ArticlePDF Available

Januskinase Inhibitors to Treat Rheumatoid Arthritis: Real World Data Match Clinical Trial Results. An Evaluation by BioReg, the Austrian Registry for Biologicals, Biosimilars, and Targeted Synthetic DMARDS in the Treatment of Inflammatory Rheumatic Diseases

Authors:
  • Rheumatological office
  • RHZ Oberlaa

Abstract and Figures

Background: Whether the safety and efficacy of Januskinase Inhibitors (JAKis) in daily routine treatment of
Content may be subject to copyright.
Januskinase Inhibitors to Treat Rheumatoid Arthritis: Real World Data Match
Clinical Trial Results. An Evaluation by BioReg, the Austrian Registry for
Biologicals, Biosimilars, and Targeted Synthetic DMARDS in the Treatment of
Inflammatory Rheumatic Diseases
Burkhard F. Leeb1,2,3*, Peter Spellitz3,4, Gabriele Eichbauer-Sturm3,5, Manfred Herold3,6, Miriam
Stetter3,7, Rudolf Puchner3,8, Franz St. Singer3, Ruth Fritsch-Stork3,9,10
1Department of Internal Medicine-Rheumatology, Karl Landsteiner Private University for Health Sciences, Krems, Austria;
2Department of Rheumatology, Office and Karl Landsteiner Institute for Clinical Rheumatology, Hollabrunn, Austria; 3BioReg
(Austrian Registry for Biologicals, Biosimilars and tsDMARDs in Rheumatology); Vienna, Austria; 4Department of Out-Patient,
Rheumazentrum Wien Oberlaa Center, Vienna, Austria; 5Department of Rheumatology and Nephrology, Office, Linz, Austria;
6Department of Rheumatology, Autoimmune Laboratory Medical University-Innsbruck, Innsbruck, Austria; 7Department of
Rheumatology, Office, Amstetten, Austria; 8Department of Rheumatology and Gastroenterology, Office, Wels, Austria; 9Department
of Rheumatology, Sigmund Freud Private University, Vienna, Austria; 10Department of Medical Hanusch Hospital, Rheumatology
and Ludwig Boltzmann Institute for Osteology, Vienna, Austria
ABSTRACT
Background: Whether the safety and efficacy of Januskinase Inhibitors (JAKis) in daily routine treatment of
Rheumatoid Arthritis (RA) match the respective clinical trial results is of crucial clinical interest, however, growing,
but still limited corresponding evidence is available.
Patients and methods: Data, including adverse events, disease activity scores, patient-related outcomes, and response
as well as persistence rates, of all RA patients receiving JAKi treatment registered in the BioReg database were
retrieved. The results were described according to those of pivotal clinical trials of JAKis. Analyses were performed using
descriptive statistics, such as mean value comparisons, and by drawing patient trajectories.
Results: One-hundred-and twenty-two patients (mean age 64 years; 83.9% female, 60.5% Rheumatoid Factor (RF)
positive; n=74 receiving Baricitinib and n=48 receiving Tofacitinib) were included from 2017 on. Significant
differences occurred in the initial disease activity (mean Disease Activity Score including 28-joint-count (DAS28) 6.5
versus 3.8) between the trials and real-world data. In the registry, an insufficient response was observed in 24 (32.4%)
and 21 (43.8%) patients, adverse events occurred in 16 (21.6%), and 12 (25.0%); the mean duration of treatment was
1.34 yrs. and 1.5 years. in patients receiving Baricitinib and Tofacitinib, respectively. The response rates were like
those of the clinical trials, whereas adverse events were less frequently reported in the registry and no safety signals
occurred. All scores applied showed a positive course and the results of mean value analysis and patient trajectories
demonstrated the benefits of JAKis; the health assessment questionnaire values remained relatively stable, in contrast
to the trial results.
Conclusion: Real World data for JAKis comply to a high extent with clinical trial results. This may contribute to an
increased confidence in the therapeutic status of these drugs.
Rheumatology: Current Research Research Article
Correspondence to: Burkhard F. Leeb, Department of Internal Medicine-Rheumatology, Karl Landsteiner Private University for Health Sciences,
Krems, Austria, Tel: +436644165686; E-mail: burkhard.leeb@stpoelten.lknoe.at/leeb.rheuma@aon.at
Received: June 02, 2021; Accepted: June 16, 2021; Published: June 23, 2021
Citation: Leeb BF, Spellitz F, Sturmc GE, Herold M, Stetter M, Puchner R, et al. (2021) Januskinase Inhibitors to Treat Rheumatoid Arthritis: Real
World Data Match Clinical Trial Results an Evaluation by BioReg, the Austrian Registry for Biologicals, Biosimilars, and Targeted Synthetic
DMARDS in the Treatment of Inflammatory Rheumatic Diseases. Rheumatology (Sunnyvale). 11.288.
Copyright: © 2021 Leeb BF, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which
permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Rheumatology (Sunnyvale), Vol.11 Iss.5 No:1000288 1
Keywords: Rheumatoid Arthritis (RA); Januskinase Inhibitors (JAKis); Baricitinib; Clinical trial results
INTRODUCTION
Rheumatoid Arthritis (RA), the most common chronic
inflammatory disease, is a progressive, systemic autoimmune
disease characterized by inflammation of the synovial
membrane. This inf lammation, aside from pain, stiffness,
swelling, and loss of function, may also cause bone erosion,
ultimately leading to irreversible joint destruction and disability
[1]. Other systemic symptoms of RA include fatigue, anemia,
and osteoporosis, and RA patient’s the overall risk to develop,
example, cardiovascular diseases and infection is considerably
increased [2,3]. Over the last decades, major advances in RA
treatment have been achieved [4], due to the development of
new biologic Disease-Modifying Drugs
Factor-alpha (TNF- α
Meeting the obvious medical need for further effective
treatments, a different therapeutic approach, targeting
intracellular activation pathways, particularly Janus Kinases
(JAKs), has been applied. JAK-Inhibitors (JAKis) have been
shown to be more efficacious than methotrexate (MTX) and/or
anti-TNF-a agents in clinical trials; providing an alternative for
patients with an inadequate clinical response to biologic or non-
biologic Disease Modifying Anti Rheumatic Drugs (DMARDs)
[8]. Studies with the first two JAKis, Tofacitinib (Tofa) and
Baricitinib (Bari) reimbursed in Austria since October 2017, and
Baricitinib (Bari), reimbursed in Austria since August 2018, have
demonstrated efficacy in several RA patient groups either as
monotherapy or in combination with MTX and Adalimumab,
as a comparator [9]. Those studies lead to a licensing of Bari and
Tofa as monotherapy or in combination with MTX in patients
with moderate-to-severe RA not adequately responding to one or
more DMARDs [10,11].
At the time point this evaluation was carried out, almost no
Real-World Evidence (RWE) for the application of both drugs
was available, which could allow a better understanding of the
JAKis treatment impact in more diverse patient populations.
Meanwhile, a feast of papers from different countries and
cultures dealing with this topic have been published, providing
evidence important for the clinically working rheumatologist,
whether the risk/benefit ration of Januskinase Inhibitors (JAKis)
in daily routine treatment of Rheumatoid Arthritis (RA) can be
regarded reasonable, and which patient groups could expect
benefits from JAKI treatment. However, most of the papers deal
with different questions, in part comparing JAKis with
biological, or giving information about treatment strategies;
similarly to our investigations, the sample sizes are relatively
small, even from big countries like Germany and Italy [12-16].
The objectives of the present evaluation of these registry data
were to evaluate the safety and efficacy of JAKis in routine
treatment of RA in real-life as well as to investigate whether real-
life data match the respective clinical trial results to increase the
knowledge about the position of JAKis in routine therapy of RA
and possibly create a hypothesis for future studies. This paper
presents the respective results from Austria, a relatively small
country with a highly developed health care system, practically
providing access to the drug to all patients who need.
PATIENTS AND METHODS
Real-life data of Austrian RA patients treated with biologicals,
Biosimilars, and Targeted Synthetic DMARDs (tsDMARDs)
including JAKis have been collected in the BioReg Project
database. BioReg, the Austrian registry for patients with chronic
rheumatic diseases treated with these agents, was established in
2010. Meanwhile, more than 3100 patients from 28 centers have
been enrolled into this database. The RA data core set
encompasses joint counts, the Disease Activity score including an
28-joint count (DAS28-ESR), the Clinical Disease Activity Index
(CDAI), and the Health Assessment Questionnaire (HAQ-DI),
additionally, the Rheumatoid Arthritis Disease Activity Index-5
(RADAI-5) [17-21]. Moreover, CRP, ESR, RF, and anti-CCP are
recorded, as well as comorbidities and adverse events. Female or
male patients, treated with Biologicals, Biosimilars or
tsDMARDs, older than 18 years, personally independent with
inflammatory rheumatic diseases, such as RA, Psoriatic
Arthritis, Ankylosing Spondylitis, and other diseases treated
with Biologicals, Biosimilars and tsDMARDs, according to the
licensed indications or generally accepted "off label” use can be
included in BioReg. Personal dependence and withdrawal of
informed consent are the only exclusion criteria. The ethics
committee of Lower Austria has approved the study design of
BioReg (reference number GS4-EK-085-2009) in 2009, and this
approval is renewed annually [6]. All patients gave their written
informed consent before inclusion into the registry. Data
collection in BioReg started in 2010. For this study with a
deadline of May 31st, 2019, the data of all RA patients receiving
Bari or Tofa in the BioReg database were retrieved. Patient
characteristics, including the most prevalent comorbidities, are
given in detail in Tables 1 and 2. According to the primary goal
of the registry, all safety issues and adverse events are
documented, collected, and evaluated, not limited to treatment
withdrawal. These are listed in Tables 3 and 4.
Personal data Baricitinib (n=74) Tofacitinib (n=48)
Age (years; min-max) 64 (26-84) 64 (35-89)
Female sex 80.30% 87.50%
RF pos 60.50% 60.40%
Leeb BF, et al.
Rheumatology (Sunnyvale), Vol.11 Iss.5 No:1000288 2
efficacy in an additional number of
(bDMARDS) with impro-
ved patients compared to
conventional treatments, Conventional Synthetic DMARDs
(csDMARDs), resulting in the change of
However, despite all advances, a
[5].
patients fail to achieve an acceptable
treatment recommend-
ations considerable propor-
tion of clinical response or
drug tolerance to the available treatments [6], including biologic
compounds targeting Tumor Necrosis ),
Interleukin-6 receptor (IL-6r), Interleukin1 (IL-1), the co-stimul-
ation pathway, or B-cells [6,7].
ACPA pos. 52,8% 57,6%
Initial DAS28-ESR
(mean ± SD)
3.83 ( ± 1.2) 3.80 ( ± 1.4)
Height (cm, min-max) 166 (150-182) 167 (153-183)
Weight (kg, min-max) 74,7 (45-130) 71.4 (43-119)
Duration (years;
mean ± SD)
14.1 ( ± 9.7) 12.9 ( ± 9.39)
Biologic naive (n, %) 14 (19%) 8 (17%)
≤ 2 biologics (n, %) 32 (43%) 34 (71%)
>2 biologics (n, %) 28 (38%) 6 (12%)
Co-morbidities (n, %) 48 (65%) 34 (71%)
Table 1: Demographic data of the JAKI treated patients in
BioReg.
Co-morbidities Baricitinib (n=48, multiple answers possible)
No co-morbidity recorded 26
Vascular diseases 15
Endocrine diseases 14
Heart diseases 12
Bone diseases 8
Psychiatric diseases 6
Pulmonary diseases 6
Gastrointestinal diseases 1
Co-morbidities Tofacitinib (n=34, multiple answers possible)
No co-morbidity recorded 14
Heart diseases 12
Endocrine diseases 12
Vascular diseases 11
Bone diseases 5
Psychiatric diseases 4
Pulmonary diseases 1
Gastrointestinal diseases 1
Table 2: Patients’ comorbidities.
BioReg patients Baricitinib (n=74) Tofacitinib (n=48)
Duration of therapy
(median, min-max)
1.25 yrs. (0.25-2.5) 1.25 yrs. (0.25-2.5)
Discontinuation of
therapy (n, %)
13 (17.6%) 14 (29.2%)
Insufficient response*
(n, %)
24 (32.4%) 21 (43.8%)
Adverse events* (n,
%)
16 (21.6%) 12 (25.0%)
*Not all leading to discontinuation of treatment
Table 3: Therapy characteristics of patients in BioReg.
AEs Baricitinib (n=16)
Infections and parasitic diseases 6
Hematological diseases 2
Ear and labyrinthic diseases 2
Endocrine diseases 1
Genitourinary and breast diseases 1
Gastrointestinal diseases 1
Neurological disorders 1
Vascular diseases 1
Heart diseases 1
AEs Tofacitinib (n=12)
Infections and parasitic diseases 5
Gastrointestinal diseases 2
Ear and labyrinthic diseases 1
Skin diseases 1
Neurological disorders 1
Vascular diseases 1
Neoplasms 1
Table 4: Adverse events in JAKi treated patients (MedRA-SOC
terms).
In Austria, LAKIs are licensed for RA patients with moderate
disease activity as expressed by an accepted disease activity scale
(example, DAS28>3.2) after failure of at least one biological.
Bari and Tofa are reimbursed in Austria since October 2017 and
August 2018, respectively, in case of failure to one bDMARD. For
this study, the following scores were retrieved from the registry:
Disease Activity score including a 28-joint count (DAS28-ESR),
Leeb BF, et al.
Rheumatology (Sunnyvale), Vol.11 Iss.5 No:1000288 3
Clinical Disease Activity Index (CDAI), and Health Assessment
Questionnaire (HAQ-DI). Additionally, the Rheumatoid
Arthritis Disease Activity Index-5 (RADAI-5) was documented
[12-16]; not all instruments are applied in each patient, as the
instruments used depend on the participant’s routine practice.
Safety data were gathered according to MedDRA definitions and
are depicted in a descriptive manner.
The registry comprised 1,408 patients with rheumatoid arthritis
at the time of data retrieval, of which 74 patients were treated
with Baricitinib and 48 patients with Tofacitinib together
amounting to 8.7% of all RA-patients included in the registry.
For analytic purposes, patient trajectories (“Spaghetti-blots”),
depicting the course of the disease in an individual patient and
mean value comparisons on a group level (as a conservative
approach) were performed [22]. Propensity score matching with
clinical trial data from the RA-BEACON study with Bari and
the ORAL Step trial with Tofa [23-25] would have been a more
stringent analysis, however, as we had no access to the raw data
of the clinical trials, it was not possible to perform such an
analysis. Thus, the results of the analysis of data obtained from
BioReg were described in comparision data obtained from clini-
cal trials with JAKis. Due to the inherent differences of data
stemming from controlled trials versus registry data in terms of,
among others, patient characteristics, timepoints, and frequency
of control visits as well as the stringency of data completion, a
descriptive rather than a statistical comparison was carried out
and should serve to generate a hypothesis.
RESULTS
Seventy-four patients were treated with Bari, their mean age was
64 (26-84) years, 80.3% were female, and 60.5% were positive
for Rheumatoid Factor (RF) and 52.8% for Antibodies against
Citrullinated Protein (ACPA). Forty-eight patients were treated
with Tofa, their mean age was 64 (range: 35-89) years, 87.5%
were female, and 60.4% were RF and 57.6 were ACPA positive.
JAKi treatment was initiated preferentially after one or two
unsuccessful treatment attempts with biologics, namely, in 32
(43%) Bari treated patients, and in 34 (71%) patients treated
with Tofa respectively, which is in line with the Austrian
reimbursement regulations for these drugs. Fourteen pts (19%)
received Baricitinib as their first treatment after csDMARDS
failure, and 8 pts (17%) Tofa, respectively, see in Table 1. Co-
morbidities were evident in 48 pts on Bari (65%) and in 34 pts
on Tofa (73%). The most prevalent comorbidities were vascular,
endocrine, and heart diseases in Table 2.
The median treatment duration was 1.25 (0.25-2.5) years for
both medications, [R] also the withdrawal rates (Bari 17.6%,
Tofa 29.2%) were similar. After one year of treatment,
approximately 80% of the patients were found still on JAKi
therapy in Table 3. Safety data of the BIOREG registry were
recorded according to the MedDRA classification system and
are depicted in Table 4. Most frequent were infectious/parasitic
diseases with a cumulative incidence of 8.1% for Bari and 10.4%
for Tofa. According to the SOC terms, vascular disease occurred
in one patient each on Bari or Tofa, and neoplasms were noted
in one patient on Tofa Table 4. An insufficient response as
assessed by the physician’s personal global evaluation was
observed in 24 (32.4%) and 21 (43.8%) patients and adverse
events occurred in 16 (21.6%) and 12 (25.0%) patients receiving
Bari and Tofa, respectively Table 3 and Table 4. During the
observation period up until the writing of this manuscript, none
of the patients died or developed deep venous thrombosis. The
DAS28-ESR was available for 50 pts. on Bari and 42 on Tofa,
CDAI for 42/33, HAQ-DI for 57/40 pts., and RADAI-5 for
41/44 pts., respectively.
The mean baseline DAS28 was 3.83 ± 1.21 and 3.80 ± 1.41,
respectively, for the Bari- and the Tofa-treated patients, with
corresponding CDAI values of 15.7, and 20.3 ± 14.4. The
respective RADAI-5 values were 4.4 ± 2.2 and 4.2 ± 2.4). All
disease activity score values indicate moderate disease activity at
the time of JAKi initiation, with mean HAQ values of 1.1 ± 0.7
and 1.2 ± 0.8 for the Bari- resp. Tofa-treated patients in Figures
1a and 1b. During the treatment period, all parameters showed
improvement, which is demonstrated by the course of DAS28
for both drugs as, shown in Figures 2a and 2b. Mean analyses
and patient trajectories, which outline the individual courses,
showed the beneficial effects of JAKis on the disease course up
to 24 weeks for Bari and 18 weeks for tofa. The analysis of the
CDAI and RADAI-5 courses was similar, apart from some
individual fluctuations as shown in Figure 3a and 3b.
Figure 1: DAS28 of the Baricitinib-treated patients. (a)
Trajectories showing the individual courses; (b) Conventional
bar graphs depicting mean values.
Figure 2: DAS28 of the Tofacitinib-treated patients. (a)
Trajectories showing the individual courses; (b) Conventional
bar graphs depicting the mean values.
Figure 3: Patient trajectories for the RADAI-5 (a) in the
Baricitinib (b) in the Tofacitinib treated patients.
Leeb BF, et al.
Rheumatology (Sunnyvale), Vol.11 Iss.5 No:1000288 4
Comparison with Bari in the RA-BEACON trial
The RA-BEACON trial was performed as a 6-month phase 3
clinical trial investigating the efficacy and safety of Bari in
combination with csDMARDS in patients not adequately
responding to prior therapy with a biological. This situation very
much mirrors the characteristics of patients in BioReg treated wi-
th Bari, which was also the case in the Austrian patients included
in the RA-BEACON study. [24]. In BioReg, 81% of the 74
patients treated with Bari had a treatment history of one or
more biologics. Therefore, we compared the registry data with
the patient group (n=177) treated with 4 mg Bari/day from the
RA-BEACON trial [24]. Whereas sex distribution and disease
duration were similar, patients in the BioReg were older (64 ±
12 vs.
BioReg displayed less severe disease activity mean
DAS28-ESR: 3.83 ± 1.2 vs. 6.6 ± 1.1, mean HAQ 1.1 ± 0.7) vs.
1.7 ± 1.6 compared to those in the RA-BEACON trial. The
mean DAS28 and HAQ reductions at month six in the RA-
BEACON trial were 1.7 and 0.4, respectively [24], whereas in
BioReg, the respective reduction of DAS28 was 1.1, and the
mean HAQ was found unchanged after six months. In the RA-
BEACON trial, 88% of the patients completed the 6-month
treatment, whereas in BioReg, 82.4% of the patients remained
on Bari throughout the observation period, which lasted for
1.25 (0.25-2.5) years. The response rate, although difficult to
compare in the clinical trial, was primarily assessed with the
ACR criteria [26], whereas that in BioReg above all was assessed
according to the physician’s discretion; 67.6% of the patients in
BioReg were assessed to have a positive response, whereas in the
RA-BEACON trial, only 30% achieved an ACR response of
50%. For the sake of better comparability, we also compared the
percentage of patients achieving a DAS28ESR<3.2 at week 24.
In BioReg, 48% (15 of 31) patients achieved the low disease
activity threshold, while in the trial this percentage was 26%.
Adverse events were more frequently reported in the clinical
trial than in the registry (77% vs. 21.6%) and no new signals
with respect to AEs were observed.
Comparison with Tofa in the ORAL-step trial
This study was performed as a 6-month phase 3 clinical trial
investigating the efficacy and safety of Tofa in combination with
MTX in patients with insufficient response to TNF inhibitors.
This was also the case in 83% of patients BioReg treated with
Tofa, suggesting similar patient populations in BioReg and the
patient group (n=122) treated with 5 mg Tofa/twice a day in the
ORAL step trial [25]. Gender distribution, RF positivity, and
disease duration were similar in both groups, whereas the
patients in BioReg were older (64 ± 12.2 vs. 55.1 ± 11.3) years
and less active in their disease (DAS28-ESR 3.8 ± 1.4 vs. 6.5 ±
1.1, HAQ: 1.2 ± 0.8 vs. 1.6 ± 0.7) than those in the trial. The
mean DAS28 and HAQ reductions at 6 months in ORAL step
trial were 1.6 and 0.5, respectively [25], in contrast to 1.0 and
0.3 in BioReg. In the ORAL step trial, 80.5% of the patients
completed the 6-month treatment, whereas, in BioReg, 70.8%
of the patients remained on Tofa throughout the median
observation period of 1.25 (0.25-2.5) years. Whereas 51.1% of
the patients achieved an ACR response of 50% in the ORAL
step one, 56.2% of the patients were evaluated by their treating
physicians to have a sufficient response in BioReg. A
DAS28ESR<3.2 at week 24 was achieved by 44% (12 of 27) in
BioReg, in contrast to 25% in the trial. Adverse events were
more frequently reported in the clinical trial than in the registry
(42.9% vs. 25.0%). It is important to note that no new or
unexpected adverse events occurred in the patients in BioReg.
DISCUSSION
The JAKis Bari and Tofa rapidly and very successfully found their
position in the therapeutic armamentarium for RA [8]. This is
also apparent in Austria with 8.7% of RA patients enrolled in
BioReg two and a half years after licensing. In terms of gender
and disease duration, patients in BioReg correspond to the
patient population of the pivotal RA-BEACON and ORAL
STEP ONE trials, however, BioReg patients are older, suffer
from a higher number of comorbidities, and display a lower
disease activity than the clinical trial cohorts. Altogether, data
from the BioReg registry paint a reassuring picture of safety and
affectivity similar/better to the ones in the pivotal trials. To
translate data from successful clinical trials acquired from a
highly selected patient population into clinical practice, data
from registries, which include patients as comprehensively as
possible, are important [27,28]. Due to the relative recent
approval of Baricitinib and Tofacitinib by the European
Medicines Agency in 2017, registry data pertaining to JAKi from
Europe are limited and have mostly been presented at meetings
[29] However, due to earlier approval in other states, sporadic
real-life data have been published, e.g. from the US, Australia or
Switzerland [22] The main scope of these reports was a
comparison with other bDMARDs, in which the non-inferiority
of JAKi compared to anti TNF known from clinical trials was
underscored [12-14]. Mueller et al., on the other hand,
investigated the safety of Tofa assessed by the reason to stop the
medication and laboratory abnormalities. Again, clinical trial
data were corroborated, and no new relevant safety signals were
described [13]. The present report from the BioReg registry
differs from most publications due to the comprehensive
description of patient characteristics, safety data, and
effectiveness data. Furthermore, the direct reference between
clinical trial data and BioReg data provides additional insight
beyond the data provided from other registries. Here we show
that the efficacy of JAKi not only applies to the clinical trial
population of patients in their mid 50ies with high disease
activity but is used safely and effectively in patients>60 years old
with only moderate disease activity reflected in the lower DAS28
values in BioReg vs. trials with Bari and Tofa [24,25]. In Austria,
in contrast to many other countries, biological and tsDMARDs
are usually initiated at this (rather low) disease activity stage [6].
The role of high DAS28 as a predictor of clinical response has
been acknowledged in several trials of biological therapy [29,
30]. However, also in registry data with lower initial disease
activity values, a significant improvement could generally be
observed [31] a positive contribution of a lower baseline DAS28
to the achievement of low activity has also been observed in a
Japanese report presented at the EULAR 2020 [32].
Altogether, patients in the trials as well as in the registry finally
developed a low moderate disease activity on average, which was
Leeb BF, et al.
Rheumatology (Sunnyvale), Vol.11 Iss.5 No:1000288 5
56 ± 11 years and less likely to be RF positive (60.6%
Patients in
±
72%).
demonstrated in several disease scores. The BioReg data,
also included the RADAI-5, not applied in the trials, which also
mirrored the course of the composite indexes [15]. Additionally,
visualizing the individual disease activity score courses by patient
trajectories using BioReg revealed a beneficial course in most of
the patients, despite some fluctuations. Patient trajectories may
allow better insight into the course of the disease and may offer
the opportunity to better estimate the individual patient’s
chance for success [17]. The improvement of the HAQ values
was less prominent in the registry than in the clinical trials, with
lower baseline HAQ values, comparable disease duration despite
higher mean age in the BioReg patients. Apart from the lower
disease activity, which may have contributed to the lower HAQ
score, the sociocultural variety in the participants of
multinational trials may have played a role. At the end of the
day, the JAKis proved to be effective to a remarkably similar
extent, although without formal statistical comparison, in
clinical trial population and the real world setting of the
Austrian registry.
As expected, adverse events were more frequently reported in
the clinical trial than in the registry (77% vs. 21.6%). Bari and
Tofa were found to be well tolerated by the patients in the
registry with an overall incidence of infection of approximately
10% as the most frequent adverse event. To our knowledge, this
is the first report of any infection derived from registry data, and
it implicates no new safety signals in the use of JAKi in real life.
However, the strikingly higher rate of adverse events in the trials
vs. the registry is evident and can-at least partly-be explained by
the rigorous monitoring procedures of clinical trials, which
cannot be guaranteed in registries.
There are some shortcomings of this study. The differences
between clinical trials and BioReg are evident: In the registry, no
close monitoring is performed; data are collected with differing
frequencies and extent. However, the registries reflect the real-
life setting, considering the above-mentioned shortcomings. The
number of included patients in our Austrian registry is smaller
compared to globally performed clinical trials or some other
registries, but the evidence published in that respect is also
founded on relatively small patient numbers [13,15]. However,
the advantage of our registry data is the homogenous patient
population with little sociocultural differences, a highly
developed social insurance system, the unusual older age, and
low disease activity of the BioReg patients. A caveat is the
inclusion of all patients treated with JAKis, and not only
biological non-responders [33]. However, we believe that
following the new recommendations [34], JAKis will increasingly
be used in biological naïve patients, justifying the inclusion of
these patients.
Large, non-interventional observations, such as registries, seek to
include as many patients as possible irrespective of
comorbidities, age, disease activity, or other confounding
factors, in contrast to the highly selected patient population of
clinical investigations, to reflect daily practice as precisely as
possible. Therefore, it is of particular importance for clinical
practice to undertake efforts to verify or falsify the results of
clinical trials by real-world evidence, which has been just recently
strengthened by the German Institute for Quality and Economy
in Health Care (IQWiG) [35]. Of course, propensity matching
had constituted a method to achieve more robust results.
However, the nature of the registry data and the impossibility to
access the clinical trials’ raw data made it seem appropriate not to
perform other than our primarily descriptive analysis. Registry
data may serve as a possibility to generate hypotheses for future
research. Such studies should comprise as many real-world data
as possible as well as raw data from clinical trials with a
propensity score analysis to validate our observations so that the
efficacy and safety of JAKis is like clinical trial results and real-
world data in RA.
CONCLUSION
In conclusion, for the Austrian patients enrolled in BioReg, the
application of JAKis in clinical practice leads to results
comparable to those of clinical trials, even though the patient
population was older and had lower disease activity. This may
contribute to an increased confidence of the treating physicians
in the therapeutic properties of these drugs.
ETHICS APPROVAL
The ethics committee of Lower Austria has approved the study
design of BioReg (reference number GS4-EK-085-2009) in 2009,
and this approval is renewed annually (most recently
02/14/2020).
DISCLOSURES
BFL Clinical trials: TRB, Roche, Consultancies: AbbVie,
Amgen, Roche, MSD, Pfizer, Celgene, Grünenthal, Kwizda, Eli-
Lilly, Novartis, Sandoz.
Speakers ‘bureau: AbbVie, Roche, MSD, Pfizer, Actiopharm,
Boehringer-Ingelheim, Kwizda, Celgene, Sandoz, Grünenthal,
Eli-Lilly.
RP: Consultancies: AbbVie, Amgen, Pfizer, Celgene,
Grünenthal, Eli-Lilly; Speakers ‘bureau: AbbVie, BMS, Janssen,
Kwizda, MSD, Pfizer, Celgene, Grünenthal, Eli-Lilly, PS, GES,
MS, MH, FStS, RFS declared no conflicts of interest.
FUNDING
BioReg is a non-profit organization supported by ABBVIE
GmbH; AMGEN GmbH, CELGENE GmbH; BIOGEN
GmbH, ELI-LILLY GmbH, FRESENIUS-KABI Austria GmbH;
JANSSEN-CILAG Austria GmbH, MERCK SHARP and
DOHME GmbH; PFIZER CORPORATION AUSTRIA
GmbH, ROCHE AUSTRIA GmbH; SANDOZ Pharma GmbH,
UCB Pharma GmbH through an unrestricted educational grant.
This study was supported by ELI-LILLY GmbH, Austria.
AUTHOR CONTRIBUTIONS
BFL: Contributed to data acquisition, design of the study, data
analysis, manuscript writing.
PS, GES, MH, MS, RP: Data acquisition, data analysis, study
design.
Leeb BF, et al.
Rheumatology (Sunnyvale), Vol.11 Iss.5 No:1000288 6
FSS, RFS: Data analysis, study design, manuscript writing.
REFERENCES
1. McInnes IB, Schett G. Pathogenetic insights from the treatment of
rheumatoid arthritis. Lancet. 2017;389(10086):2328-2337.
2. Hochberg MC, Johnston SS, John AK. The incidence and prevalence
of extra-articular and systemic manifestations in a cohort of newly-
diagnosed patients with rheumatoid arthritis between 1999 and 2006.
Curr Med Res Opin. 2008;24(2):469-480.
3. Coates LC, FitzGerald O, Helliwell PS, Paul C. Psoriasis, psoriatic
arthritis, and rheumatoid arthritis: Is all inflammation the same?
Semin Arthritis Rheum 2016;46(3):291-304.
4. Burmester GR, Pope JE. Novel treatment strategies in rheumatoid
arthritis. Lancet. 2017;389(10086):2338-2348.
5. Sparks JA. Rheumatoid arthritis. Ann Intern Med
2019;170(1):ITC1-ITC16.
6. Rintelen B, Zwerina J, Herold M, Singer F, Hitzelhammer J, Halder
W, et al. Validity of data collected in BIOREG, the Austrian register
for biological treatment in rheumatology: Current practice of
bDMARD therapy in rheumatoid arthritis in Austria. BMC
Musculoskelet Disord. 2016;17(1):1-8.
7. Singh JA, Saag KG, Bridges Jr SL, Akl EA, Bannuru RR, Sullivan MC,
et al. 2015 American College of Rheumatology guideline for the
treatment of rheumatoid arthritis. Arthritis Rheumatol. 2016;68(1):
1-26.
8. Taylor PC. Clinical efficacy of launched JAK inhibitors in rheumatoid
arthritis. Rheumatology. 2019;58(1):i17-i26.
9. Emery P, Pope JE, Kruger K, Lippe R, DeMasi R, Lula S, et al. Efficacy
of monotherapy with biologics and JAK inhibitors for the treatment of
rheumatoid arthritis: A systematic review. Adv Ther. 2018;35(10):
1535-1563.
10. Olumiant. Utrecht, the Netherlands: Eli Lilly Nederland BV. 2019.
11. Xeljanz. United Kingdom: Pfizer Limited. 2019.
12. Reed GW, Gerber RA, Shan Y, Takiya L, Dandreo KJ, Gruben D, et
al. Real-world comparative effectiveness of Tofacitinib and tumor
necrosis factor inhibitors as monotherapy and combination therapy
for treatment of rheumatoid arthritis. Rheumatol Ther. 2019;6(4):
573-586.
13. Mueller RB, Hasler C, Popp F, Mattow F, Durmisi M, Souza A, et al.
Effectiveness, tolerability, and safety of tofacitinib in rheumatoid
arthritis: A retrospective analysis of real-world data from the St. Gallen
and Aarau cohorts. J Clin Med. 2019;8(10):1548.
14. Bird P, Littlejohn G, Butcher B, Smith T, da Fonseca Pereira C,
Witcombe D, et al. Real-world evaluation of effectiveness, persistence,
and usage patterns of tofacitinib in treatment of rheumatoid arthritis
in Australia. Clin Rheumatol. 2020;39(9):2545-2551.
15. Perrone V, Losi S, Rogai V, Antonelli S, Fakhouri W, Giovannitti M,
et al. Real-World Analysis of Therapeutic Patterns in Patients Affected
by Rheumatoid Arthritis in Italy: A Focus on Baricitinib. Rheumatol
Ther. 2020;7(3):657-665.
16. Bayat S, Tascilar K, Kaufmann V, Kleyer A, Simon D, Knitza J, et al.
AB0330 High remission rates in ra-real life data from bariticinib. Ann
Rheum Dis. 2020; 79: 1463-1464.
17. Prevoo ML, Van'T Hof M, Kuper HH, Van Leeuwen MA, Van De
Putte LB, Van Riel PL. Modified disease activity scores that include
twentyeightjoint counts development and validation in a prospective
longitudinal study of patients with rheumatoid arthritis. Arthritis
Rheum. 1995;38(1):44-48.
18. Aletaha D, Nell VP, Stamm T, Uffmann M, Pflugbeil S, Machold K,
Smolen JS. Acute phase reactants add little to composite disease
activity indices for rheumatoid arthritis: validation of a clinical activity
score. Arthritis Res Ther. 2005;7(4): R796-R806.
health assessment questionnaire format. Arthritis Rheum.
1999;42(10):2220-2230.
20. Leeb BF, Haindl PM, Maktari A, Nothnagl T, Rintelen B. Patient-
centered rheumatoid arthritis disease activity assessment by a modified
RADAI. J Rheumatol. 2008;35(7):1294-1299.
21. Leeb BF, Sautner J, Mai HT, Haindl PM, Deutsch C, Rintelen B. A
comparison of patient questionnaires and composite indexes in
routine care of rheumatoid arthritis patients. Joint Bone Spine.
2009;76(6):658-664.
22. Taylor PC, Emery P, Weinblatt ME, Mysler E, Rubbert-Roth A, Jia B,
et al. THU0102Patient disease trajectories in Baricitinib-treated
patients with rheumatoid arthritis and inadequate response to
methotrexate. Ann Rheum Dis. 2019;78(1):322-323.
23. Rosenbaum PR, Rubin DB. The central role of the propensity score in
observational studies for causal effects. Biometrika. 1983;70(1):41-55.
24. Taylor PC, Keystone EC, van der Heijde D, Weinblatt ME, del
Carmen Morales L, Reyes Gonzaga J, et al. Baricitinib vs. placebo or
adalimumab in rheumatoid arthritis. N Engl J Med. 2017;376(7):
652-662.
25. Van Vollenhoven RF, Fleischmann R, Cohen S, Lee EB, García
Meijide JA, Wagner S, et al. Tofacitinib or adalimumab vs. placebo in
rheumatoid arthritis. N Engl J Med. 2012;367(6):508-519.
26. Felson DT, Anderson JJ, Boers M, Bombardier C, Furst D, Goldsmith
C, et al. American College of Rheumatology preliminary definition of
improvement in rheumatoid arthritis. Arthritis Rheum. 1995;38(6):
727-735.
27. Genovese MC, Fleischmann R, Combe B, Hall S, Rubbert-Roth A,
Zhang Y, 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(10139):
2513-2524.
28. Leeb BF. Clinical studies-real world data. An area of tension?
Rheuma Plus. 2019;18:189-190.
29. Daien CI, Morel J. Predictive factors of response to biological disease
modifying antirheumatic drugs: Towards personalized medicine.
Mediators of inflammation. 2014;2014:386148.
30. Iwamoto N, Aramaki T, Ichinose K, Ueki Y, Kawakami A. AB0344
Factors which contribute persistency and optimal use of Tofacitinib;
Lessons from long term daily clinical use of Tofacitinib. Ann Rheum
Dis. 2020;79(1):1471-1472.
31. Lauper K, Mongin D, Iannone F, Kristianslund EK, Kvien TK,
Nordström DC, et al. Comparative effectiveness of TNF inhibitors
and tocilizumab with and without conventional synthetic disease-
modifying antirheumatic drugs in a pan-European observational
cohort of bio-naïve patients with rheumatoid arthritis. Semin Arthritis
Rheum. 2020;50(1):17-24.
32. Kamiya M. Efficacy of JAK Inhibitors in refractory Rheumatoid
Arthritis. Ann Rheum Dis. 2020;79(1):1472.
33. Masahiro H, Takahashi N, Kojima T. AB0348 the effectiveness
and safety of Baricitinib after insufficient response to bDMARDs
or tsDMARDs in patients with RA from Japanese multi-center
registry: 24 weeks outcomes. Ann Rheum Dis. 2020;79(1):
1473-1474.
34. Smolen JS, Landewé RBM, Bijlsma JWJ et al, EULAR
recommendations RA 2019 EULAR recommendations for the
management of rheumatoid arthritis with synthetic and biological
disease-modifying antirheumatic drugs: 2019 update. Ann Rheum Dis.
2020;79(6):685-699.
Leeb BF, et al.
Rheumatology (Sunnyvale), Vol.11 Iss.5 No:1000288 7
19. Pincus T, Swearingen C, Wolfe F. Toward a multidimensional Health
Assessment Questionnaire (MDHAQ): Assessment of advanced
activities of daily living and psychological status in the patientfriendly
35. Deutsches Netzwerk Versorgungsforschung e.V. Pressemitteilung Das
Deutsche Netzwerk Versorgungsforschung begrüßt
neue Möglichkeiten zur Einbindung qualitativ hochwertiger
Registerdaten in die Arzneimittel-Nutzenbewertung, press release.
2020.
... Additionally, we report greater improvement in CDAI disease activity, as well as pain and physical functioning. Furthermore, results presented here echo findings described in other real-world studies; patients treated with baricitinib are reported to have high remission and LDA, regardless of treatment history, from 6 to 12 months of treatment [27][28][29], while other reports have noted similar proportions of patients achieving these outcomes at 12 months, regardless of treatment with baricitinib, TNFi, or OMA [30]. ...
Article
Full-text available
RA-BE-REAL is a 3-year, multinational, prospective, observational study of adult patients with rheumatoid arthritis (RA) evaluating time to discontinuation of initial RA treatment along with patient baseline characteristics. This study’s primary objective was to assess the time to discontinuation of initial baricitinib, any other targeted synthetic disease-modifying anti-rheumatic drug (tsDMARD), or any biologic disease-modifying anti-rheumatic drug (bDMARD) treatment for all causes (excluding sustained clinical response) over 24 months in a European population. Patients initiated treatment with baricitinib (cohort A) or any bDMARD or tsDMARD (cohort B) for the first time. This study’s primary objective was to assess the time to discontinuation of initial baricitinib, any other targeted synthetic disease-modifying anti-rheumatic drug (tsDMARD), or any biologic disease-modifying anti-rheumatic drug (bDMARD) treatment for all causes (excluding sustained clinical response) over 24 months in a European population. Comparative effectiveness analyses, over 24 months, included time to treatment discontinuation for all causes (excluding sustained clinical response), percentage of patients achieving Clinical Disease Activity Index (CDAI) remission or low disease activity (LDA), as well as mean changes from baseline for CDAI, pain visual analogue scale, and the Health Assessment Questionnaire-Disability Index (HAQ-DI). For this European subpopulation, comparative analyses were performed using a frequentist model averaging (FMA) framework based on a data-driven machine learning causal inference approach to compare time to discontinuation, effectiveness, rates of remission or LDA, and patient-reported outcomes over 24 months comparing baricitinib with TNFi, as well as non-TNFi and tsDMARD grouped as other mechanism of action (OMA) drugs. In the European sample of RA-BE-REAL, patients with RA treated with baricitinib experienced fewer discontinuations in comparison to those treated with tumour necrosis factor inhibitors or OMA. Overall, patients naïve to b/tsDMARDs achieved a higher rate of LDA and remission compared with experienced patients. A significantly greater proportion of patients treated with baricitinib achieved LDA compared with b/tsDMARDs. This real-world data can better inform clinicians about baricitinib effectiveness and drug survival when prescribing treatment for patients with RA across different subpopulations.
Article
Full-text available
Baricitinib, an oral selective Janus kinase (JAK)1/JAK2 inhibitor, is approved as monotherapy or in combination with methotrexate for treating adults with moderate-to-severe active rheumatoid arthritis (RA) and provides improvements in clinical signs, symptoms and patient-reported outcomes. Currently, baricitinib is approved for treating RA in more than 75 countries. In several pivotal Phase II and III RA trials (RA-BALANCE, RA-BEGIN, RA-BEAM, RA-BUILD, RA-BEACON, RA-BEYOND), up to seven years of baricitinib treatment was well tolerated and provided rapid and sustained efficacy, which was confirmed in real-world settings. Safety signals for another JAK inhibitor, tofacitinib, have emerged, as observed in the post-marketing Phase IIIb/IV trial Oral Rheumatoid Arthritis Trial (ORAL) Surveillance; safety signals were subsequently highlighted in a retrospective study of baricitinib and consequently new recommendations and warnings and precautions for all JAK inhibitors have been issued. Ongoing studies to further characterise and clarify the benefit:risk of JAK inhibitors include registries and controlled trials. This capstone review summarises clinical and real-world data outlining the benefit:risk profile of baricitinib, confirming that the improved disease activity and physical function of patients with RA treated with this JAK inhibitor observed in clinical trials is translated into effectiveness in clinical practice, with a low rate of discontinuations.
Article
Full-text available
Background Recent developments of targeted treatments such as targeted synthetic DMARDs (tsDMARDs) increase the chances of a sustained low disease activity (LDA) or remission state for patients suffering rheumatoid arthritis (RA). tsDMARDs such as baricitinib, an oral inhibitor of the Janus Kinases (JAK1/JAK2) was recently approved for the treatment of RA with an inadequate response to conventional (cDMARD) and biological (bDMARD) therapy. (1, 2). Objectives Aim of this study is to analyze the effect of baricitinb on disease activity (DAS28, LDA) in patients with RA in real life, to analyze drug persistance and associate these effects with various baseline characteristics. Methods All RA patients were seen in our outpatient clinic. If a patient was switched to a baricitinib due to medical reasons, these patients were included in our prospective, observational study which started in April 2017. Clinical scores (SJC/TJC 76/78), composite scores (DAS28), PROs (HAQ-DI; RAID; FACIT), safety parameters (not reported in this abstract) as well as laboratory biomarkers were collected at each visit every three months. Linear mixed effects models for repeated measurements were used to analyze the time course of disease activity, patient reported outcomes and laboratory results. We estimated the probabilities of continued baricitinib treatment and the probabilities of LDA and remission by DAS-28 as well as Boolean remission up to one year using survival analysis and explored their association with disease characteristics using multivariable Cox regression. All patients gave informed consent. The study is approved by the local ethics. Results 95 patients were included and 85 analyzed with available follow-up data until November 2019. Demographics are shown in table 1. Mean follow-up duration after starting baricitinib was 49.3 (28.9) weeks. 51 patients (60%) were on monotherapy. Baricitinib survival (95%CI) was 82% (73% to 91%) at one year. Cumulative number (%probability, 95%CI) of patients that attained DAS-28 LDA at least once up to one year was 67 (92%, 80% to 97%) and the number of patients attaining DAS-28 and Boolean remission were 31 (50%, 34% to 61%) and 12(20%, 9% to 30%) respectively. Median time to DAS-28 LDA was 16 weeks (Figure 1). Cox regression analyses did not show any sufficiently precise association of remission or LDA with age, gender, seropositivity, disease duration, concomitant DMARD use and number of previous bDMARDs. Increasing number of previous bDMARDs was associated with poor baricitinib survival (HR=1.5, 95%CI 1.1 to 2.2) while this association was not robust to adjustment for baseline disease activity. Favorable changes were observed in tender and swollen joint counts, pain-VAS, patient and physician disease assessment scores, RAID, FACIT and the acute phase response. Conclusion In this prospective observational study, we observed high rates of LDA and DAS-28 remission and significant improvements in disease activity and patient reported outcome measurements over time. References [1]Keystone EC, Taylor PC, Drescher E, Schlichting DE, Beattie SD, Berclaz PY, et al. Safety and efficacy of baricitinib at 24 weeks in patients with rheumatoid arthritis who have had an inadequate response to methotrexate. Annals of the rheumatic diseases. 2015 Feb;74(2):333-40. [2]Genovese MC, Kremer J, Zamani O, Ludivico C, Krogulec M, Xie L, et al. Baricitinib in Patients with Refractory Rheumatoid Arthritis. The New England journal of medicine. 2016 Mar 31;374(13):1243-52. Figure 1. Cumulative probability of low disease activity or remission under treatment with baricitinib. Disclosure of Interests Sara Bayat Speakers bureau: Novartis, Koray Tascilar: None declared, Veronica Kaufmann: None declared, Arnd Kleyer Consultant of: Lilly, Gilead, Novartis,Abbvie, Speakers bureau: Novartis, Lilly, David Simon Grant/research support from: Else Kröner-Memorial Scholarship, Novartis, Consultant of: Novartis, Lilly, Johannes Knitza Grant/research support from: Research Grant: Novartis, Fabian Hartmann: None declared, Susanne Adam: None declared, Axel Hueber Grant/research support from: Novartis, Lilly, Pfizer, EIT Health, EU-IMI, DFG, Universität Erlangen (EFI), Consultant of: Abbvie, BMS, Celgene, Gilead, GSK, Lilly, Novartis, Speakers bureau: GSK, Lilly, Novartis, Georg Schett Speakers bureau: AbbVie, BMS, Celgene, Janssen, Eli Lilly, Novartis, Roche and UCB
Article
Full-text available
IntroductionThe objective of this study was to evaluate treatment patterns in patients with rheumatoid arthritis (RA), with a focus on the utilization of baricitinib, an oral highly selective Janus kinase 1 and 2 inhibitor, in an Italian real-world setting.Methods This observational retrospective analysis was based on data collected in selected Italian administrative databases. Patients aged ≥ 18 years with a diagnosis of RA defined by hospitalization discharge diagnosis (International Classification of Diseases, Ninth Revision, Clinical Modification code 714.0) or by disease exemption code 006 for RA in 2018 were included. The index date (ID) was defined as the date of first prescription for a drug indicated for RA during the inclusion period. Patients without a prescription for biologic/targeted synthetic disease-modifying antirheumatic drugs (b/tsDMARDs) before the ID were considered to be b/tsDMARD naïve. A further analysis was performed on patients only receiving baricitinib.ResultsA total of 41,290 RA patients were enrolled, of whom 55.6% were not treated with conventional synthetic DMARDs (csDMARDs) or b/tsDMARDs, 39.4% were receiving therapy with csDMARDs, and 5.0% were using b/tsDMARDs. In the latter group, 2.7% (n = 56) were receiving therapy with baricitinib. In 2018, 13.2% of csDMARD-treated patients switched to b/tsDMARDs, of whom 4.3% (n = 93) of these switched to baricitinib. In total, 149 patients (mean age ± standard deviation 57.6 ± 12.1; 12.8% male) had a baricitinib prescription, of whom 51% were b/tsDMARD naïve. At baseline, 61.7% of baricitinib users were receiving combination therapy with csDMARDs plus corticosteroids, 26.2% were receiving combination therapy with corticosteroids, and 8.1% were receiving combination therapy with csDMARDs; 4% were receiving baricitinib monotherapy. During follow-up, the proportion of patients receiving baricitinib monotherapy increased to 38.9%, while 26.9, 18.8, and 15.4% of baricitinib users received combination therapy with corticosteroids, csDMARDs plus corticosteroids, and csDMARDs, respectively.Conclusion This study provides a current view of the treatment patterns in Italian patients with RA in a real-world setting of daily clinical practice, with a focus on baricitinib utilization.
Article
Full-text available
Objectives To provide an update of the European League Against Rheumatism (EULAR) rheumatoid arthritis (RA) management recommendations to account for the most recent developments in the field. Methods An international task force considered new evidence supporting or contradicting previous recommendations and novel therapies and strategic insights based on two systematic literature searches on efficacy and safety of disease-modifying antirheumatic drugs (DMARDs) since the last update (2016) until 2019. A predefined voting process was applied, current levels of evidence and strengths of recommendation were assigned and participants ultimately voted independently on their level of agreement with each of the items. Results The task force agreed on 5 overarching principles and 12 recommendations concerning use of conventional synthetic (cs) DMARDs (methotrexate (MTX), leflunomide, sulfasalazine); glucocorticoids (GCs); biological (b) DMARDs (tumour necrosis factor inhibitors (adalimumab, certolizumab pegol, etanercept, golimumab, infliximab), abatacept, rituximab, tocilizumab, sarilumab and biosimilar (bs) DMARDs) and targeted synthetic (ts) DMARDs (the Janus kinase (JAK) inhibitors tofacitinib, baricitinib, filgotinib, upadacitinib). Guidance on monotherapy, combination therapy, treatment strategies (treat-to-target) and tapering on sustained clinical remission is provided. Cost and sequencing of b/tsDMARDs are addressed. Initially, MTX plus GCs and upon insufficient response to this therapy within 3 to 6 months, stratification according to risk factors is recommended. With poor prognostic factors (presence of autoantibodies, high disease activity, early erosions or failure of two csDMARDs), any bDMARD or JAK inhibitor should be added to the csDMARD. If this fails, any other bDMARD (from another or the same class) or tsDMARD is recommended. On sustained remission, DMARDs may be tapered, but not be stopped. Levels of evidence and levels of agreement were mostly high. Conclusions These updated EULAR recommendations provide consensus on the management of RA with respect to benefit, safety, preferences and cost.
Article
Full-text available
Introduction No published studies exist comparing the effectiveness of tofacitinib with other advanced therapies for the treatment of rheumatoid arthritis (RA) in real-world clinical practice. Here, we report differences in effectiveness of tofacitinib compared with standard of care, tumor necrosis factor inhibitors (TNFi), with or without concomitant methotrexate (MTX), using US Corrona registry data. Methods This observational cohort study included RA patients receiving tofacitinib (from 6 November 2012; N = 558) or TNFi (from 1 November 2001; N = 8014) with or without MTX until 31 July 2016. Efficacy outcomes at 6 months included modified American College of Rheumatology 20% responses, Clinical Disease Activity Index (CDAI) and Pain. Outcomes were compared between patients receiving TNFi and tofacitinib with or without MTX and by line of therapy. Outcomes within therapy lines were compared using propensity-score matching; between-group differences were estimated using mixed-effects regression models. Results Patients receiving tofacitinib had longer RA duration and a greater proportion had previously received biologics than those receiving TNFi; other baseline characteristics were comparable. In patients receiving second- and third-line TNFi therapy, CDAI low disease activity/remission response rates were significantly better with concomitant MTX. Too few patients received tofacitinib as second line for meaningful assessment. No significant differences were observed in outcomes between tofacitinib as monotherapy and tofacitinib with concomitant MTX. Conclusions In clinical practice, TNFi efficacy is improved with concomitant MTX in the second and third line. In the third/fourth line, patients are likely to achieve similar efficacy with tofacitinib monotherapy, or TNFi or tofacitinib in combination with MTX. Funding Pfizer Inc
Article
Full-text available
: Introduction: Tofacitinib is an oral JAK inhibitor indicated for the treatment of rheumatoid arthritis (RA). The efficacy and safety of tofacitinib have been shown in several randomized clinical trials. The study presented here aimed to assess the clinical tolerability and effectiveness of tofacitinib among RA patients in real life. Methods: Consecutive patients between January 2015 and April 2017 with RA who fulfilled the American College of Rheumatology (ACR)/European League Against Rheumatism (EULAR) 2010 criteria were included in a prospectively designed analysis of retrospective data. Patients were initiated on tofacitinib 5 mg bid. The primary objective was to analyze the safety of tofacitinib in a real-life cohort. Safety was assessed by the reasons to stop tofacitinib during follow up and changes of liver enzymes, hemoglobin, and creatinine. The secondary outcome was to analyze the frequency of and time to achieve low disease activity (LDA) and remission as defined by 28 joint count disease activity score (DAS28). Results: A total of 144 patients were treated with tofacitinib. A total of 84.9% of patients were pre-exposed to at least one biological agent. The average DAS28 at the initiation of tofacitinib was 4.43. A total of 50.0% of patients were positive for rheumatoid factor and 49.0% for ACPA. The mean follow up was 1.22 years (range 10d–3.7a) after initiation of tofacitinib treatment. A total of 94 (64.4%) patients remained on tofacitinib during follow-up. The average time to stop tofacitinib was 190.0 days. Reasons to stop tofacitinib were: insufficient response (n = 23), gastrointestinal symptoms (n = 18), infection (n = 5), myalgia (n = 2), remission (n = 2), headache (n = 2), cough, blue finger syndrome, intolerance, heartburn, psoriasis, and increased liver enzymes (all n = 1). Increased alanine amino transferase (ALAT) or aspartate amino transferase (ASAT) > 2× upper limit of normal (ULN) were detected in 3.3% and 4.4% of patients, respectively. Hemoglobin decrease of >10% was detected in 15.1% of the patients and decreased lymphocytes <500/μL in 3.4%. An increase of creatinine >20% was detected in 9.4% of patients. A total of 62.9% and 50.0% of the patients achieved low disease activity (LDA) or remission after a median of 319 and 645 days, respectively. These rates were significantly higher in patients naïve to biologic agents as compared to patients pre-exposed to biologics (LDA: naïve 100% 92 d, pre-exposed 57.0% 434 d, p ≤ 0.001; remission: naïve 86.7% 132 d, pre-exposed 44.1%, 692 d, p = 0.001). Conclusions: Tofacitinib is a safe and effective treatment option for patients with RA. Tofacitinib may induce high rates of LDA and remission in patients with active disease, even after the use of one or more biologics, though the rate appeared higher in patients naïve to biologics. Tofacitinib may be a valuable option in a treat-to-target approach. Our data demonstrate that Janus kinase (JAK) inhibitors are safe and efficacious in real life patients.
Article
Full-text available
Tofacitinib and baricitinib are the first orally available, small-molecule inhibitors of Janus kinase (JAK) enzymes to be approved for the treatment of RA. Tofacitinib is a selective JAK1, 3 inhibitor with less activity against JAK2 and TYK2 and baricitinib is a selective, oral JAK1, 2 inhibitor with moderate activity against TYK2 and significantly less activity against JAK3. Both drugs have undergone extensive phase III clinical trials in RA and demonstrated rapid improvements in disease activity, function and patient-reported outcomes as well as disease modification. Tofacitinib 5 mg bd, was approved by the Federal Drug Administration in 2012 for the treatment of RA in patients who are intolerant or unresponsive to MTX. An extended release formulation for the treatment of RA was approved by Federal Drug Administration in 2016. In 2017 the European Medicines Agency approved tofacitinib 5 mg bd in combination with MTX and baricitinib 4 mg and 2 mg once daily for the treatment of moderate to severe active RA in adult patients who are intolerant or unresponsive to one or more conventional synthetic DMARDs.
Article
Background Recently, tofacitinib treatment in rheumatoid arthritis (RA) is thought to be not inferior to other biologic disease-modifying antirheumatic drugs (bDMARDS) such as TNF-inhibitor. However, approval of tofacitinib for treatment with RA is relatively recent as compared to other bDMARDs, therefore until now, little knowledge about long term efficacy and safety of tofacitinib in real-world settings or about which patients should be initiated tofacitinib are available. Thus, we need evidence from real-world setting for optimal use of tofacitinib. Objectives To investigate the efficacy and safety of tofacitinib and to identify factors which contribute persistency and efficacy of tofacitinib treatment during 2 years. Methods 148 patients, for whom tofacitinib was initiated until January 2018 were enrolled. All patients received 5 mg of tofacitinib twice daily and were followed for 2 years. Clinical disease activity indicated by disease activity score (DAS)28-ESR as well as adverse events (AEs) were evaluated. Statistical analysis was performed to determine which baseline variables influenced the persistency and efficacy of tofacitinib. Results 92 patients (62.2 %) continued tofacitinib for 2 years. Clinical disease activity rapidly and significantly decreased, and this efficacy continued throughout the 2 years: i.e., DAS28-ESR decreased from 5.13 ± 1.42 at baseline to 4.02 ± 1.11 at 4 weeks and 3.91 ± 1.32 at 2 years (P<0.0001, vs. baseline). 55 AEs including 22 herpes zoster infection occurred during tofacitinib treatment. 27 patients discontinued tofacitinib due to lack of efficacy. Multivariable logistic analysis showed that the number of bDMARDs previously used and age were associated with discontinuation of tofacitinib treatment due to lack of efficacy(table 1). Another set of multivariable logistic analysis revealed that lower disease activity at baseline contributed the achievement of DAS-low disease activity(odds ratio= 1.56, 95% confidence interval: 0.48-0.85). In the concomitant use of MTX or without MTX, the Δ values of DAS28-ESR from baseline to 24 months were -1.62, -1.13, respectively(P=0.12). And, the efficacy of patients after switching from tocilizumab(TCZ), which also inhibit IL-6 as same as JAK inhibitor, were not inferior as compared to non-switching from TCZ (Δ DAS28-ESR;-1.07, -1.61, respectively P=0.10). Table 2. Univariate Model Multivariate Model OR(95% CI) P-value OR(95% CI) P-value Age (per 1-year increase) 0.92(0.88-0.95) <0.001* 0.91(0.87-0.95) <0.001* Disease duration (per 1-year increase) 0.97(0.97-1.02) 0.21 Concomitant MTX use (yes/no) 0.95(0.39-2.31) 0.91 Concomitant oral steroid use (yes/no) 1.84(0.76-4.42) 0.16 1.61(0.60-4.34) 0.349 Number of previous use of bDMARDs (per drug) 1.26(0.99-1.62) 0.05 1.39(1.05-1.84) 0.02* DAS-ESR at baseline (per 1 increase) 1.02(0.76-1.36) 0.92 LDA achievement at 1 year (yes/no) 0.56(0.76-1.49) 0.23 RF positive (yes/no) 0.91(0.32-2.56) 0.85 ACPA positive (yes/no) 0.76(0.27-2.18) 0.62 OR odds raio, 95% CI 95% confidence interval, MTX methotrexate, bDMARDs biologic disease-modifying antirheumatic drugs, ACPA anti-citrullinated protein antibodies, RF rheumatoid factor *P<0.05 Conclusion Our present study suggests that tofacitinib is effective in real-world settings even without concomitant MTX. Our results also suggest that for continuous use of tofacitinib without lack of efficacy, use tofacitinib earlier during switching strategy for RA patients who have failed to be treated with bDMARDs is better. Disclosure of Interests None declared
Article
IntroductionThe aim of this study was to describe the real-world evidence for effectiveness, treatment persistence, and treatment patterns among patients in the community with rheumatoid arthritis treated with the JAK inhibitor tofacitinib.Methods This was a retrospective, non-interventional cohort study that extracted data for new users of tofacitinib or biologic disease-modifying antirheumatic drugs (bDMARDs) from the Australian Optimizing Patient outcomes in Australian RheumatoLogy (OPAL) dataset between March 2015 and September 2018. Patients were propensity score matched at a 1:2 tofacitinib to bDMARD ratio based on age, sex, and selected baseline treatment combinations. Treatment effectiveness was evaluated using disease status measures. Treatment persistence was calculated and the percentage of patients receiving monotherapy or combination therapy at treatment initiation was evaluated.ResultsData from 2810 patients were extracted and 1950 patients were included in the matched population (1300 bDMARD initiators and 650 tofacitinib initiators). Patients were predominantly aged 55 to 74 years (57.8%) and female (81.2%). After 18 months of treatment, 52.4% and 57.8% of patients had achieved disease activity score (DAS) remission in the bDMARD and tofacitinib groups, respectively. The median treatment persistence for tofacitinib was similar to that for bDMARDs: 34.2 months (95% CI 32.2 to not reached) and 33.8 months (95% CI 28.8 to 40.4), respectively. In the overall population, more patients were prescribed tofacitinib as monotherapy (43.4%) compared with bDMARD monotherapy (33.4%).Conclusions Tofacitinib demonstrated treatment effectiveness and persistence similar to bDMARDs. Overall, there was a trend for more use of tofacitinib as monotherapy than bDMARDs.Key Points• This study provides real-world evidence regarding effectiveness, treatment persistence, and treatment patterns, among patients with rheumatoid arthritis (RA) in the community being treated with tofacitinib.• The study suggests that tofacitinib is an effective and enduring intervention in RA with tofacitinib persistence and effectiveness comparable to bDMARDs.
Article
Objectives: To compare treatment effectiveness in rheumatoid arthritis (RA) patients naïve to biological disease-modifying antirheumatic drugs (bDMARDs) treated with tocilizumab (TCZ) or TNF-inhibitor (TNFi) with (-combo) or without (-mono) conventional synthetic DMARDs (csDMARDs). Methods: Patients with RA across 7 European registries, naïve to bDMARDs who initiated treatment with TCZ or TNFi from 2009 to 2016 were included. Drug retention rate was analyzed using Kaplan�Meier and Cox models, and CDAI over time by mixed models. The proportions of patients reaching CDAI low disease activity (LDA) and remission after one year were corrected for attrition. Results: 6713 TNFi-combo, 3762 TNFi-mono, 646 TCZ-combo and 384 TCZ-mono were eligible. Crude median retention was 3.67 years (95%CI 3.41�3.83) for TNFi-combo, 4.14 (3.77�4.62) for TNFi-mono, 2.98 (2.76�3.34) for TCZ-combo and 3.63 years (3.34�5.03) for TCZ-mono. After adjustment for covariates, country and year of treatment initiation stratification, hazards of discontinuation were lower for TCZ-mono (0.60, 95% CI 0.52�0.69) and TCZ-combo (0.66, 95% CI 0.54�0.81) compared to TNFi-combo. Adjusted CDAI evolution was not significantly different between groups. CDAI LDA and remission corrected for attrition were similar between TCZ with or without csDMARDs and TNFi-combo. Conclusion: In routine care across 7 European countries, the adjusted drug retention, adjusted CDAI over time and attrition-corrected response proportion for RA patients were similar for bio-naïve patients if treated with TNFi-combo, TCZ-combo or TCZ-mono.
Conference Paper
Background In RA-BEAM phase 3 study, baricitinib (bari) 4mg demonstrated clinical efficacy in patients (pts) with rheumatoid arthritis (RA) and an inadequate response to methotrexate. Bari, a selective Janus kinase 1/2 inhibitor, is approved for the treatment of moderate to severe active RA in adults in >50 countries. For improved treatment strategy, it is important to understand whether the pt population is composed of distinct pt groups with differing treatment responses and associated baseline characteristics. Objectives To identify different treatment trajectories, based on CDAI improvement, in bari 4mg treated pts over 52 weeks; and examine the associated clinical disease measures, structural damage score and baseline characteristics. Methods Growth Mixed Model (GMM), a novel latent class mixed model, was used to classify the longitudinal disease patterns instead of predefining a clinical responder at a specific time point. GMM was applied to observed CDAI values for bari 4mg pts, continuously treated from 0-52 weeks or up to rescue, to cluster pts into subgroups based on their trajectory patterns. Following identification of the groups, baseline pt characteristics and disease measures were compared between groups. Results Bari 4mg treated pts (N=487) were classified into 3 groups based on their CDAI trajectory patterns: Group 1 (n=344, 71%), Group 2 (n=56, 11%), and Group 3 (n=87, 18%) (Figure). Group 1 had lower baseline CDAI (34, Table 1), achieved CDAI≤10 (low disease activity, LDA) rapidly and maintained LDA up to 52 weeks. Group 2 had higher CDAI at baseline (48, Table 1), responded quickly, and although pts took longer to attain LDA, they continued to show CDAI improvement. Group 3 had similar baseline CDAI values (48, Table 1) to Group 2 but higher baseline damage (mean total Sharp score [mTSS] of 50, versus 41 for Groups 1 and 2, Table 1). Most Group 3 pts did not achieve LDA but continued to show improvement over time. The majority of pts had no radiographic progression with the highest proportion in Group 1 (Table 2). The trajectories of average pain VAS, Health Assessment Questionnaire–Disability Index (HAQ-DI), tender joint count 28, and swollen joint count 28 were consistent in the 3 groups. View this table: • View inline • View popup Abstract THU0102 –Table 1 Baseline characteristics: View this table: • View inline • View popup Abstract THU0102 – Table 2 Response rates in patient groups, n (%) W=week Conclusion Baseline severity is associated with different treatment trajectories. With bari treatment, majority of pts achieved LDA and had no structural progression. Pts with high baseline disease activity were associated with longer time to achieve LDA. Pts with higher baseline structural damage in addition to high disease activity were less likely to achieve LDA, but consistent with the other groups, had similar low rate of joint damage progression. Long-term maintenance and continued improvement in CDAI were observed with bari treatment. • Download figure • Open in new tab • Download powerpoint Abstract THU0102 – Figure 1 Disclosure of Interests Peter C. Taylor Grant/research support from: Celgene, Galapagos, Eli Lilly, UCB, Consultant for: AbbVie, Galapagos, Gilead, Eli Lilly, Pfizer Inc, Paul Emery Grant/research support from: Pfizer, MSD, AbbVie, Bristol-Myers Squibb, Roche, Consultant for: Pfizer, MSD, AbbVie, Bristol-Myers Squibb, UCB, Roche, Novartis, Gilead,Samsung, Sandoz and Lilly, Michael E. Weinblatt Shareholder of: Stock option: CanFite, Lycera, Scipher, Inmedix, Grant/research support from: Crescendo Bioscience, Bristol Myers Squibb, Sanofi, Consultant for: AbbVie, Amgen, Bristol-Myers Squibb, CanFite, Corrona, Crescendo, GlaxoSmithKline, Gilead, Horizon, Lilly, Lycera, Merck, Novartis, Pfizer, Roche, Samsung, Scipher, Set Point, Eduardo Mysler Grant/research support from: AbbVie, Bristol-Myers Squibb, Eli Lilly, Pfizer, Novartis, Janssen, Grant/research support from: AbbVie, Bristol-Myers Squibb, Eli Lily, Janssen, Medimmune, Pfizer Inc, and Roche, Consultant for: AbbVie, Bristol-Myers Squibb, Eli Lily, Janssen, Medimmune, Pfizer Inc, and Roche, Speakers bureau: AbbVie, Bristol-Myers Squibb, Eli Lilly, Pfizer, Novartis, Janssen, Speakers bureau: AbbVie, Bristol-Myers Squibb, Eli Lily, Janssen, Medimmune, Pfizer Inc, and Roche, Andrea Rubbert-Roth Consultant for: Chugai, Eli Lilly, Roche, and Sanofi, Speakers bureau: AbbVie, Bristol-Myers Squibb, Chugai, Hexal/Novartis, Janssen, Eli Lilly, Merck Sharp & Dohme, Pfizer, Roche, and Sanofi, Bochao Jia Shareholder of: Eli Lilly and Company, Employee of: Eli Lilly and Company, Luna Sun Shareholder of: Eli Lilly and Company, Employee of: Eli Lilly and Company, Yushi Liu Shareholder of: Eli Lilly and Company, Employee of: Eli Lilly and Company, Yun-Fei Chen Shareholder of: Eli Lilly and Company, Employee of: Eli Lilly and Company, Anabela Cardoso Shareholder of: Eli Lilly and Company, Employee of: Eli Lilly and Company, Yoshiya Tanaka Grant/research support from: Abbvie, Astellas, Bristol-Myers Squibb, Chugai, Daiichi-Sankyo, Eisai, Mitsubishi-Tanabe, MSD, Ono, Taisho-Toyama, Takeda, Speakers bureau: Abbvie, Asahi-kasei, Astellas, Bristol-Myers Squibb, Chugai, Daiichi-Sankyo, Eli Lilly, Eisai, Glaxo-Smithkline, Janssen, Mitsubishi-Tanabe, Novartis, Pfizer Japan Inc, Sanofi, Takeda, UCB, YL Biologics