You are on page 1of 15

Drugs

DOI 10.1007/s40265-017-0835-9

ADIS DRUG EVALUATION

Tofacitinib: A Review in Rheumatoid Arthritis


Sohita Dhillon1

Ó Springer International Publishing AG 2017

Abstract Tofacitinib (XeljanzÒ) is a potent, selective JAK common adverse events (AEs) in tofacitinib recipients.
inhibitor that preferentially inhibits Janus kinase (JAK) 1 However, the incidence of herpes zoster (HZ) was higher
and JAK3. In the EU, oral tofacitinib 5 mg twice daily is with tofacitinib than in the general RA population,
indicated for the treatment of moderate to severe active although infections were clinically manageable. When
rheumatoid arthritis (RA) in adult patients who have added to background methotrexate, tofacitinib was nonin-
responded inadequately to, or who are intolerant of, one or ferior to adalimumab in terms of efficacy, and both com-
more DMARDs. Several clinical studies of B 24 months’ bination therapies had generally similar tolerability
duration showed that tofacitinib monotherapy (as first- or profiles. Although additional comparative studies are nee-
second-line treatment) and combination therapy with a ded to more definitively position tofacitinib relative to
conventional synthetic DMARD (csDMARD; as second- bDMARDs and other targeted synthetic DMARDs, current
or third-line treatment) was effective in reducing signs and evidence indicates that oral tofacitinib is a useful option for
symptoms of disease and improving health-related quality the treatment of patients with RA.
of life (HR-QOL), with benefits sustained during long-term
therapy (B 96 months). Tofacitinib monotherapy inhibited
progression of structural damage in methotrexate-naı̈ve
Tofacitinib: clinical considerations in RA
patients during B 24 months’ treatment, with beneficial
effects also seen in patients receiving tofacitinib plus
Modulates immune and inflammatory responses by
methotrexate as second-line therapy for 12 months.
inhibiting the JAK family of kinases, preferentially
Tofacitinib was generally well tolerated during
JAK1 and JAK3
B 114 months’ treatment, with most adverse events of mild
or moderate severity. The tolerability profile of tofacitinib Effective in methotrexate-naı̈ve and DMARD-
was generally similar to that of biological DMARDs experienced patients, including those with inadequate
(bDMARDs), with infections and infestations the most response to TNF inhibitors
Reduces signs and symptoms of disease and
The manuscript was reviewed by: R. M. Fleischmann, Metroplex improves HR-QOL as monotherapy and in
Clinical Research Center, University of Texas Southwestern Medical combination with csDMARDs, with benefits
Center, Dallas, TX, USA; E. B. Lee, Division of Rheumatology,
Department of Internal Medicine, Seoul National University College
sustained during long-term therapy
of Medicine, Seoul, Republic of Korea; D. L Scott, Department of Has beneficial effects on radiographic progression
Clinical Rheumatology, Kings College London, Weston Education
Centre, London, UK. Most common AEs are infections/infestations,
generally of mild or moderate severity; incidence of
& Sohita Dhillon
demail@springer.com
HZ is higher than in the general RA population

1
Springer, Private Bag 65901, Mairangi Bay, Auckland 0754,
New Zealand
S. Dhillon

1 Introduction 34.0 nmol/L) [7]. Tofacitinib had limited selectivity for


other kinases in the human genome [8]. In cellular assays,
Rheumatoid arthritis (RA) is a chronic, progressive, tofacitinib preferentially inhibited signalling by hetero-
autoimmune disease characterized by inflammatory syn- dimeric cytokine receptors that associate with JAK3 and/or
ovitis and articular destruction. Conventional synthetic JAK1 [5, 7]. Preclinical studies showed that tofacitinib
disease-modifying antirheumatic drugs (csDMARDs; e.g. attenuated JAK1/JAK3-mediated signalling of IL-2, -4, -6,
methotrexate, sulfasalazine) and biological DMARDs -7, -15 and -21, as well as IFNa and IFNc, resulting in the
(bDMARDs; TNF inhibitors, IL-6 inhibitors) have been the modulation of immune and inflammatory responses [7, 9].
mainstay of disease management in patients with RA [1]. For instance, in vitro and in vivo assays of samples from
However, despite various treatment modalities and the RA patients showed that tofacitinib dose-dependently
recommended treat-to-target approach for disease man- inhibited IL-17 and IFNc production by, and proliferation
agement, many patients still do not achieve therapeutic of, synovial and peripheral blood CD4? T cells, resulting
targets, indicating an unmet need for additional therapies in the inhibition of IL-6 production in synovial fibroblasts,
[2, 3]. Recent advances in the understanding of interactions inhibition of IL-8 production in CD14? monocytes and
between immune receptors and downstream signalling decreased cartilage damage [9].
pathways have led to the development of small-molecule Tofacitinib rapidly reduced C-reactive protein (CRP)
antirheumatic agents targeting key nodes in these pathways levels in patients with RA, with these reductions main-
[1]. The Janus kinase (JAK) family of tyrosine kinases are tained throughout therapy [5]. Treatment discontinuation
critical for the signalling of cytokines that bind to type I for 2 weeks did not completely reverse the change in CRP
and II cytokine receptors, including cytokines responsible levels, suggesting that tofacitinib has a longer duration of
for driving inflammatory processes implicated in the pharmacodynamic activity than its half-life [5]. In RA
pathogenesis of RA (e.g. type I interferons and several patients with inadequate response to methotrexate, tofaci-
interleukins) [1, 4]. JAKs were therefore considered logical tinib reduced the synovial expression of key genes impli-
targets for pharmacological manipulation in inflammatory cated in the pathogenesis of RA (e.g. matrix
diseases, leading to the development of JAK inhibitors. metalloproteinases 1 and 3, and chemokines CCL2,
Tofacitinib (XeljanzÒ) is one such JAK inhibitor approved CXCL10 and CXCL13), and reduced synovial STAT1 and
in several countries worldwide, and recently in the EU, for STAT3 phosphorylation (which correlated with clinical
the treatment of patients with RA. This narrative review is improvement), suggesting that IFN and IL-6 signalling
written from the EU perspective [5], focusing on the play a key role in the synovial response to tofacitinib-
therapeutic efficacy and tolerability of tofacitinib in mediated JAK blockade [10]. Tofacitinib (with or without
patients with moderate to severe active RA and summa- methotrexate) also reduced bone marrow oedema and
rizing relevant pharmacological data. inhibited progression of structural damage in methotrexate-
naı̈ve RA patients (as assessed by MRI endpoints) [11]. In
addition, studies in patients with RA and in rodent models
2 Pharmacodynamic Properties of Tofacitinib of RA suggested that tofacitinib reduced structural damage
to the arthritic joints by suppressing osteoclast-mediated
The pharmacological properties of tofacitinib have been bone resorption through decreased RANKL production
reviewed in detail previously [4, 6]; therefore, a brief [12, 13].
overview is presented here. Tofacitinib is a potent, selec- During B 6 months’ therapy, tofacitinib dose-depen-
tive inhibitor of the JAK family of tyrosine kinases [7, 8], dently decreased NK cell counts (by median 35%) and
comprising four non-receptor tyrosine kinases, JAK1, increased B cell counts (by median & 30%) from baseline
JAK2, JAK3 and tyrosine kinase 2 (TYK2), with each [14]. By contrast, during long-term therapy (median
having specificity for a different set of cytokine receptors exposure 5 years), NK cell counts increased (by med-
[4]. Cytokine binding to its receptor activates receptor-as- ian & 73%) from baseline, but there was no further
sociated JAKs, which act as docking sites for signal increase in B cell counts (median increase from base-
transducers and activators of transcription (STATs) [4]. line & 3%) [14]. Changes in CD3?, CD4? and CD8? T
STATs, after being phosphorylated by the receptor-asso- cells correlated with changes in absolute lymphocyte
ciated JAKs, dissociate from the receptor units, dimerise counts (ALC; Sect. 5.2). Importantly, changes in lympho-
with each other and translocate to the cell nucleus where cyte subset counts were not related to the occurrence of
they regulate gene transcription [4]. In in vitro enzymatic serious or opportunistic infections or herpes zoster (HZ)
assays, tofacitinib potently inhibited JAK1, JAK2 and [5], and were reversed following temporary treatment
JAK3 [half maximal inhibitory concentrations (IC50) discontinuation [14]. Tofacitinib was associated with dose-
1.6–3.2 nmol/L] and, to a lesser degree, TYK2 (IC50 dependent and moderate increases in lipid parameters (e.g.
Tofacitinib: A Review

15–20% increase in LDL-C and HDL-C) during [18, 19, 21–24] or 2010 revised criteria (C 4 tender or
B 24 months’ therapy, with maximum effects seen within painful joints, C 4 swollen joints, CRP of C 3 mg/L and
6 weeks and levels remaining generally stable thereafter; class I–III functional capacity) [20]. Two studies assessed
mean LDL-C/HDL-C and ApoB/ApoA1 ratios were gen- radiographic outcomes and required that patients addi-
erally unchanged [5]. Statins (e.g. atorvastatin) rapidly tionally have C 3 distinct joint erosions on hand and wrist
(within 4–6 weeks) reduced tofacitinib-associated increa- or foot radiographs, or a positive test for IgM rheumatoid
ses in total cholesterol, LDL-C, triglyceride and ApoB factor (RF) or antibodies to citrullinated peptide (anti-CCP)
levels [15]. Small, reversible increases in mean serum [18, 21]. Patients included had a mean age of approxi-
creatinine levels have also been seen with tofacitinib mately 50–55 years and were largely female (79–87%).
therapy, which plateaued early and did not appear to be This section focuses on results relevant to the approved
associated with progressive worsening of renal function or dosage of tofacitinib 5 mg twice daily. With the exception
acute renal failure [16]. of one study [20], all studies included a tofacitinib 10 mg
Tofacitinib may diminish the immunogenicity of coad- twice daily study arm, results for which are presented in the
ministered vaccines (e.g. the 23-valent pneumococcal table for completeness, but not discussed further. Copri-
polysaccharide vaccine, particularly when used in combi- mary endpoints were assessed sequentially, using a step-
nation with methotrexate [17]); therefore, it is recom- down approach to assign significance.
mended that patients be brought up to date with all
immunisations prior to initiating tofacitinib therapy and 4.1 As Combination Therapy After Inadequate
live vaccines not be coadministered with tofacitinib [5]. Response to cs/bDMARDs

The efficacy of tofacitinib compared with placebo and/or


3 Pharmacokinetic Properties of Tofacitinib adalimumab (as combination therapy with DMARDs) was
assessed in the ORAL Scan, ORAL Standard, ORAL Sync,
Tofacitinib is rapidly absorbed, with peak plasma con- ORAL Step and ORAL Strategy in patients with RA who
centrations reached within 0.5–1 h after administration; had an inadequate response to methotrexate [20–22], C 1
steady-state is reached in 24–48 h and there is negligible cs/bDMARD [23] or C 1 TNF inhibitor [24] (Table 1). The
accumulation after twice daily dosing [5]. The oral mean disease duration ranged between 5 and 13 years.
bioavailability of tofacitinib is 74% and its volume of
distribution after intravenous administration is 87 L. 4.1.1 Compared with Placebo
Approximately 40% of tofacitinib is bound to plasma
proteins (largely albumin). Clearance of tofacitinib is via 4.1.1.1 Clinical Responses The addition of tofacitinib
hepatic metabolism (70%) and renal excretion (30%). 5 mg twice daily to a stable dose of methotrexate or C 1
Tofacitinib is metabolised primarily by CYP3A4 and to a csDMARD was more effective in improving signs and
minor extent by CYP2C19; 35% of a tofacitinib dose is symptoms of disease than the addition of placebo to
excreted as eight metabolites that are 10-fold less potent background therapy (Table 1) [21–24]. At month 3 in the
than tofacitinib, and 65% of the dose is excreted as ORAL Step study and at month 6 in the ORAL Scan,
unchanged drug. Tofacitinib is rapidly eliminated with a ORAL Standard and ORAL Sync studies, significantly
half-life of & 3 h [5]. more patients in the tofacitinib than placebo groups
achieved ACR20 (coprimary endpoint) ACR50 or ACR70
responses (Table 1) [21–24]. Tofacitinib had a rapid onset
4 Therapeutic Efficacy of Tofacitinib of action, as indicated by significantly higher ACR20,
ACR50 and ACR70 response rates within 2–4 weeks of
The therapeutic efficacy of oral tofacitinib as monotherapy therapy (p\0.05 vs. placebo [22, 24] or baseline [21, 23]).
[18–20] or combination therapy [20–24] in patients with Moreover, in a post hoc analysis of the ORAL Scan study,
RA was assessed in several 6- to 24-month, randomized, ACR20, ACR50 and ACR70 response rates at month 6
double-blind, multicentre, phase 3 or 3b/4 ORAL studies. were significantly (p\0.05) higher in the tofacitinib group
Patients included were either naı̈ve to methotrexate therapy than in the placebo group regardless of the background
[18] or had an inadequate response to C 1 cs/bDMARD dose of methotrexate [low (B 12.5 mg/week), moderate
[19–23] or C 1 TNF-inhibitor [24]. They had to be ([12.5 to \17.5 mg/week) or high (C 17.5 mg/week)]
C 18 years old and have active RA according to the [25].
American College of Rheumatology (ACR) 1987 revised In the ORAL Scan study, owing to the step-down
criteria (C 6 tender or painful joints, C 6 swollen joints and assessment of coprimary endpoints, and as no statistically
either an ESR of [28 mm/h or CRP of [7 mg/L) significant between-group difference was seen for
S. Dhillon

Table 1 Efficacy of oral tofacitinib 5 or 10 mg twice daily in rheumatoid arthritis patients with inadequate response to cs/bDMARDs in
6-[19, 24], 12-[20, 22, 23] or 24- [21] month, randomized, double-blind, multicentre, phase 3 [19, 21–24] or 3b/4 [20] studies
Study (prior therapy) Timept Treatment (no. of ptsc) Response ratesa (% pts) Remissiona,b LDAa,b Change from BL
(months)
ACR20 ACR50 ACR70 (% pts) (% pts) HAQ-DI [BL]d

ORAL Scan [21] 6 TOF 5 ? MTXe (321) 51.5***f 32.4*** 14.6*** 7.2f,g 14.3*** - 0.40f,g,h [1.4]
(MTX-IR) TOF 10 ? MTX (316) e
61.8*** f
43.7*** 22.3*** 16.0*** f
28.4*** – 0.54***f,h [1.4]
PL ? MTXi,e (160) 25.3f 8.4 1.3 1.6f 3.1 - 0.15f,h [1.3]
ORAL Standard [22] 6 TOF 5 ? MTXe (204) 51.5***f 37*j 20*j 6.2*f - 0.55***f,h [1.5]
(MTX-IR) TOF 10 ? MTX (201) e
52.6*** f
35* j
22* j
12.5*** f
– 0.61***f,h [1.5]
e f j j f
ADA ? MTX (204) 47.2*** 29* 10* 6.7* - 0.49***f,h [1.5]
i,e j j
PL ? MTX (108) 28.3 12 2.5 1.1 - 0.24f,h [1.5]
ORAL Sync [5, 23] 6 TOF 5 ? csDMARDe (315) 52.1***f 34*** 13*** 8.5**f - 0.44***f,h [1.4]
(cs/bDMARD-IR) TOF 10 ? csDMARDe (318) 56.6***f 36*** 16*** 12.5***f – 0.53***f,h [1.4]
PL ? csDMARDi,e (159) 30.8f 12.5 3.1 2.6f - 0.16f,h [1.5]
ORAL Step [24] 3 TOF 5 ? MTXe (133) 41.7**f 26.5*** 13.6*** 6.7*f 14.3* - 0.43***f [1.6]
(TNFi-IR) TOF 10 ? MTX (134) e
48.1*** f
27.8*** 10.5** 8.8* f
20.8*** – 0.46***f [1.5]
i,e f f
PL ? MTX (132) 24.4 8.4 1.5 1.7 5.0 - 0.18f [1.6]
ORAL Strategy 6 TOF 5 (384) 65 38f 18 10 21 - 0.52j [1.6]
[20] (MTX-IR) TOF 5 ? MTXe (376) 73 46f,k 25 12 27 - 0.58j [1.6]
e f,k
ADA ? MTX (386) 71 44 21 12 27 - 0.54j [1.6]
12 TOF 5 62 39 21 11 23 - 0.57j
e
TOF 5 ? MTX 70 48 29 15 27 - 0.62j
ADA ? MTXe 68 46 16 17 33 - 0.63j
ORAL Solo [19] 3 TOF 5 (243) 59.8***f 31.1*** 15.4** 5.6f 12.5* - 0.5***f [1.5]
(cs/bDMARD-IR) TOF 10 (245) 65.7***f 36.8*** 20.3*** 8.7f 17.0*** – 0.57***f [1.5]
i f f
PL (122) 26.7 12.5 5.8 4.4 5.3 - 0.2f [1.5]

ACR ‘x’ C x% improvement in ACR criteria, ADA subcutaneous adalimumab 40 mg every two weeks, bid twice daily, BL baseline, DAS28-ESR Disease Activity Score
28-erythrocyte sedimentation rate, cs/bDMARD conventional synthetic/biological disease modifying anti-rheumatic drug(s), HAQ-DI Health Assessment Questionnaire-
Disability Index score (lower scores indicate less disability), IR inadequate response, LDA low disease activity, MTX methotrexate, PL placebo, pts patients, timept timepoint,
TNFi TNF-inhibitors, TOF tofacitinib
*p B 0.05, **p B 0.01, ***p\0.001
a
Missing values imputed using nonresponse imputation
b
Proportion of pts in remission (DAS28-ESR of \2.6) or with LDA (DAS28-ESR of B 3.2 [19, 21, 24] or \3.2 [20])
c
Full analysis set [20–24] or modified intent-to-treat population [19]
d
Least-squares mean changes from BL; all BL values are means
e
Stable doses of MTX 15–25 mg/week [20, 21] or 7.5–25 mg/week [22, 24], stable doses of DMARDs (e.g. B 25 mg/week of MTX) [23]
f
Primary [20] or coprimary [19, 21–24] endpoints; significance of coprimary endpoints was assessed using a step-down approach
g
Owing to the step-down assessment of coprimary endpoints, and as no significant between-group difference was seen for radiographic progression, significance was not
declared
h
Assessed at month 3
i
In ORAL Solo and Step, at 3 months, PL recipients were switched to TOF 5 or 10 mg bid [19, 24]. In ORAL Scan, Standard and Sync, PL recipients who did not have C 20%
improvement in swollen and tender joint counts at 3 months were switched to TOF 5 or 10 mg bid; the remaining PL recipients switched to TOF 5 or 10 mg bid at 6 months
[21–23]
j
Values estimated from graphs
k
TOF ? MTX was noninferior to ADA ? MTX

radiographic progression [as assessed by the modified total DAS28-ESR remission and low disease activity rates at
Sharp score (mTSS); Sect. 4.1.1.2], significance was not month 3 in the ORAL Step study were significantly higher
declared for the proportion of patients with DAS28-ESR in the tofacitinib groups than in the placebo groups
remission (score of \2.6) at month 6 (coprimary endpoint; (Table 1) [22–24]. Patients in the tofacitinib group also had
Table 1) [21]. However, the rate of DAS28-ESR low dis- significantly (p B 0.05 vs. placebo [21, 22, 24] or baseline
ease activity (score of B 3.2) was significantly higher ([4- [23]) greater improvements in the DAS-ESR at month 3
fold) in the tofacitinib group than in the placebo group [24] or 6 [21–23] in the four studies.
(Table 1) [21]. The DAS28-ESR remission rates at month Where reported, ACR20, ACR50 and ACR70 response
6 in the ORAL Standard and ORAL Sync studies, and the rates [21–24], DAS28-ESR remission and low disease
Tofacitinib: A Review

Table 2 Radiographic outcomes with oral tofacitinib 5 or 10 mg twice daily in randomized, double-blind, multicentre, phase 3 studies in
patients with rheumatoid arthritis who had inadequate response to methotrexate [21] or who were methotrexate naı̈ve [18]

Study Treatment (no. of ITT pts) Timepoint LS mean change from BL in scores No radiographic
(months) progressiona (% pts)
mTSS (BL) Erosion (BL) JSN (BL)

ORAL Scan [21] TOF 5 ? MTX (321) 6 0.12b (31.1) 0.07c (13.8) 0.05c (17.3) 88.8**
b c
TOF 10 ? MTX (316) 0.06* (37.3) 0.02 (17.7) 0.02c (19.6) 86.9*
PL ? MTX (160) 0.47b (32.6) 0.17c (14.4) 0.32c (18.2) 77.7
TOF 5 ? MTX 12 0.29 0.18c 0.1*c 86.0**
TOF 10 ? MTX 0.05** 0.0c 0.03*c 86.4**
c
PL ? MTX 0.92 0.29 0.61c 74.1
ORAL Start [18] TOF 5 (373) 6 0.2***b (19.1) 0.1* (9.1) 0.1* (10.0) 87.1***
TOF 10 (397) \0.1***b (17.9) 0.1*** (9.1) 0.1* (8.8) 89.3***
MTX (186) 0.8b (16.1) 0.4 (8.4) 0.3 (7.7) 73.7
TOF 5 24 0.6*** 0.2*** 0.4* 79.9***
TOF 10 0.3*** 0.2*** 0.1*** 83.7***
MTX 2.1 1.0 1.1 64.9
MTX stable dose of 15–25 mg/week [21] or target-dose after titration of 20 mg/week [18]
BL baseline, JSN joint-space narrowing, LS least-squares, PL placebo, pts patients, mTSS van der Heijde modified total Sharp score, MTX
methotrexate, TOF tofacitinib
*p B 0.05, **p\0.01, ***p\0.001
a
Proportion of patients with a B 0.5 unit increase from BL in mTSS
b
Coprimary endpoints; significance assessed using a step-down approach
c
Values estimated from graphs

activity rates [21, 23, 24] and the change from baseline in in terms of the mTSS (B 0.5 unit increase from baseline) at
DAS28-ESR [21–24] were maintained or improved during months 6 and 12 (Table 2) and no radiographic progression
B 12 months’ treatment with tofacitinib plus background in terms of the erosion score (B 0.5 unit increase from
DMARD. baseline) at month 12 (92.0 vs. 83.5%; p B 0.05). More-
over, post hoc analyses in subgroups of patients with
4.1.1.2 Radiographic Responses At month 6 in ORAL prognostic factors predictive of greater progression of
Scan, there was no significant difference in the least square structural joint damage (i.e. anti-CCP positive, DAS28-
(LS) mean change from baseline in the mTSS between ESR[5.1, anti-CCP and/or RF positive with erosion score
patients receiving tofacitinib 5 mg twice daily and those C 3, baseline mTSS greater than baseline median mTSS
receiving placebo, in addition to background methotrexate value) showed greater benefits with tofacitinib than pla-
therapy (coprimary endpoint; Table 2) [21]. However, cebo at 6 and 12 months (95% CIs generally excluded ‘0’)
when results were analysed using a trimmed analysis (to [21].
assess the effect of outliers), a significant (p\0.05) benefit
of tofacitinib over placebo was seen both at months 6 and 4.1.1.3 Health-Related Quality of Life In the ORAL
12 (e.g. at 1% trimming the between-group differences in Standard, ORAL Sync and ORAL Step studies, the addi-
mTSS at months 6 and 12 were - 0.37 and - 0.68, tion of tofacitinib 5 mg twice daily to background
respectively) [26]. The LS mean change from baseline in DMARD significantly improved physical function, as
the mTSS at month 12, the erosion score at months 6 and assessed by the LS mean change from baseline to month 3
12, and the joint-space narrowing score at month 6 did not in the HAQ-DI score (coprimary endpoint; Table 1)
differ significantly between the tofacitinib and placebo [22–24]. In ORAL Scan, significance was not declared for
groups. However, at month 12, the LS mean change from the HAQ-DI score (coprimary endpoint; Table 1) owing to
baseline in the joint-space narrowing score was signifi- the step-down assessment of coprimary endpoints [21]. In
cantly smaller in patients receiving tofacitinib than in those ORAL Scan, ORAL Standard and ORAL Sync, a signifi-
receiving placebo, indicating less structural damage cant improvement in the LS mean change from baseline in
(Table 2). Significantly more patients in the tofacitinib the HAQ-DI score with tofacitinib was evident within
group than placebo group had no radiographic progression 1–2 weeks of treatment (p\0.05 vs. placebo [21, 27] or
S. Dhillon

baseline [23]), with the magnitude of response maintained Physical function improved from baseline to a generally
or improved during B 12 months’ therapy [21, 23, 27]. similar extent in patients receiving tofacitinib plus
Fatigue (FACIT-F) scores and pain scores were also methotrexate, adalimumab plus methotrexate or tofacitinib
improved from baseline to a significantly (p\0.01) monotherapy, as assessed by the LS mean change from
greater extent in the tofacitinib groups than in the placebo baseline in the HAQ-DI score at months 6 and 12 (Table 1)
groups at month 3 in ORAL Standard, ORAL Sync and [20]. In addition, similar proportions of patients in the three
ORAL Step and at month 6 in ORAL Scan treatment groups had a MCID in the HAQ-DI score at
[21, 24, 27, 28]. At month 3, other health-related quality month 6 (70, 67 and 66% in the respective groups) and
of life (HR-QOL) outcomes in ORAL Standard, ORAL month 12 (64, 64 and 63%) [20]. Other HR-QOL outcomes
Sync and ORAL Step, including the patient’s global at these timepoints, including pain, FACIT-F, PtGA, SF-36
assessment (PtGA) score, SF-36 physical component PCS and MCS scores, improved from baseline to a gen-
summary (PCS) score, SF-36 mental component summary erally similar extent with tofacitinib plus methotrexate and
(MCS) score (in ORAL Sync and ORAL Step), Medical adalimumab plus methotrexate, with improvements in the
Outcomes Study (MOS) sleep score (in ORAL Standard tofacitinib combination therapy group numerically greater
and ORAL Sync) and scores for most SF-36 domains than those in the tofacitinib monotherapy group [30].
were improved from baseline to a significantly (p\0.05) The ORAL Standard study also included an adalimumab
greater extent in tofacitinib than placebo recipients plus methotrexate treatment arm (Table 1) and although
[24, 27, 28]. Significant improvements with tofacitinib the efficacy of tofacitinib plus methotrexate was not
relative to placebo in some measures were seen as early directly compared, the magnitude of efficacy responses in
as within 1 month of treatment, e.g. pain scores in ORAL the two treatment arms was similar [22]. Treatment with
Standard, ORAL Sync and ORAL Step [27–29]. Signifi- tofacitinib or adalimumab resulted in significant (p\0.05)
cantly (p\0.05) more patients receiving tofacitinib than improvements versus placebo in HR-QOL outcomes,
placebo in ORAL Standard, ORAL Sync and ORAL Step including HAQ-DI (Table 1), PtGA, FACIT-F and SF-36
had improvements of greater than or equal to the mini- PCS scores [27].
mum clinically important differences (MCIDs) in HR-
QOL outcomes, including HAQ-DI, pain, fatigue, PtGA, 4.2 As Monotherapy After Inadequate Response
SF-36 PCS, SF-36 MCS (in ORAL Sync and ORAL Step) to Methotrexate
and several SF-36 domains [27–29]. MCID was defined
as a decrease from baseline of C 0.22 points in HAQ-DI Tofacitinib 5 mg twice daily as monotherapy was effective in
scores and C 10 points in PtGA and pain scores; and an reducing signs and symptoms of disease and improving
increase from baseline of C 4 points in FACIT-F scores, physical function in the 6-month, phase 3 ORAL Solo study in
C 2.5 points in SF-36 PCS and MCS scores and C 5- patients with RA (mean disease duration & 8 years) who had
points in SF-36 domain scores. an inadequate response to C 1 cs/bDMARD (Table 1) [19]. At
month 3, ACR20 (coprimary endpoint), ACR50 and ACR70
4.1.2 Compared with Adalimumab response rates were significantly higher ([2-fold) in patients
receiving tofacitinib monotherapy than in those receiving
In the ORAL Strategy study in patients with RA who had placebo (Table 1). Significant (p B 0.05) between-group dif-
inadequate response to methotrexate, tofacitinib 5 mg ferences in ACR20, ACR50 and ACR70 response rates were
twice daily plus methotrexate was noninferior to adali- evident by 2–4 weeks of treatment and generally sustained
mumab plus methotrexate in terms of the ACR50 response during 6 months’ treatment with tofacitinib (ACR20, ACR50
at month 6 (primary endpoint; Table 1). Tofacitinib and ACR70 response rates at month 6 were 65, 42 and 22%,
monotherapy did not achieve statistical noninferiority to respectively; values estimated from graphs). The DAS28-ESR
either tofacitinib plus methotrexate or adalimumab plus remission rate (coprimary endpoint) at month 3 did not differ
methotrexate for the primary endpoint (Table 1). The significantly between the tofacitinib and placebo groups
ACR50 response achieved in all three treatment groups (Table 1); however, the DAS28-ESR low disease activity rate
was maintained during 12 months’ therapy (Table 1) [20]. (Table 1) and the LS mean change from baseline in DAS28-
The ACR20 and ACR70 response rates and DAS28-ESR ESR [- 2.0 vs. - 1.1 (baseline 6.7); p\0.001] was signifi-
remission and low disease activity rates at month 6 were cantly greater in tofacitinib than placebo recipients [19]. Fol-
generally similar in patients receiving tofacitinib plus lowing 6 months’ treatment with tofacitinib, 9.8% of patients
methotrexate or adalimumab plus methotrexate, with the were in remission, 22.0% had low disease activity and the LS
rates in both combination therapy groups being numeri- mean change from baseline in DAS28-ESR was - 2.4 [19].
cally greater than the rates in the tofacitinib monotherapy Physical function at month 3, as assessed by the LS
group (Table 1). mean change from baseline in the HAQ-DI score
Tofacitinib: A Review

(coprimary endpoint), was significantly improved with signs and symptoms of disease and inhibiting progression
tofacitinib relative to placebo (Table 1) [19, 31]. Other of structural joint damage in the 24-month, phase 3
HR-QOL outcomes at month 3, including PtGA, pain, ORAL Start study in methotrexate-naı̈ve patients with RA
FACIT-F, and SF-36 PCS and MCS scores, were also [18]. For the majority (65.5%) of patients the interval
significantly improved (p\0.05) with tofacitinib relative to between diagnosis and enrolment was \2 years and for
placebo. Moreover, the improvements from baseline in 40% of patients the duration was \6 months (median
HAQ-DI, PtGA, pain, and SF-36 PCS and MCS scores 0.7–0.8 years); the mean disease duration was & 3 years.
with tofacitinib exceeded the MCIDs [31]. Tofacitinib had At month 6, ACR70 (coprimary endpoint), ACR20 and
a rapid onset of action, with significant (p\0.01 vs. pla- ACR50 response rates were significantly higher in tofac-
cebo) benefits in HAQ-DI, PtGA and pain scores evident as itinib than methotrexate recipients (Table 3), with signif-
early as week 2 of treatment and the magnitude of response icant (p\0.001) improvements seen as early as month 1
with tofacitinib sustained during 6 months’ therapy of therapy and sustained over 24 months’ treatment.
[19, 31]. DAS28-ESR remission and DAS28-ESR low disease
At month 6, the ACR20, ACR50 and ACR70 response activity rates at month 6 were almost 2-fold higher with
rates and HAQ-DI scores in patients who switched from tofacitinib than methotrexate (Table 3), with the magni-
placebo to tofacitinib at month 3 (see Table 1 for study tude of response generally sustained over 24 months’
details) were generally similar to those in patients who had therapy. In addition, the proportion of patients achieving a
received tofacitinib for 3 months [19]. ‘good or moderate’ EULAR response was significantly
Tofacitinib monotherapy in this clinical setting was also (p\0.001) higher in tofacitinib than methotrexate recip-
assessed in the ORAL Strategy study. Results showed that ients at month 6 (79 vs. 61%) [18].
monotherapy did not achieve statistical noninferiority to Tofacitinib monotherapy was associated with signifi-
combination treatment with tofacitinib plus methotrexate cantly less radiographic damage than methotrexate
or adalimumab plus methotrexate, while the two combi- monotherapy [18]. At month 6, the mTSS (coprimary
nation therapies were noninferior to each other (Table 1; endpoint), erosion score and joint-space narrowing score
see Sect. 4.1.2) [20]. increased from baseline to a significantly smaller extent in
tofacitinib than methotrexate recipients, with benefits
4.3 As Monotherapy in Methotrexate-Naı̈ve maintained at month 24 (Table 2). The proportion of
Patients patients with no radiographic progression (B 0.5 unit
increase in mTSS) at month 24 was also significantly
Tofacitinib 5 mg twice daily as monotherapy was more higher in the tofacitinib than the methotrexate group
effective than methotrexate monotherapy in reducing (Table 2) [18].

Table 3 Efficacy of oral tofacitinib 5 or 10 mg twice daily in methotrexate-naı̈ve patients with rheumatoid arthritis in the 24-month, ran-
domized, double-blind, multicentre, phase 3 ORAL Start study [18]

Study Timepoint Treatment Response ratesa (% pts) Remissiona,b LDAa,b Change from
(months) (no. of FAS pts) BL HAQ-DI [BL]c
ACR20 ACR50 ACR70 (% pts) (% pts)

ORAL Start 6 TOF 5 (373) 71.3*** 46.6*** 25.5***d 14.6* 27.8*** - 0.8*** [1.5]
TOF 10 (397) 76.1*** 56.4*** 37.7***d 21.8*** 38.2*** – 0.9*** [1.5]
MTXe (186) 50.5 26.6 12.0d 7.6 14.0 - 0.6 [1.5]
24 TOF 5 64.2*** 49.3*** 34.4*** 20.8*** 34.8*** - 0.9***
TOF 10 64.2*** 49.2*** 37.6*** 22.3*** 36.0*** – 1.0***
MTXe 42.4 28.3 15.2 9.9 15.8 - 0.7
ACR ‘x’ C x% improvement in ACR criteria, BL baseline, DAS28-ESR Disease Activity Score 28-erythrocyte sedimentation rate, FAS full
analysis set, HAQ-DI Health Assessment Questionnaire – Disability Index score (lower scores indicate less disability), LDA low disease activity,
MTX methotrexate, pts patients, TOF tofacitinib
*p B 0.05, **p B 0.01, ***p\0.001
a
Missing values imputed using nonresponse imputation
b
Proportion of pts in remission (DAS28-ESR of \2.6) or with LDA (DAS28-ESR of B 3.2)
c
Least-squares mean changes from BL; all BL values are means
d
Coprimary endpoints; significance of coprimary endpoints was assessed using a step-down approach
e
MTX target-dose after titration of 20 mg/week
S. Dhillon

At month 6, physical function (assessed by the LS mean (mean change 0.25 and 1.17), erosion score (0.21 and 0.68)
change from baseline in the HAQ-DI score) had improved to and joint-space narrowing score (0.24 and 0.78) was seen
a significantly greater extent in tofacitinib than methotrexate with tofacitinib; baseline was the last visit in the index
recipients (Table 3), with significant (p\0.05) between- study prior to LTE entry. The proportion of patients with
group differences seen from month 1 and sustained over no radiographic progression (change from baseline in
24 months’ therapy [18, 32]. Tofacitinib reduced fatigue mTSS of B 0.5) decreased during this period from 84.5% at
(FACIT-F scores) and arthritis pain to a significantly greater month 12 to 72.2% at month 36, and the proportion of
extent than methotrexate at month 6, with significant patients with no new erosions (change from baseline in
(p\0.05) between-group differences seen at month 1 and erosion score of B 0.5) decreased from 92.3 to 85.3%. At
generally maintained during 24 months’ therapy [18, 32]. months 12 and 36, x-ray data were available from 1003
Other HR-QOL outcomes, including PtGA, and SF-36 PCS (86.8%) and 858 (76%) of patients, respectively [35].
scores, were also significantly (p\0.05) improved with
tofacitinib relative to methotrexate at months 3, 6, 12 and 4.5 Other Studies
24; improvements in PtGA scores were seen from month 1
onwards [32]. In addition, significantly (p\0.05) more Post hoc pooled data from five phase 3 ORAL studies
tofacitinib than methotrexate recipients at month 6 had (Solo, Scan, Standard, Sync and Step) and four phase 2
MCIDs in PtGA scores and in three SF-36 domains (phys- studies showed that tofacitinib was effective in improving
ical functioning, bodily pain and mental health) [32]. the signs and symptoms of RA when used before or after
bDMARDs [36]. For instance, at month 3, ACR20
4.4 Long-Term Efficacy responses were achieved in 60.3% of tofacitinib 5 mg
twice daily recipients versus 26.5% of placebo recipients
Pooled data from two open-label long-term extension (p\0.0001) in the bDMARD-naı̈ve group (n = 2812) and
(LTE) studies (a 72-month Japanese study A3921041 and in 43.4 versus 24.6% (p\0.0001) of patients in the
the ongoing ORAL Sequel study, data cut-off March 2017) bDMARD-experienced group (n = 705) [36]. Another
involving patients participating in phase 1–3 studies, post hoc pooled analysis of data from five phase 3 ORAL
showed that the efficacy of tofacitinib (5 or 10 mg twice studies (Solo, Scan, Standard, Sync and Step) suggested
daily with or without DMARDs) was sustained for at least that the treatment response may be greater if tofacitinib
96 months [33, 34]. In 1421 patients receiving tofacitinib 5 mg twice daily is initiated earlier in the course of RA,
5 mg twice daily (with or without background DMARDs), thereby potentially reducing the time patients experience
ACR20 response rates at months 1 and 48 were 73.8 and active disease [37].
74.4%, respectively; ACR50 response rates were 49.8 and Tofacitinib 5 mg twice daily (with or without DMARDs)
49.6; ACR70 response rates were 28.8 and 34.1; DAS28- improved RA outcomes regardless of body mass index at
ESR remission was achieved by 23.1 and 21.6% of patients; baseline (\25, 25 to \30, and C 30 kg/m2) [38] or prior
DAS28-ESR low disease activity was achieved by 39.3 and therapy (inadequate response/intolerance to methotrexate,
42.4% of patients; the mean DAS28-ESR was 3.69 and bDMARD or csDMARD other than methotrexate) [39] in
3.56; and the mean HAQ-DI score was 0.78 and 0.81 [33]. post hoc pooled analyses of six phase 3 [38] and 14 phase 2
The total tofacitinib exposure in this analysis was 3215 or 3 trials [39]. In addition, the Japanese LTE study
patient-years (PY) and the mean (maximum) duration of (A3921041) [40], a post hoc subgroup analysis of the
therapy was 826 (1844) days [33]. Similar results were seen ORAL Sync study [41] and post hoc pooled analyses of
at 96 months, with 77.9, 59.5 and 41.7% of patients who phase 2, 3 and/or LTE studies [38, 42–44] suggested that the
received tofacitinib 5 or 10 mg twice daily maintaining efficacy of tofacitinib 5 mg twice daily (with or without
ACR20, ACR50 and ACR70 responses, respectively; the DMARDs) was generally consistent between Chinese [41],
mean DAS28-ESR was 3.34 and the mean HAQ-DI was Japanese [40] or Latin American [42] patients and the
0.77. By month 96, 4967 patients had received tofacitinib 5 overall tofacitinib population; between patients in the USA
or 10 mg twice daily with or without background DMARDs and rest of the world [43]; and between younger
for a mean (maximum) duration of 3.5 (9.4) years [34]. (\65 years) and older (aged C 65 years) patients [44].
Assessment of radiographic progression in a radio-
graphic substudy of ORAL Sequel (n = 1156) showed that
patients receiving tofacitinib 5 or 10 mg twice daily (with 5 Tolerability of Tofacitinib
or without DMARDs) had limited structural progression
during longer-term therapy (mean treatment duration after The tolerability profile of oral tofacitinib in patients with
LTE entry 1113 days; data cut off March 2017) [35]. At active RA is based on the pivotal studies discussed in Sect.
months 12 and 36, a slight increase from baseline in mTSS 4, pooled data from two LTEs (study A3921041 and ORAL
Tofacitinib: A Review

Sequel study; Sect. 4.4) [5, 34, 45], and pooled data from 4.9 years with background csDMARDs) [47]. AE-associ-
phase 1–3 studies (including ORAL Start, ORAL Solo, ated withdrawal rates with tofacitinib were higher than
ORAL Scan, ORAL Standard, ORAL Sync and ORAL rates associated with loss of efficacy (21.6 vs. 3.1%), with
Step) and the LTE studies [5, 45]. The latter pooled anal- the most common reasons for treatment discontinuation
ysis included data from a total of 6194 patients (referred to being infections and infestations (8.8%), investigations
as the long-term safety population hereafter) who had (4.2%) and benign, malignant or unspecified neoplasms
received tofacitinib (all doses, with or without DMARDs) (3.2%). Drug survival and treatment discontinuation rates
for a mean duration of 3.13 years (median 3.4 years; data for the two tofacitinib doses were generally similar [47].
cut-off of 31 March 2015; accumulated total exposure of
19406 PY, based on B 8.5 years of exposure) [5, 45, 46]. 5.1 Infections
Tofacitinib was generally well tolerated in patients with
RA, with a tolerability profile generally similar to that of During B 8.5 years’ treatment with tofacitinib 5 mg twice
bDMARDs. In the long-term safety population, the inci- daily in the long-term safety population, the incidence rate
dence of treatment-emergent adverse events (AEs) with of infections was 43.8 patients/100 PY (48.9 patients/100
tofacitinib (all doses) was 136.9 patients/100 PY, the rate PY with tofacitinib monotherapy and 41.0 patients/100 PY
of discontinuation because of AEs was 7.5 patients/100 PY with tofacitinib plus DMARDs) and the rate of serious
and that of serious treatment-emergent AEs (SAEs) was 9.4 infections was 2.4 patients/100 PY, with the most com-
patients/100 PY [45]. During the first 3 months of treat- mon serious infections being pneumonia, HZ, UTI, cel-
ment in 6- to 24-month randomized, controlled studies lulitis, gastroenteritis and diverticulitis [5]. Significant
(n = 1849 tofacitinib and 1079 placebo recipients), treat- (p\0.05) risk factors for serious infections included use
ment-emergent AEs occurred in 51.4% of patients receiv- of glucocorticoids at baseline, higher baseline HAQ-DI
ing tofacitinib 5 mg twice daily (vs. 51.8% of placebo score, prior confirmed post-baseline lymphopenia (\500
recipients), with headache (4.5 vs. 2.4%), upper respiratory cells/mm3), line of therapy (third-line vs. second-line) and
tract infections (URTI; 3.8 vs. 3.8%), nasopharyngitis (3.5 geographical region (e.g. Asian patients vs. patients in the
vs. 3.2%), diarrhoea (3.5 vs. 2.6%) and nausea (2.8 vs. USA/Canada) [45]. Age was also a significant (p\0.05)
2.3%) reported most frequently (incidence [2.5%) [46]. risk factor for serious infections [45], with a two-fold
SAEs occurred in approximately 2.7% of patients each in higher incidence in the elderly (aged C 65 years) than in
the tofacitinib and placebo groups during this period, with younger patients (aged \65 years) (4.8 vs. 2.4 patients/
the most common SAEs being infections and infestations, 100 PY) [5].
occurring in 0.8 and 0.4% of patients in the respective Viral reactivation and cases of herpes virus reactivation
groups [46]. The adverse event-related withdrawal rate was (e.g. HZ), as well as opportunistic infections, such as
3.8 and 3.4% in the tofacitinib and placebo groups, tuberculosis (TB) and other mycobacterial infections,
respectively; the most common infections leading to cryptococcus and histoplasmosis, have also been reported
treatment withdrawal in tofacitinib recipients were HZ and with tofacitinib therapy [5]. In the long-term safety popu-
pneumonia [5, 46]. lation, HZ was reported in 703 patients (incidence 3.9
The tolerability profile of tofacitinib (5 or 10 mg twice patients/100 PY), active TB in 36 patients (0.2 patients/100
daily with or without DMARDs) in the pooled LTEs PY; four patients had latent TB at screening with adequate
[B 9.4 years’ (B 114 months) therapy] was generally con- therapy) and opportunistic infections excluding TB in 61
sistent with that seen during short-term treatment [34]. patients (0.3 patients/100 PY) receiving tofacitinib (all
Infections and infestations occurred most commonly doses) [45]. The incidence of HZ appears to be higher in
(69.6% of patients) in tofacitinib recipients, with the most several patient subgroups, including Asian patients (par-
frequently reported infections being nasopharyngitis ticularly Japanese and Korean patients) and patients with
(19.1%), URTI (17.9%), bronchitis (12.6%) and urinary long-standing RA who had prior treatment with C 2
tract infection (UTI; 12.5%). SAEs occurred in 29.4% of bDMARDs; patients with an ALC of \1000 cells/mm3
tofacitinib recipients (9.13 patients/100 PY), with serious may also be at increased risk of HZ [5]. There was no
infections reported in 8.9% of patients (2.46 patients/100 notable increase in the incidence rate of serious infections
PY); 23.9% of tofacitinib recipients discontinued treatment or HZ during long-term therapy [45]. Owing to a risk of HZ
because of AEs [34]. with tofacitinib, consideration should be given to prophy-
An earlier pooled analysis of the two LTE studies (data lactic zoster vaccination, particularly in high risk patients
cut-off March 2015; n = 4867), showed that during a mean [5]; tofacitinib did not adversely affect humoral and cell-
(maximum) treatment duration of 3.0 (7.9) years, the mediated immune responses against, and the safety of, live
median overall drug survival of tofacitinib 5 or 10 mg zoster virus vaccine in patients with RA who had pre-ex-
twice daily was 5.0 years (5.1 years as monotherapy and isting immunity to varicella zoster virus [48].
S. Dhillon

5.2 Cardiovascular Safety and Haematological \1000 cells/mm3 was reported in 0.08% of patients [5].
Changes An ALC of \750 cells/mm3 was associated with an
increased incidence of serious infections, but no clear
Tofacitinib was associated with a low incidence of car- relationship was seen between an ANC of \1000 cells/
diovascular (CV) events [45]. In the long-term safety mm3 and the occurrence of serious infections [5].
population, 71 patients experienced Major Adverse Car-
diovascular Events (MACE; CV death and non-fatal CV 5.3 Malignancies and Other Adverse Events
events) during B 8.5 years’ treatment with tofacitinib (all
doses); the incidence rate of MACE ranged between 0.2 During B 8.5 years’ treatment with tofacitinib (all doses) in
and 0.7 patients/100 PY over this period [45]. the long-term safety population, 173 patients (incidence 0.9
Moderate increases in lipid levels seen with tofacitinib patients/100 PY) had malignancies [excluding non-me-
(Sect. 2) appear to be non-atherogenic [46]. However, an lanoma skin cancer (NMSC)], which included lung cancer
association between increased lipid levels and higher risk (0.2 patients/100 PY), breast cancer (0.2 patients/100 PY)
of cardiovascular disease (CVD) cannot be excluded, par- [45] and lymphomas (0.1 patients/100 PY) [51]. NMSC
ticularly in the RA population which is at increased risk of occurred at an incidence of 0.6 patients/100 PY (118
CVD [46]. A post hoc pooled analysis of six phase 3 patients) [45], which was no higher than that in the general
ORAL studies (Start, Solo, Scan, Standard, Sync and Step) RA population [46]. No notable increase in the incidence rate
assessed the impact of changes in lipids and high-sensi- of malignancies was evident during long-term therapy [45].
tivity (hs) CRP on CV risk by using the Framingham and Although the incidence of lymphoma was low in the tofac-
Reynolds risk scores to calculate the 10-year risk of itinib clinical trials, the risk of developing lymphoma may be
developing CVD [49]. Results showed that the Framing- higher in patients with RA than in the general population,
ham CV risk score was significantly higher with tofacitinib particularly in patients with highly active disease [5].
(5 or 10 mg twice daily with or without DMARDs) than GI perforations were reported infrequently (n = 22
with placebo in ORAL Sync (both doses) and ORAL Step patients) with tofacitinib (all doses) in the long-term safety
(10 mg dose), and significantly higher than with population (incidence 0.11 patients/100 PY) [45]. All tofac-
methotrexate in ORAL Start (both doses) (all p\0.05). itinib recipients with GI perforations were receiving con-
The Reynolds risk score, which includes hsCRP as a comitant NSAIDs or corticosteroids, 13 patients had a history
variable, was generally reduced with tofacitinib relative to of diverticulitis or diverticulosis and two patients had a his-
placebo in five studies (all p\0.05, except p = non- tory of gastric ulcers [45]; treatment with NSAIDs and/or
significant for the 5 mg dose in ORAL Step) and did not corticosteroids and prior history of diverticulitis are known
differ significantly from placebo in ORAL Start [49]. risk factors for GI perforations in patients with RA [52].
Clinically relevant decreases in haemoglobin levels of Confirmed increases in liver enzymes (ALT or AST) of
B 7 g/dL were reported infrequently (\2% of patients) greater than three times the upper limit of normal (ULN)
with tofacitinib (5 or 10 mg twice daily with or without were uncommon in patients receiving tofacitinib (with or
DMARDs) during B 7 years’ (B 84 months) therapy [50]. without DMARDs) [5]; during B 9.4 years’ (B 114 months)
There have also been reports of neutropenia and lym- therapy with tofacitinib (5 or 10 mg twice daily with or
phopenia in tofacitinib recipients [5]. After an initial without DMARDs) in the LTEs, 5.7 and 3.3% of patients
transient increase in the mean ALC during the first had increases in ALT and AST of C 3 times the ULN,
3 months of treatment with tofacitinib (5 or 10 mg twice respectively [34].
daily with or without DMARDs), lymphocyte counts
decreased gradually until month 48 and stabilized there- 5.4 Compared with Adalimumab
after [50]. During B 8.5 years’ treatment in the long-term
safety population, a confirmed decrease in ALC of \500 The 12-month ORAL Strategy study showed that the tol-
cells/mm3 and 500–750 cells/mm3 was reported in 1.3% erability profile of tofacitinib plus methotrexate was gen-
and 8.4% of patients, respectively [5]. Most patients with erally similar to that of adalimumab plus methotrexate,
an ALC of \500 cells/mm3 achieved counts C 500 cells/ with no new safety concerns reported in either combination
mm3 after & 3 weeks of treatment discontinuation therapy group or in the tofacitinib monotherapy group [20].
[14, 50]. Mean neutrophil counts decreased during the first In tofacitinib monotherapy, tofacitinib plus methotrexate
3 months of treatment with tofacitinib (5 or 10 mg twice and adalimumab plus methotrexate recipients, treatment-
daily with or without DMARDs) and then remained rela- emergent AEs occurred in 59, 61 and 66% of patients,
tively stable during B 66 weeks’ therapy [50]. During respectively, treatment-related AEs in 26, 30 and 35% of
B 8.5 years’ treatment in the long-term safety population, a patients and SAEs in 9, 7 and 6% of patients; corre-
confirmed decrease in absolute neutrophil count (ANC) of sponding AE-related withdrawal rates were 6, 7 and 10%.
Tofacitinib: A Review

Most AEs in all treatment groups were of mild to moderate therapies were started late during the course of the disease
severity, with the most common being URTI (7, 10 and 8% [2, 53]. Today, disease modification has become a
with tofacitinib, tofacitinib plus methotrexate and adali- cornerstone of RA management and is a key requirement
mumab plus methotrexate, respectively), nasopharyngitis for all modern antirheumatic agents [2]. A variety of
(6, 4 and 5%), UTI (3, 4 and 4%), nausea (3, 4 and 4%) and pharmacotherapeutic options are available, including
increase in ALT levels (2, 6 and 7%). AEs of special (i) csDMARDs, such as methotrexate (anchor drug) and
interest also occurred at generally similar rates in tofaci- sulfasalazine (ii) bDMARDs, including TNF inhibitors
tinib, tofacitinib plus methotrexate and adalimumab plus (e.g. adalimumab, etanercept) and IL-6 inhibitors (e.g.
methotrexate recipients, including serious infections (2, 3 tocilizumab) (iii) biosimilars, including infliximab and
and 2%, respectively), HZ (1, 2 and 2%), TB (0, 1 and 0%) etanercept biosimilars (iv) and targeted synthetic DMARDs
and malignancies (excluding NMSC;\1, 0 and 0%) [20]. (tsDMARDs), such as JAK inhibitors (e.g. tofacitinib,
baricitinib).
Current EU treatment guidelines recommend adopting a
6 Dosage and Administration of Tofacitinib treat-to-target approach for the management of RA, with
the aim of achieving remission or sustained low disease
In the EU, oral tofacitinib in combination with methotrexate activity [2]. Treatment should be initiated soon after
is indicated for the treatment of moderate to severe active diagnosis, with the choice of therapy based on disease
RA in adult patients who have responded inadequately to, or activity and other patient factors (e.g. progression of
who are intolerant of, one or more DMARDs [5]. Tofaci- structural damage), comorbidities, safety profile of agents,
tinib may be administered as monotherapy in patients who and medical and societal costs. Methotrexate should be part
are intolerant of methotrexate or when treatment with of the first treatment strategy in patients with active RA,
methotrexate is inappropriate. The recommended dosage of unless contraindicated or not tolerated, in which case
tofacitinib is 5 mg administered twice daily, with or without another csDMARD (leflunomide or sulfasalazine) may be
food. No dosage adjustment is needed when tofacitinib is used. If the treatment target is not achieved within
coadministered with methotrexate or when given to patients 3–6 months and in the absence of unfavourable prognostic
with mild [creatinine clearance (CLCR) 50–80 mL/min] or factors, switching to a, or the addition of another,
moderate (CLCR 30–49 mL/min) renal impairment or mild csDMARD is recommended. When unfavourable prog-
hepatic impairment (Child Pugh A) [5]. However, tofaci- nostic factors are present (e.g. moderate to high activity,
tinib dosage should be reduced to 5 mg once daily in high acute phase reactant levels or high swollen joint
patients with severe renal impairment (CLCR\30 mL/min) counts), a bDMARD or tsDMARD should be combined
or moderate hepatic impairment (Child Pugh B), and when with the csDMARD; current practice is to start with a
coadministering tofacitinib with potent CYP3A4 inhibitors bDMARD, owing to the long-term experience with these
(e.g. ketoconazole) or drugs that are both moderate inhibi- agents. If the treatment target is still not reached, use of any
tors of CYP3A4 and potent inhibitors of CYP2C19 (e.g. other bDMARD or tsDMARD is recommended [2].
fluconazole) [5]. Recently, two tsDMARDs tofacitinib [20] and barici-
Tofacitinib is contraindicated in patients with active TB, tinib [54] have been approved in the EU for the treatment
serious (e.g. sepsis) or opportunistic infections, in patients of patients with moderate to severe active RA, and have
with severe hepatic impairment, and during pregnancy and been included as treatment options in the current EU [2]
lactation. Regular monitoring of patients is recommended, and NICE [55, 56] treatment guidelines (consult individual
and temporary or permanent discontinuation of tofacitinib guidelines for further details). Tofacitinib is a potent,
may be required for the management of AEs and laboratory selective JAK inhibitor that preferentially inhibits sig-
abnormalities associated with tofacitinib therapy (Sect. 5). nalling by heterodimeric cytokine receptors that associate
[5]. Local prescribing information should be consulted for with JAK3 and/or JAK1 (Sect. 2). It is an organic small
further information, including dosage and administration molecule with convenient oral twice daily dosing relative
details, contraindications, warnings and precautions. to bDMARDs, which are proteinaceous molecules admin-
istered subcutaneously or intravenously [57]. An extensive
phase 3–4 ORAL trial programme in patients with RA
7 Place of Tofacitinib in the Management showed that tofacitinib (monotherapy and/or combination
of Rheumatoid Arthritis therapy with csDMARDs) was effective in reducing signs
and symptoms of disease and improving HR-QOL in a
Management strategies for RA have changed substantially range of patient populations, including patients naı̈ve to
since the 1990s when there were few and minimally methotrexate, patients with inadequate response to
effective therapeutic agents and available antirheumatic cs/bDMARDs and patients with inadequate response to
S. Dhillon

TNF inhibitors (Sect. 4). Tofacitinib monotherapy was suggested that the risk of serious infections with tofacitinib
more effective than methotrexate or placebo, but was was generally similar to that with bDMARDs (risk ratio
generally less effective than combination therapy with versus placebo of 2.2 for tofacitinib 5 mg twice daily vs.
tofacitinib or adalimumab plus methotrexate; the two 1.0–2.27 for bDMARDs) [59].
combination therapies were noninferior to each other [20]. However, the incidence of HZ (Sect. 5.1) was approxi-
Tofacitinib had a rapid onset of response with improve- mately 1.5- to 2-fold higher in tofacitinib recipients than
ments in signs and symptoms, and physical function gen- patients receiving placebo or bDMARDs [60], which
erally evident within 2–4 weeks of treatment and benefits appears to be a class effect [58]. Similar results were seen
sustained during long-term therapy (B 96 months) (Sect. in a real-world study (based on 2006–2013 US Medicare
4). data and 2010–2014 US Marketscan data), which showed
In terms of structural preservation, tofacitinib that the risk of HZ was 2-fold higher with tofacitinib than
monotherapy was superior to methotrexate monotherapy in bDMARDs (hazard ratio 2.01 vs. 1.0–1.17) [61]. Impor-
inhibiting progression of structural damage (assessed by tantly, in clinical studies, HZ was manageable, with the
mTSS) in ORAL Start in methotrexate-naı̈ve patients [18], majority of patients recovering and no HZ-related deaths
with treatment benefits maintained for up to 24 months reported with tofacitinib [46]. Although an understanding
(Sect. 4.3). In ORAL Scan in patients with inadequate of the biological mechanism of the increased risk of viral
response to methotrexate [21], although the addition of reactivation with tofacitinib is currently lacking, it has been
tofacitinib to background methotrexate did not demonstrate suggested that tofacitinib-mediated inhibition of IFN sig-
superiority over the addition of placebo in inhibiting pro- nalling via JAK1 receptor may play a role, as antiviral
gression of structural damage (assessed by mTSS), signif- defences in humans rely upon IFN signalling [60].
icantly more patients receiving tofacitinib experienced no Tofacitinib did not appear to increase the risk of
radiographic disease progression (Sect. 4.1.1.2). Moreover, malignancies, GI perforations or MACE during long-term
patients with unfavourable prognostic factors experienced therapy (Sect. 5). Changes in laboratory parameters with
greater benefits with tofacitinib than placebo, according to tofacitinib were generally mild or moderate, and reversible
post hoc analyses (Sect. 4.1.1.2). In addition, when data with treatment interruption (Sect. 5).
from both studies were analysed using a trimmed analysis Although assessed for B 114 months, the overall clinical
(to assess the effect of outliers), a significant (p B 0.05) experience with tofacitinib is less than that with
benefit of tofacitinib over placebo in terms of mTSS was bDMARDs. In addition, apart from a head-to-head com-
seen in both ORAL Start and ORAL Scan [26]. It was parison of tofacitinib with adalimumab (in addition to
suggested that the difference in structural preservation background methotrexate) in ORAL Strategy [20], there
outcomes between the ORAL Start and ORAL Scan studies are no direct comparisons of tofacitinib and bDMARDs.
may be because of differences in the sensitivity of the two However, indirect network meta-analyses suggest that the
study populations in terms of revealing a treatment benefit efficacy and safety profile of tofacitinib is generally similar
over the short treatment duration [46]. Patients with inad- to that of bDMARDs [62, 63]. There are also no clinical
equate response to methotrexate may be less sensitive due trials comparing the efficacy of tofacitinib with that of oral
to a smaller potentially responsive subset and a lower rate baricitinib once daily, which preferentially inhibits JAK1
of structural progression than methotrexate-naı̈ve patients and JAK2 and has demonstrated efficacy in similar clinical
[46]. settings [54]. In one clinical study (RA-BEAM), when
Tofacitinib was generally well tolerated in clinical added to background methotrexate, baricitinib was more
studies (Sect. 5), with a tolerability profile generally similar effective than adalimumab in reducing signs and symptoms
to that of bDMARDs [58], such as adalimumab (Sect. 5.4). of disease and inhibiting progression of structural joint
During B 114 months’ therapy, the most common treat- damage in patients with active RA who had inadequate
ment-emergent AEs with tofacitinib were infections and response to methotrexate [64]. Although not directly
infestations, of which URTI and nasopharyngitis were the compared, the tolerability profile of baricitinib appears to
most frequent (Sect. 5). Infections were also the most be generally similar to that of tofacitinib, with both agents
common SAEs, with pneumonia and HZ among the most associated with an increased risk of HZ [58]. Baricitinib
frequently reported (Sect. 5.1). A systematic review and has also been associated with cases of deep vein throm-
meta-analysis of data from 66 randomized controlled trials bosis (DVT) and pulmonary embolism (PE) (5 cases during
and 22 LTE studies of bDMARDs and tofacitinib 6637 PY experience) [65], and the summary of product
Tofacitinib: A Review

characteristics (SPC) carries a warning regarding the same tsDMARDs, current evidence indicates that oral tofacitinib
(UK SPC [66]). A signal for DVT and PE has not been is a useful option for the treatment of patients with RA.
confirmed with tofacitinib despite longer clinical experi-
ence than that with baricitinib.
Drug survival or the time to discontinuation of medi- Data Selection Tofacitinib: 178 records identified
cations is largely determined by the efficacy and tolera-
Duplicates removed 38
bility of a drug, and assesses the long-term impact of
medication on the course of the disease [67]. Early results Excluded at initial screening (e.g. press releases; news 12
in the clinical setting showed that tofacitinib had a median reports; not relevant drug/indication)
drug survival of 5 years (Sect. 5). In the real-world setting, Excluded during initial selection (e.g. preclinical study; 8
persistence at 12 months was found to be generally similar reviews; case reports; not randomized trial)
for tofacitinib, adalimumab and etanercept in a retrospec- Excluded during writing (e.g. reviews; duplicate data; 50
tive US cohort study in bDMARD naı̈ve-patients with RA small patient number; nonrandomized/phase I/II trials)
who had received prior methotrexate [68]. Additional Cited efficacy/tolerability articles 37
studies would help to determine how tofacitinib compares Cited articles not efficacy/tolerability 33
with bDMARDs in this respect during long term therapy.
Search Strategy EMBASE, MEDLINE and PubMed from 2013 to
In addition to the efficacy and tolerability of antirheu- present. Previous Adis Drug Evaluation published in 2013 was
matic drugs, the convenience of drug intake, as determined hand-searched for relevant data Clinical trial registries/databases
by the frequency and route of administration, may impact and websites were also searched for relevant data. Key words were
patients’ adherence to treatment and ultimately to the Tofacitinib, Xeljanz, CP-690550, rheumatoid arthritis, BID, twice
daily. Records were limited to those in English language. Searches
failure or success of therapy [69, 70], particularly if the last updated 27 October 2017
drugs have similar efficacy and tolerability (e.g.
bDMARDs and tsDMARDs [62, 63]). Tofacitinib has
convenient oral twice daily administration, which may be
Acknowledgements During the peer review process, the manufac-
preferred by patients, potentially increasing satisfaction turer of tofacitinib (XeljanzÒ) was also offered an opportunity to
and adherence, and thereby improving clinical outcomes. A review this article. Changes resulting from comments received were
recent study examining patient preferences in the treatment made on the basis of scientific and editorial merit.
of RA using a discrete-choice approach suggested that an
Compliance with Ethical Standards
oral DMARD that does not have to be combined with
methotrexate and is not administered (only) every Funding The preparation of this review was not supported by any
1–2 weeks may be a favourable second-line option in external funding.
patients with RA [70]. Well-designed studies are needed to
Conflict of interest Sohita Dhillon is a salaried employee of Adis/
assess adherence/compliance to therapy with tofacitinib Springer, is responsible for the article content and declares no rele-
and any potential impact this may have on clinical vant conflicts of interest.
outcomes.
The cost-effectiveness of treatment is also an important Additional information about this Adis Drug Review can be found at
http://www.medengine.com/Redeem/ED1CF06073837128.
factor in determining the choice of therapy in RA, and
current NICE guidelines indicate that tofacitinib and
baricitinib are cost-effective after cs/bDMARDs in some References
clinical settings [55, 56]. Additional well-designed cost-
effectiveness analyses would help to more definitively 1. O’Shea JJ, Laurence A, McInnes IB, et al. Back to the future: oral
targeted therapy for RA and other autoimmune diseases. Nat Rev
position tofacitinib relative to cs/bDMARDs, including
Rheumatol. 2013;9(3):173–82.
biosimilar DMARDs. 2. Smolen JS, Landewe R, Bijlsma J, et al. EULAR recommenda-
In conclusion, B 114 months’ clinical experience in tions for the management of rheumatoid arthritis with synthetic
patients with active RA showed that tofacitinib and biological disease-modifying antirheumatic drugs: 2016
update. Ann Rheum Dis. 2017;76(6):960–77.
monotherapy (as first- and second-line treatment) and
3. Winthrop KL, Strand V, van der Heijde DM, et al. The unmet
combination therapy with methotrexate (as second- and need in rheumatology: reports from the Targeted Therapies
third-line treatment) was effective and generally well tol- meeting 2016. Clin Exp Rheumatol. 2016;34(4 Suppl 98):69–76.
erated. When added to background methotrexate, tofaci- 4. Hodge JA, Kawabata TT, Krishnaswami S, et al. The mechanism
of action of tofacitinib—an oral Janus kinase inhibitor for the
tinib and adalimumab were noninferior in terms of efficacy
treatment of rheumatoid arthritis. Clin Exp Rheumatol.
and had generally similar tolerability profiles. Although 2016;34(2):318–28.
additional comparative studies are needed to more defini- 5. Pfizer Ltd. Xeljanz (tofacitinib): summary of product character-
tively position tofacitinib relative to bDMARDs and other istics. 2017. http://www.ema.europa.eu/docs/en_GB/document_
S. Dhillon

library/EPAR_-_Product_Information/human/004214/WC50022 23. Kremer J, Li ZG, Hall S, et al. Tofacitinib in combination with


4911.pdf. Accessed 31 Oct 2017. nonbiologic disease-modifying antirheumatic drugs in patients
6. Scott LJ. Tofacitinib: a review of its use in adult patients with with active rheumatoid arthritis: a randomized trial. Ann Intern
rheumatoid arthritis. Drugs. 2013;73(8):857–74. Med. 2013;159(4):253–61.
7. Meyer DM, Jesson MI, Li X, et al. Anti-inflammatory activity 24. Burmester GR, Blanco R, Charles-Schoeman C, et al. Tofacitinib
and neutrophil reductions mediated by the JAK1/JAK3 inhibitor, (CP-690,550) in combination with methotrexate in patients with
CP-690,550, in rat adjuvant-induced arthritis. J Inflamm. active rheumatoid arthritis with an inadequate response to tumour
2010;7:41. necrosis factor inhibitors: a randomised phase 3 trial. Lancet.
8. Karaman MW, Herrgard S, Treiber DK, et al. A quantitative 2013;381(9865):451–60.
analysis of kinase inhibitor selectivity. Nat Biotechnol. 25. Fleischmann R, Mease PJ, Schwartzman S, et al. Efficacy of
2008;26(1):127–32. tofacitinib in patients with rheumatoid arthritis stratified by
9. Maeshima K, Yamaoka K, Kubo S, et al. The JAK inhibitor background methotrexate dose group. Clin Rheumatol.
tofacitinib regulates synovitis through inhibition of interferon- 2017;36(1):15–24.
gamma and interleukin-17 production by human CD4 ? T cells. 26. Landewe RB, Connell CA, Bradley JD, et al. Is radiographic
Arthritis Rheum. 2012;64(6):1790–8. progression in modern rheumatoid arthritis trials still a robust
10. Boyle DL, Soma K, Hodge J, et al. The JAK inhibitor tofacitinib outcome? Experience from tofacitinib clinical trials. Arthritis Res
suppresses synovial JAK1-STAT signalling in rheumatoid Ther. 2016;18(1):212.
arthritis. Ann Rheum Dis. 2015;74(6):1311–6. 27. Strand V, van Vollenhoven RF, Lee EB, et al. Tofacitinib or
11. Conaghan PG, Ostergaard M, Bowes MA, et al. Comparing the adalimumab versus placebo: patient-reported outcomes from a
effects of tofacitinib, methotrexate and the combination, on bone phase 3 study of active rheumatoid arthritis. Rheumatology
marrow oedema, synovitis and bone erosion in methotrexate- (Oxford). 2016;55(6):1031–41.
naive, early active rheumatoid arthritis: results of an exploratory 28. Strand V, Kremer JM, Gruben D, et al. Tofacitinib in combina-
randomised MRI study incorporating semiquantitative and tion with conventional disease-modifying antirheumatic drugs in
quantitative techniques. Ann Rheum Dis. 2016;75(6):1024–33. patients with active rheumatoid arthritis: patient-reported out-
12. LaBranche TP, Jesson MI, Radi ZA, et al. JAK inhibition with comes from a phase III randomized controlled trial. Arthritis Care
tofacitinib suppresses arthritic joint structural damage through Res (Hoboken). 2017;69(4):592–8.
decreased RANKL production. Arthritis Rheum. 2012;64(11): 29. Strand V, Burmester GR, Zerbini CA, et al. Tofacitinib with
3531–42. methotrexate in third-line treatment of patients with active
13. Kitano M, Kitano S, Sekiguchi M, et al. Early effect of tofacitinib rheumatoid arthritis: patient-reported outcomes from a phase III
on osteoclast regulator in rheumatoid arthritis [abstract no. trial. Arthritis Care Res (Hoboken). 2015;67(4):475–83.
AB0394]. Ann Rheum Dis. 2016;75(Suppl 2):1040. 30. Strand V, Mysler E, Moots RJ, et al. Tofacitinib with and without
14. van Vollenhoven R, Choy E, Lee EB, et al. Tofacitinib, an oral methotrexate versus adalimumab with methotrexate for the
Janus kinase inhibitor, in the treatment of rheumatoid arthritis: treatment of rheumatoid arthritis: patient-reported outcomes from
changes in lymphocytes and lymphocyte subset counts and a phase 3b/4 randomized trial [abstract no. 1906]. Arthritis
reversibility after up to 8 years of tofacitinib treatment [abstract Rheumatol. 2017;69(Suppl 10).
no. THU0199]. Ann Rheum Dis. 2016;75(Suppl 2). 31. Strand V, Kremer J, Wallenstein G, et al. Effects of tofacitinib
15. McInnes IB, Kim HY, Lee SH, et al. Open-label tofacitinib and monotherapy on patient-reported outcomes in a randomized
double-blind atorvastatin in rheumatoid arthritis patients: a ran- phase 3 study of patients with active rheumatoid arthritis and
domised study. Ann Rheum Dis. 2014;73(1):124–31. inadequate responses to DMARDs. Arthritis Res Ther.
16. Isaacs JD, Zuckerman A, Krishnaswami S, et al. Changes in 2015;17:307.
serum creatinine in patients with active rheumatoid arthritis 32. Strand V, Lee EB, Fleischmann R, et al. Tofacitinib versus
treated with tofacitinib: results from clinical trials. Arthritis Res methotrexate in rheumatoid arthritis: patient-reported outcomes
Ther. 2014;16(4):R158. from the randomised phase III ORAL Start trial. RMD Open.
17. Winthrop KL, Silverfield J, Racewicz A, et al. The effect of 2016;2(2):e000308.
tofacitinib on pneumococcal and influenza vaccine responses in 33. Wollenhaupt J, Silverfield J, Lee EB, et al. Safety and efficacy of
rheumatoid arthritis. Ann Rheum Dis. 2016;75(4):687–95. tofacitinib, an oral janus kinase inhibitor, for the treatment of
18. Lee EB, Fleischmann R, Hall S, et al. Tofacitinib versus rheumatoid arthritis in open-label, longterm extension studies.
methotrexate in rheumatoid arthritis. N Engl J Med. J Rheumatol. 2014;41(5):837–52.
2014;370(25):2377–86. 34. Wollenhaupt J, Silverfield J, Lee EB. Tofacitinib, an oral janus
19. Fleischmann R, Kremer J, Cush J, et al. Placebo-controlled trial kinase inhibitor, in the treatment of rheumatoid arthritis: safety
of tofacitinib monotherapy in rheumatoid arthritis. N Engl J Med. and efficacy in open-label, long-term extension studies over
2012;367(6):495–507. 9 years [abstract no. 522]. Arthritis Rheumatol. 2017;69(Suppl
20. Fleischmann R, Mysler E, Hall S, et al. Efficacy and safety of 10).
tofacitinib monotherapy, tofacitinib with methotrexate, and adali- 35. van der Heijde D, Wollenhaupt J, Cohen SB, et al. Assessment of
mumab with methotrexate in patients with rheumatoid arthritis radiographic progression in patients with rheumatoid arthritis
(ORAL Strategy): a phase 3b/4, double-blind, head-to-head, ran- treated with tofacitinib: data from an open-label long-term
domised controlled trial. Lancet. 2017;390(10093):457–68. extension study over 3 years [abstract no. 533]. 2017;69(Suppl
21. van der Heijde D, Tanaka Y, Fleischmann R, et al. Tofacitinib 10).
(CP-690,550) in patients with rheumatoid arthritis receiving 36. Charles-Schoeman C, Burmester G, Nash P, et al. Efficacy and
methotrexate: twelve-month data from a twenty-four-month safety of tofacitinib following inadequate response to conven-
phase III randomized radiographic study. Arthritis Rheum. tional synthetic or biological disease-modifying antirheumatic
2013;65(3):559–70. drugs. Ann Rheum Dis. 2016;75(7):1293–301.
22. van Vollenhoven RF, Fleischmann R, Cohen S, et al. Tofacitinib 37. Hall S, Nash P, Rischmueller M, et al. Efficacy of tofacitinib in
or adalimumab versus placebo in rheumatoid arthritis. N Engl J patients who are inadequate responders to disease-modifying
Med. 2012;367(6):508–19. antirheumatic drugs according to early versus late duration of
Tofacitinib: A Review

rheumatoid arthritis: post-hoc analysis of data from phase 3 trials 53. Burmester GR, Pope JE. Novel treatment strategies in rheumatoid
[abstract no. 1609]. Arthritis Rheumatol. 2016;68(Suppl 10). arthritis. Lancet. 2017;389(10086):2338–48.
38. Dikranian A, Gonzalez-Gay MA, Wellborne F, et al. The efficacy 54. Eli Lilly Nederland B.V. OlumiantÒ (baricitinib): summary of
of tofacitinib in patients with rheumatoid arthritis stratified by product characteristics. 2017. http://www.ema.europa.eu/docs/
baseline body mass index [abstract no. 2371]. Arthritis en_GB/document_library/EPAR_-_Product_Information/human/
Rheumatol. 2017;69(Suppl 10). 004085/WC500223723.pdf. Accessed 31 Oct 2017.
39. Tesser J, Gül A, Olech E, et al. Efficacy and safety of tofacitinib 55. National Institute of Health and Care Excellence. Tofacitinib for
in patients with rheumatoid arthritis and inadequate response or moderate to severe rheumatoid arthritis: technology appraisal
intolerance to prior therapies [abstract no. 2493]. Arthritis guidance. 2017. https://www.nice.org.uk/guidance/ta480. Acces-
Rheumatol. 2017;69(Suppl 10). sed 31 Oct 2017.
40. Yamanaka H, Tanaka Y, Takeuchi T, et al. Tofacitinib, an oral 56. National Institute of Health and Care Excellence. Baricitinib for
Janus kinase inhibitor, as monotherapy or with background moderate to severe rheumatoid arthritis. 2017. https://www.nice.
methotrexate, in Japanese patients with rheumatoid arthritis: an org.uk/guidance/ta466. Accessed 31 Oct 2017.
open-label, long-term extension study. Arthritis Res Ther. 2016. 57. Mocsai A, Kovacs L, Gergely P. What is the future of targeted
https://doi.org/10.1186/s13075-016-0932-2. therapy in rheumatology: biologics or small molecules? BMC
41. An Y, Li Z, Wu Q. Efficacy and safety of tofacitinib in chinese Med. 2014;12:43.
patients with active rheumatoid arthritis: subgroup analysis from 58. Winthrop KL. The emerging safety profile of JAK inhibitors in
a phase 3 study of tofacitinib in combination with nonbiologic rheumatic disease. Nat Rev Rheumatol. 2017;13(4):234–43.
disease-modifying antirheumatic drugs [abstract no. AB0514]. 59. Strand V, Ahadieh S, French J, et al. Systematic review and meta-
Ann Rheum Dis. 2015;74(Suppl 2). analysis of serious infections with tofacitinib and biologic dis-
42. Radominski SC, Cardiel MH, Citera G, et al. Tofacitinib, an oral ease-modifying antirheumatic drug treatment in rheumatoid
Janus kinase inhibitor, for the treatment of Latin American arthritis clinical trials. Arthritis Res Ther. 2015;17:362.
patients with rheumatoid arthritis: pooled efficacy and safety 60. Winthrop KL, Yamanaka H, Valdez H, et al. Herpes zoster and
analyses of phase 3 and long-term extension studies. Reumatol tofacitinib therapy in patients with rheumatoid arthritis. Arthritis
Clin. 2017;13(4):201–9. Rheumatol. 2014;66(10):2675–84.
43. Cohen SB, Koenig A, Wang L, et al. Efficacy and safety of 61. Curtis JR, Xie F, Yun H, et al. Real-world comparative risks of
tofacitinib in US and non-US rheumatoid arthritis patients: herpes virus infections in tofacitinib and biologic-treated patients
pooled analyses of phase II and III. Clin Exp Rheumatol. with rheumatoid arthritis. Ann Rheum Dis. 2016;75(10):1843–7.
2016;34(1):32–6. 62. Vieira MC, Zwillich SH, Jansen JP, et al. Tofacitinib versus
44. Curtis JR, Schulze-Koops H, Takiya L, et al. Efficacy and safety biologic treatments in patients with active rheumatoid arthritis
of tofacitinib in older and younger patients with rheumatoid who have had an inadequate response to tumor necrosis factor
arthritis. Clin Exp Rheumatol. 2017;35(3):390–400. inhibitors: results from a network meta-analysis. Clin Ther.
45. Cohen SB, Tanaka Y, Mariette X, et al. Long-term safety of 2016;38(12):2628–41.e5.
tofacitinib for the treatment of rheumatoid arthritis up to 63. Bergrath E, Gerber RA, Gruben D, et al. Tofacitinib versus
8.5 years: integrated analysis of data from the global clinical biologic treatments in moderate-to-severe rheumatoid arthritis
trials. Ann Rheum Dis. 2017;76(7):1253–62. patients who have had an inadequate response to nonbiologic
46. European Medicines Agency. Xeljanz (tofacitinib): assessment DMARDs: systematic literature review and network meta-anal-
report. 2017. http://www.ema.europa.eu/docs/en_GB/document_ ysis. Int J Rheumatol. 2017;2017:8417249.
library/EPAR_-_Public_assessment_report/human/004214/ 64. Taylor PC, Keystone EC, van der Heijde D, et al. Baricitinib
WC500224913.pdf. Accessed 31 Oct 2017. versus placebo or adalimumab in rheumatoid arthritis. N Engl J
47. Pope J, Keystone E, Jamal S, et al. Persistence of tofacitinib in Med. 2017;376(7):652–62.
the treatment of rheumatoid arthritis in open-label, long-term 65. Weinblatt M, Taylor PC, Burmester GR, et al. Cardiovascular
extension studies up to 8 years [abstract no. 1602]. Arthritis safety during treatment with baricitinib in rheumatoid arthritis
Rheumatol. 2016;68(Suppl 10). [abstract no. 2352]. Arthritis. Rheumatol. 2017;69(Suppl
48. Winthrop K, Wouters A, Choy E, et al. Assessment of 10):2352.
immunogenicity of live zoster vaccination in rheumatoid arthritis 66. Eli Lilly and Co Ltd. Olumiant (baricitinib): UK summary of
patients on background methotrexate before and after initiating product characteristics. 2017. https://www.medicines.org.uk/emc/
tofacitinib or placebo [abstract no. FRI0110]. Ann Rheum Dis. medicine/32997#. Accessed 31 Oct 2017.
2016;75(Suppl 2):468. 67. Souto A, Maneiro JR, Gomez-Reino JJ. Rate of discontinuation
49. Nurmohamed M, Choy E, Charles-Schoeman C, et al. Impact of and drug survival of biologic therapies in rheumatoid arthritis: a
tofacitinib treatment compared with placebo or methotrexate on systematic review and meta-analysis of drug registries and health
cardiovascular risk scores in six phase 3 randomized controlled care databases. Rheumatology (Oxford). 2016;55(3):523–34.
trials [abstract no. 2966]. Arthritis Rheumatol. 2017;69(Suppl 68. Smith T, Harnett J, Gruben D, et al. Real-world experience with
10). tofacitinib versus adalimumab and etanercept in biologic-naive
50. Schulze-Koops H, Strand V, Nduaka C, et al. Analysis of patients with RA previously treated with methotrexate: data from
haematological changes in tofacitinib-treated patients with a US administrative healthcare insurance claims database [ab-
rheumatoid arthritis across phase 3 and long-term extension stract no. 2831]. Arthritis Rheumatol. 2017;69(Suppl 10).
studies. Rheumatology (Oxford). 2017;56(1):46–57. 69. Marengo MF, Suarez-Almazor ME. Improving treatment adher-
51. Mariette X, Chen C, Biswas P, et al. Lymphoma in the tofacitinib ence in patients with rheumatoid arthritis: what are the options?
rheumatoid arthritis clinical development program. Arthritis Care Int J Clin Rheumtol. 2015;10(5):345–56.
Res (Hoboken). 2017. https://doi.org/10.1002/acr.23421. 70. Alten R, Kruger K, Rellecke J, et al. Examining patient prefer-
52. Curtis JR, Lanas A, John A, et al. Factors associated with gas- ences in the treatment of rheumatoid arthritis using a discrete-
trointestinal perforation in a cohort of patients with rheumatoid choice approach. Patient Prefer Adherence. 2016;10:2217–28.
arthritis. Arthritis Care Res (Hoboken). 2012;64(12):1819–28.

You might also like