1/3‐mediated cytokine receptors (γc cytokine receptors; IL‐2, IL‐4, IL‐7, and IL‐15) appeared to be numerically greater with tofacitinib vs other JAK inhibitors; inhibition of JAK2/TYK2‐mediated cytokine receptors (IL‐12, IL‐23, and EPO) appeared to be numerically greater with baricitinib vs other JAK inhibitors; and inhibition of JAK1/JAK2‐mediated cytokine receptors (IFNγ and G‐CSF) and JAK2‐mediated cytokine receptors (TPO, IL‐3, and GM‐CSF) appeared to be numerically greater with upadacitinib vs other JAK inhibitors.

Figure 1.

Figure 1

Cytokine receptor inhibitory concentrations for modeled exposures of tofacitinib 5 mg BID, baricitinib 4 mg QD, upadacitinib 15 mg QD, and filgotinib/metabolite 200 mg QD.

BID, twice daily; CD, cluster of differentiation; EPO, erythropoietin; G‐CSF, granulocyte colony‐stimulating factor; GM‐CSF, granulocyte‐macrophage colony‐stimulating factor; ICxx, proportion of JAK inhibitory concentration; IFN, interferon; IL, interleukin; JAK, Janus kinase; pSTAT, phosphorylated signal transducer and activator of transcription protein; QD, once daily; TPO, thrombopoietin; TYK, tyrosine kinase

4. DISCUSSION

The relationship between the clinical profiles of JAK inhibitors and their individual selectivity for JAK isoforms is not clear. To explore this, we evaluated the activity of JAK inhibitors approved or under investigation for RA treatment. Using an integrated modeling approach, we determined cytokine receptor inhibition profiles of clinically equivalent doses (based on proportion of patients meeting ACR response criteria) of tofacitinib (5 mg BID), baricitinib (4 mg QD), upadacitinib (15 mg QD), and filgotinib and its metabolite (200 mg QD).

The IC50 values determined in this study were consistent with the literature, with each JAK inhibitor demonstrating greater selectivity for JAK1 vs other isoforms.4, 5, 9, 10, 11 JAK1 is presumed to be a key target for RA and other inflammatory diseases since it associates with receptors for γc cytokines, interferons, type II cytokine receptors (eg, IL‐6), and other interleukins.5, 22 Also per the literature, tofacitinib showed comparative selectivity for JAK3, and baricitinib showed comparative selectivity for JAK2.4, 5, 9

Cytokine receptor inhibitory concentration (ICxx) profiles were generally similar among the JAK inhibitors, with small numerical differences in percentage cytokine receptor inhibition, suggesting limited differentiation of these JAK inhibitors based on in vitro pharmacology. Small differences observed included the ICxx values for IL‐6 and IL‐15 receptor inhibition. However, these differences do not appear to translate into significant differences in key clinical biomarkers. Although IL‐6 is involved in stimulating CRP production from hepatocytes,7 reduction of CRP levels from baseline to week 12 does not appear to differ by any clinically meaningful extent for these JAK inhibitors when used to treat RA: tofacitinib 5 mg BID (−10.1 mg/L23); baricitinib 4 mg QD (approximately −10 mg/L24); upadacitinib 6 mg BID (−8.8 mg/L25); and filgotinib 200 mg QD (−14.9 mg/L26). Likewise, although IL‐15 is critical for NK cell maintenance,27 reductions from baseline in NK cell counts do not appear to differ to a meaningful extent for these JAK inhibitors in RA: tofacitinib (−32.5 cells/mm3 at month 1.5, −63.5 cells/mm3 at month 6, +6.5 cells/mm3 at month 22, cross‐sectional analysis in different groups of patients4); baricitinib 4 mg QD (−57.0 cells/mm3 at week 12; −53.4 cells/mm3 at week 2428); upadacitinib 6 mg BID (approximately −50 cells/mm3 at week 1229); sufficient long‐term data are not available for filgotinib.

Tofacitinib generally demonstrated a greater relative inhibition of γc cytokine receptors vs the other JAK inhibitors. This is consistent with the selectivity of tofacitinib for JAK1 and JAK3,4, 5 compared with smaller JAK3 effects for the other JAK inhibitors.9, 10, 11 Given the association between γc cytokines and adaptive immune functions, including effects on subsets of pathologic T cells,5 one might expect to observe related clinical differences between these JAK inhibitors. It remains to be seen if the relative difference in inhibition of γc cytokine receptors vs other cytokine receptors (eg, IFN and IL‐6) with tofacitinib, compared with JAK inhibitors that spare JAK3, translates into a meaningful difference in infection risk. Meta‐analysis data suggest that the risk of serious infections is comparable for tofacitinib and baricitinib in the treatment of RA.30 Incidence rates (IRs; patients with events per 100 patient‐years) (95% confidence intervals [CI]) were: 2.7 (2.5, 3.0) for all tofacitinib doses pooled in phase 1, phase 2, phase 3, and long‐term extension (LTE) studies; and 4.8 (2.3, 9.7) for baricitinib 2 mg QD, and 3.7 (2.3, 5.8) for baricitinib 4 mg QD, in randomized controlled trials.30 Interestingly, herpes zoster (HZ), caused by varicella zoster virus reactivation,31 has been observed with each JAK inhibitor studied herein. In pooled analyses of RA clinical studies, HZ IRs (events per 100 patient‐years) were: 4.4 and 4.2 with tofacitinib 5 and 10 mg BID, respectively, vs 1.5 with placebo32; 2.7 and 4.3 with baricitinib 2‐ and 4 mg QD, respectively, vs 1.0 with placebo.33 HZ has also been reported in phase 3 RA studies of upadacitinib and filgotinib.17, 18 As such, the occurrence of HZ is likely a “class effect” of inhibiting at least JAK1,34 although viral reactivation could also be dependent on the overall impact of JAK inhibition.

Notably, IC50 values for filgotinib and its metabolite were not measurable for TPO, IL‐3, or GM‐CSF cytokine receptors (all JAK2‐dependent pathways). This may be related to the potency of filgotinib and the concentration range used in the assays. Overall, there was only minor differentiation between the effects of the JAK inhibitors on cytokine receptors mediated by JAK2/JAK2 pairs. However, tofacitinib and baricitinib appear to differentiate from each other clinically in terms of effect on platelet counts and hemoglobin levels, which could be related to the effects of JAK2 inhibition on TPO and EPO activity and production.6, 35, 36 In a pooled analysis of two LTE studies of tofacitinib 5 or 10 mg BID for RA, platelet counts initially decreased from baseline then stabilized over time, and hemoglobin levels increased from baseline over time.37 In the tofacitinib LTE study ORAL Sequel, increases in hemoglobin levels were observed from baseline to month 24, which then remained stable to month 96.38 In contrast, in an integrated analysis of phase 1b, phase 2, phase 3, and LTE RA studies, baricitinib 4 mg QD was associated with an initial increase in platelet counts, which then returned toward baseline; hemoglobin levels decreased from baseline to week 20, then returned to baseline or higher.33 Given that tofacitinib and baricitinib do not appear to differentially inhibit cytokine receptor signaling via JAK2 to a significant extent, alternative reasoning for the observed clinical difference may be important, for example, time course for inhibition. Indeed, time above IC50 may be significant.39

One observation with tofacitinib dosed QD vs BID suggests that some JAK effects may be more sensitive to daily drug holiday. In a phase 2 RA study, changes in levels of hemoglobin and neutrophils from baseline to week 24 were less pronounced with tofacitinib 20 mg QD (0.01 g/dL and −0.43 × 103/mm3, respectively) vs 10 mg BID (−0.34 g/dL and −1.20 × 103/mm3, respectively).23 While these differences are subtle, further clinical data are required to understand this phenomenon.

Lower levels of inhibition of JAK2‐dependent cytokine receptor signaling by tofacitinib, baricitinib, and upadacitinib have recently been reported,40 which contradict values reported here and in previous studies.41 However, whereas we used cytokine concentrations for all receptors that maximally induce STAT phosphorylation (>EC95), we note that McInnes et al40 utilized as much as 667‐fold lower concentrations of JAK2‐dependent cytokines. STAT phosphorylation can be more readily inhibited at lower cytokine concentrations (Figure S2), likely due to partial receptor activation, resulting in lower IC50. Physiologic cytokine concentrations vary, necessitating standardized cytokine receptor stimulation for proper comparison of inhibitor activity across different receptors. Similarly, the use of physiologic ATP concentration is important when comparing the inhibition potential of ATP‐competitive JAK inhibitors.42 The potency of upadacitinib against JAK enzymes measured in this study differs from those recently reported in Parmentier et al,43 which likely reflects differences in ATP concentrations used between studies.

Limitations of this analysis must be considered. The comparisons were based on doses of four JAK inhibitors which were considered to provide generally comparable clinical efficacy in patients with RA; these agents (and doses) have not all received regulatory approval. Furthermore, this analysis was limited to an extensive, but incomplete, list of cytokines; nevertheless, those evaluated represent key family members associated with each JAK pair, so the results are somewhat generalizable. While this analysis determined cytokine receptor inhibition profiles, the clinical impact of JAK selectivity could vary depending on factors such as patients' genetics or underlying inflammatory state (ie, comorbid diseases)8 as well as environmental factors or exogenous influences (eg, concomitant medications).

Research into inhibitors of alternative inflammatory pathways remains important. Indeed, second‐generation JAK inhibitors, preferentially selective for one JAK isoform, are being developed.6 For example, BMS‑986165 is a TYK2‐selective inhibitor under investigation for psoriasis44; and PF‐06651600 is a dual JAK3/TEC kinase‐selective inhibitor45 under investigation for alopecia (NCT02974868), Crohn's disease (NCT03395184), non‐segmental vitiligo (NCT03715829), RA (NCT02969044), and ulcerative colitis (NCT02958865). Possible differences in clinical profiles of compounds selective for TYK2 or JAK3 vs those selective for JAK1 are evidenced in early study data, although further research in more robust settings is required. For example, BMS‑986165 has not been associated with laboratory changes such as increased lipid levels,44 which can occur with IL‐6 inhibition.46

In conclusion, by applying an integrated modeling approach, the JAK inhibitors tofacitinib, baricitinib, upadacitinib, and filgotinib, at doses conveying reasonably equivalent clinical efficacy for RA, exhibited generally similar cytokine receptor inhibition profiles. Although some small numerical differences were observed, these do not appear to translate to significant differences in the JAK inhibitors' clinical profiles. At the same time, we appreciate that only robust clinical testing involving head‐to‐head studies may determine whether there are clinically meaningful differences between these JAK inhibitors. All JAK inhibitors, including novel second‐generation compounds with minimal JAK1 effects, need to be characterized via thorough preclinical, metabolic and pharmacological evaluation, adequate long‐term clinical data, and when available, real‐world experience.

DISCLOSURES

All authors are employees and shareholders of Pfizer Inc. The authors report that this research did not receive external public or private foundation funding. The study was sponsored by Pfizer Inc.

AUTHOR CONTRIBUTIONS

Participated in research design: Dowty, Lin, Jesson, Martin, Katkade, and Telliez.

Performed experiments: Dowty and Lin.

Performed data analysis: Dowty, Lin, Jesson, Hegen, Martin, Katkade, Menon, and Telliez.

Wrote or contributed to the writing of the manuscript: Dowty, Lin, Jesson, Hegen, Martin, Katkade, Menon, and Telliez.

Supporting information

 

 

 

ACKNOWLEDGMENTS

This study was sponsored by Pfizer Inc. Medical writing support, under the guidance of the authors, was provided by Sarah Piggott, MChem, of CMC Connect, a division of McCann Health Medical Communications Ltd, Glasgow, UK, and was funded by Pfizer Inc in accordance with Good Publication Practice (GPP3) guidelines (Ann Intern Med 2015;163:461‐464).

Dowty ME, Lin TH, Jesson MI, et al. Janus kinase inhibitors for the treatment of rheumatoid arthritis demonstrate similar profiles of in vitro cytokine receptor inhibition. Pharmacol Res Perspect. 2019;e00537 10.1002/prp2.537

REFERENCES

  • 1. Kuek A, Hazleman BL, Ostör AJ. Immune‐mediated inflammatory diseases (IMIDs) and biologic therapy: a medical revolution. Postgrad Med J. 2007;83:251‐260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. El‐Gabalawy H, Guenther LC, Bernstein CN. Epidemiology of immune‐mediated inflammatory diseases: incidence, prevalence, natural history, and comorbidities. J Rheumatol Suppl. 2010;85:2‐10. [DOI] [PubMed] [Google Scholar]
  • 3. Jacobs P, Bissonnette R, Guenther LC. Socioeconomic burden of immune‐mediated inflammatory diseases–focusing on work productivity and disability. J Rheumatol Suppl. 2011;88:55‐61. [DOI] [PubMed] [Google Scholar]
  • 4. Hodge JA, Kawabata TT, Krishnaswami S, et al. The mechanism of action of tofacitinib ‐ an oral Janus kinase inhibitor for the treatment of rheumatoid arthritis. Clin Exp Rheumatol. 2016;34:318‐328. [PubMed] [Google Scholar]
  • 5. Clark JD, Flanagan ME, Telliez JB. Discovery and development of Janus kinase (JAK) inhibitors for inflammatory diseases. J Med Chem. 2014;57:5023‐5038. [DOI] [PubMed] [Google Scholar]
  • 6. Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, O'Shea JJ. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov. 2017;17:78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Srirangan S, Choy EH. The role of interleukin 6 in the pathophysiology of rheumatoid arthritis. Ther Adv Musculoskelet Dis. 2010;2:247‐256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Choy EH. Clinical significance of Janus kinase inhibitor selectivity. Rheumatology (Oxford). 2018;58:953‐962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Shi JG, Chen X, Lee F, et al. The pharmacokinetics, pharmacodynamics, and safety of baricitinib, an oral JAK 1/2 inhibitor, in healthy volunteers. J Clin Pharmacol. 2014;54:1354‐1361. [DOI] [PubMed] [Google Scholar]
  • 10. Klünder B, Mohamed MF, Othman AA. Population pharmacokinetics of upadacitinib in healthy subjects and subjects with rheumatoid arthritis: analyses of phase I and II clinical trials. Clin Pharmacokinet. 2018;57:977‐988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Namour F, Diderichsen PM, Cox E, et al. Pharmacokinetics and pharmacokinetic/pharmacodynamic modeling of filgotinib (GLPG0634), a selective JAK1 inhibitor, in support of Phase IIb dose selection. Clin Pharmacokinet. 2015;54:859‐874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Galien R, Vayssière B, de Vos S, et al. Analysis of the JAK1 selectivity of GLPG0634 and its main metabolite in different species, healthy volunteers and rheumatoid arthritis patients. Arthritis Rheum. 2013;65(suppl 10). Abstract 478. Available at: https://acrabstracts.org/abstract/analysis-of-the-jak1-selectivity-of-glpg0634-and-its-main-metabolite-in-different-species-healthy-volunteers-and-rheumatoid-arthritis-patients/. [Google Scholar]
  • 13. Elwood F, Witter DJ, Piesvaux J, et al. Evaluation of JAK3 biology in autoimmune disease using a highly selective, irreversible JAK3 inhibitor. J Pharmacol Exp Ther. 2017;361:229‐244. [DOI] [PubMed] [Google Scholar]
  • 14. Riccardi K, Lin J, Li Z, et al. Novel method to predict in vivo liver‐to‐plasma Kpuu for OATP substrates using suspension hepatocytes. Drug Metab Dispos. 2017;45:576‐580. [DOI] [PubMed] [Google Scholar]
  • 15. US Food and Drug Administration . XELJANZ® (tofacitinib) highlights of prescribing information. 2017. http://labeling.pfizer.com/ShowLabeling.aspx?xml:id=959. Accessed June 4, 2018.
  • 16. US Food and Drug Administration . Olumiant (baricitinib): highlights of prescribing information. 2018. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/207924s000lbl.pdf. Accessed June 5, 2019.
  • 17. Genovese MC, Kalunian KC, Walker D, et al. Safety and efficacy of filgotinib in a Phase 3 trial of patients with active rheumatoid arthritis and inadequate response or intolerance to biologic DMARDs. Arthritis Rheumatol. 2018;70 (suppl 10). Abstract L06 Available at: https://acrabstracts.org/abstract/safety-and-efficacy-of-filgotinib-in-a-phase-3-trial-of-patients-with-active-rheumatoid-arthritis-and-inadequate-response-or-intolerance-to-biologic-dmards/. [Google Scholar]
  • 18. 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;391:2513‐2524. [DOI] [PubMed] [Google Scholar]
  • 19. Dowty ME, Jesson MI, Ghosh S, et al. Preclinical to clinical translation of tofacitinib, a Janus kinase inhibitor, in rheumatoid arthritis. J Pharmacol Exp Ther. 2014;348:165‐173. [DOI] [PubMed] [Google Scholar]
  • 20. Lamba M, Hutmacher MM, Furst DE, et al. Model‐informed development and registration of a once‐daily regimen of extended‐release tofacitinib. Clin Pharmacol Ther. 2017;101:745‐753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Vanhoutte F, Mazur M, Van der Aa A, Wigerinck P, van't Klooster G. Selective JAK1 inhibition in the treatment of rheumatoid arthritis: proof of concept with GLPG0634. Arthritis Rheum. 2012;64(suppl 10). Abstract 2489. Available at: https://acrabstracts.org/abstract/selective-jak1-inhibition-in-the-treatment-of-rheumatoid-arthritis-proof-of-concept-with-glpg0634/. [Google Scholar]
  • 22. O'Sullivan LA, Liongue C, Lewis RS, Stephenson SE, Ward AC. Cytokine receptor signaling through the Jak‐Stat‐Socs pathway in disease. Mol Immunol. 2007;44:2497‐2506. [DOI] [PubMed] [Google Scholar]
  • 23. Kremer JM, Cohen S, Wilkinson BE, et al. A phase IIb dose‐ranging study of the oral JAK inhibitor tofacitinib (CP‐690,550) versus placebo in combination with background methotrexate in patients with active rheumatoid arthritis and an inadequate response to methotrexate alone. Arthritis Rheum. 2012;64:970‐981. [DOI] [PubMed] [Google Scholar]
  • 24. Genovese MC, Kremer J, Zamani O, et al. Baricitinib in patients with refractory rheumatoid arthritis. N Engl J Med. 2016;374:1243‐1252. [DOI] [PubMed] [Google Scholar]
  • 25. 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;68:2857‐2866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. 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;76:1009‐1019. [DOI] [PubMed] [Google Scholar]
  • 27. Fehniger TA, Caligiuri MA. Interleukin 15: biology and relevance to human disease. Blood. 2001;97:14‐32. [DOI] [PubMed] [Google Scholar]
  • 28. Emery P, McInnes I, Genovese MC, et al. A7.16 characterisation of changes in lymphocyte subsets in baricitinib‐treated patients with rheumatoid arthritis in two phase 3 studies. Ann Rheum Dis. 2016;75:A62 Available at: https://ard.bmj.com/content/75/Suppl_1/A62.1. [Google Scholar]
  • 29. 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;68:2867‐2877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Strand V, Ahadieh S, DeMasi R, et al. THU0211 Meta‐analysis of serious infections with baricitinib, tofacitinib and biologic DMARDs in rheumatoid arthritis. Ann Rheum Dis. 2017;76:284 Available at: https://ard.bmj.com/content/76/Suppl_2/284.1. [Google Scholar]
  • 31. Zerboni L, Sen N, Oliver SL, Arvin AM. Molecular mechanisms of varicella zoster virus pathogenesis. Nat Rev Microbiol. 2014;12:197‐210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Winthrop KL, Yamanaka H, Valdez H, et al. Herpes zoster and tofacitinib therapy in patients with rheumatoid arthritis. Arthritis Rheumatol. 2014;66(suppl 10):2675‐2684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Smolen JS, Genovese MC, Takeuchi T, et al. Safety profile of baricitinib in patients with active rheumatoid arthritis with over 2 years median time in treatment. J Rheumatol. 2019;46:7‐18. [DOI] [PubMed] [Google Scholar]
  • 34. Winthrop KL. The emerging safety profile of JAK inhibitors in rheumatic disease. Nat Rev Rheumatol. 2017;13:320. [DOI] [PubMed] [Google Scholar]
  • 35. Skoda RC. Less Jak2 makes more platelets. Blood. 2014;124:2168‐2169. [DOI] [PubMed] [Google Scholar]
  • 36. Grozovsky R, Begonja AJ, Liu K, et al. The Ashwell‐Morell receptor regulates hepatic thrombopoietin production via JAK2‐STAT3 signaling. Nat Med. 2015;21:47‐54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Wollenhaupt J, Silverfield J, Lee EB, et al. Safety and efficacy of tofacitinib, an oral Janus kinase inhibitor, for the treatment of rheumatoid arthritis in open‐label, longterm extension studies. J Rheumatol. 2014;41:837‐852. [DOI] [PubMed] [Google Scholar]
  • 38. Wollenhaupt J, Lee EB, Curtis JR, et al. Safety and efficacy of tofacitinib for up to 9.5 years in the treatment of rheumatoid arthritis: final results of a global, open‐label, long‐term extension study. Arthritis Res Ther. 2019;21:89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. McInnes IB, Higgs R, Lee J, et al. Ex vivo comparison of baricitinib, upadacitinib, filgotinib, and tofacitinib for cytokine signaling in human leukocyte subpopulations. Arthritis Rheumatol. 2017;69(suppl 10). Available at: https://acrabstracts.org/abstract/ex-vivo-comparison-of-baricitinib-upadacitinib-filgotinib-and-tofacitinib-for-cytokine-signaling-in-human-leukocyte-subpopulations. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. McInnes IB, Byers NL, Higgs RE, et al. Comparison of baricitinib, upadacitinib, and tofacitinib mediated regulation of cytokine signaling in human leukocyte subpopulations. Arthritis Res Ther. 2019;21:183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Meyer DM, Jesson MI, Li X, et al. Anti‐inflammatory activity and neutrophil reductions mediated by the JAK1/JAK3 inhibitor, CP‐690,550, in rat adjuvant‐induced arthritis. J Inflamm. 2010;7:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Thorarensen A, Banker ME, Fensome A, et al. ATP‐mediated kinome selectivity: the missing link in understanding the contribution of individual JAK Kinase isoforms to cellular signaling. ACS Chem Biol. 2014;9:1552‐1558. [DOI] [PubMed] [Google Scholar]
  • 43. Parmentier JM, Voss J, Graff C, et al. In vitro and in vivo characterization of the JAK1 selectivity of upadacitinib (ABT‐494). BMC Rheumatol. 2018;2:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Papp K, Gordon K, Thaçi D, et al. Phase 2 trial of selective tyrosine kinase 2 inhibition in psoriasis. N Engl J Med. 2018;379:1313‐1321. [DOI] [PubMed] [Google Scholar]
  • 45. Xu H, Jesson MI, Seneviratne UI, et al. PF‐06651600, a dual JAK3/TEC family kinase inhibitor. ACS Chem Biol. 2019;14:1235‐1242. [DOI] [PubMed] [Google Scholar]
  • 46. Hashizume M, Masahiko M. IL‐6 and lipid metabolism. Inflamm Regen. 2011;31:325‐333. [Google Scholar]
  • 47. Mohamed MF, Camp HS, Jiang P, Padley RJ, Asatryan A, Othman AA. Pharmacokinetics, safety and tolerability of ABT‐494, a novel selective JAK 1 inhibitor, in healthy volunteers and subjects with rheumatoid arthritis. Clin Pharmacokinet. 2016;55:1547‐1558. [DOI] [PubMed] [Google Scholar]
  • 48. Mohamed MF, Zeng J, Song I‐H, Othman AA. THU0177 ABT‐494 pharmacokinetics following administration of the once‐daily extended‐release tablet formulation being utilized in the ongoing rheumatoid arthritis phase 3 trials. Ann Rheum Dis. 2017;76(suppl 2):268‐269. Available at: https://ard.bmj.com/content/76/Suppl_2/268.3. [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

 

 

 


Articles from Pharmacology Research & Perspectives are provided here courtesy of Wiley

RESOURCES