You are on page 1of 7

Clinical Rheumatology

https://doi.org/10.1007/s10067-020-05021-7

ORIGINAL ARTICLE

Real-world evaluation of effectiveness, persistence, and usage


patterns of tofacitinib in treatment of rheumatoid arthritis
in Australia
Paul Bird 1,2 & Geoffrey Littlejohn 1,3 & Belinda Butcher 2,4 & Tegan Smith 1 & Candida da Fonseca Pereira 5 &
David Witcombe 5 & Hedley Griffiths 1,6

Received: 15 December 2019 / Revised: 20 February 2020 / Accepted: 28 February 2020


# International League of Associations for Rheumatology (ILAR) 2020

Abstract
Introduction The 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.
Results Data 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.

Keywords Medication persistence . Real-world . Rheumatoid arthritis . Targeted synthetic disease-modifying antirheumatic
drug . TNF-inhibitors . Tofacitinib

* Belinda Butcher 3
Monash University, Clayton, Victoria, Australia
bbutcher@writesourcemedical.com.au 4
WriteSource Medical Pty Ltd, Lane Cove, New South Wales,
Australia
1 5
OPAL Rheumatology Ltd, Sydney, New South Wales, Australia Pfizer Australia, Sydney, New South Wales, Australia
2 6
University of New South Wales, Kensington, New South Wales, Barwon Rheumatology Service, Geelong, Victoria, Australia
Australia
Clin Rheumatol

Introduction There is limited data from large real-world populations to


describe the characteristics and outcomes of patients who re-
Rheumatoid diseases such as rheumatoid arthritis (RA) ceive tofacitinib for the management of RA in Australia. This
have a significant negative impact on patients’ health- study aims to use the Optimizing Patient outcome in
related quality of life (HQoL) and impose a significant Australian rheumatoLogy (OPAL) dataset to provide real-
economic burden on society. Maximization of HQoL is world evidence regarding clinical effectiveness, treatment per-
the primary goal of treatment. This is achieved through sistence, and treatment patterns, among patients with RA be-
symptom and inflammation control, prevention of pro- ing treated with tofacitinib in the post-approval setting.
gressive structural damage, preservation or normalization Similar data was collected for patients treated with
of function and social participation, and targeting remis- bDMARDs to provide context in a large real-world clinical
sion [1, 2]. Treatment of RA usually involves a multi- practice setting.
faceted approach that includes pharmacologic and non-
pharmacologic strategies. Non-pharmacologic therapy
may include physical, occupational, and psychological Materials and methods
therapy, and surgery, while pharmacological therapy usu-
ally consists of various combinations of non-steroidal Study design and setting
antiinflammatory drugs (NSAIDs), analgesics, corticoste-
roids, and synthetic or biologic disease-modifying anti- This was a retrospective, non-interventional cohort study of
rheumatic drugs (DMARDs). clinical effectiveness, treatment persistence, and treatment pat-
Recently, oral targeted synthetic DMARDs (tsDMARD), terns in patients prescribed tofacitinib or bDMARDs. Data was
including tofacitinib, have become available. Tofacitinib is a extracted from real-world patient data from the Australian
potent, selective inhibitor of the Janus kinase (JAK) family of OPAL dataset derived from 42 rheumatology clinics around
kinases [3]. Tofacitinib was approved for use in Australia in Australia. The OPAL dataset collects information from individ-
February 2015 and included in the Pharmaceutical Benefits ual clinicians’ servers during routine clinical consultations.
Scheme (PBS) for reimbursement in October 2015 for the Clinicians contributing data to OPAL use purpose-built
treatment of patients with RA. worksheets in Audit4 software (Software4Specialists,
The efficacy, safety, and impacts on HQoL of tofacitinib Australia), and this software serves as the patient’s medical
have been assessed in randomized controlled trials (RCTs) record [15]. Diagnoses were made by individual rheumatolo-
[4–11]. A review of patient reported outcomes from clinical gists rather than being criteria based. Data de-identified for
studies found that overall tofacitinib was associated with an patient, clinic, and clinician were exported from each of the
increase in quality of life and functionality and with improve- OPAL member’s local server, aggregated across all sites, and
ments in patient reported assessment of pain and global as- analyzed based on a predefined protocol.
sessment of disease [12]. However, real-world evidence com- The activities of OPAL Rheumatology Ltd. have received
pliments the knowledge gained from clinical trials and pro- overarching ethics approval from the University of New South
vides information as to how factors such as the clinical setting Wales (UNSW) Human Research Ethics Committee (HREC),
and health system influence treatment effects and outcomes. based on a patient opt-out arrangement (HC17799). This re-
Observational studies evaluating the effects of treatment in a search protocol was approved by the UNSW HREC
routine clinical environment are a key source of real-world (HC17221).
data [13].
Real-world treatment of RA with tofacitinib has only just Patient population and eligibility criteria
begun to emerge as tofacitinib was only approved in the USA
in 2012 and in Europe in 2017. A review of the real-world Patients were included if they were registered in the OPAL
evidence from countries including the USA, Japan, Taiwan, dataset with a clinical diagnosis of RA and were aged between
Switzerland, Latin America, and Russia concerning the use of 18 and 94 years of age. Patients had initiated treatment with
tofacitinib in the management of patients with RA has been tofacitinib or a bDMARD and had at least 1 year of follow-up
reported [14]. The review found that patients initiating within the sample selection window between March 2015 and
tofacitinib usually had longer disease duration; tofacitinib ap- September 2018. The bDMARDs that were approved for use
peared to be more frequently used as a monotherapy than in Australia and included in this study were abatacept,
biological DMARDS (bDMARDs) and was effective without adalimumab, anakinra, certolizumab pegol, etanercept,
background use of methotrexate. The real-world data also golimumab, infliximab, rituximab, and tocilizumab.
showed that persistence of treatment with tofacitinib and ad- Tofacitinib was the only tsDMARD included in the study as
herence to treatment were good overall and similar to that seen no other tsDMARDs were approved in Australia at the com-
for bDMARDs [14]. mencement of the study. Patients who had no visit data
Clin Rheumatol

recorded and patients who had missing start dates for covariates were methotrexate monotherapy, methotrexate
tofacitinib/bDMARD treatment were not included. Patients in combination with other csDMARD(s), csDMARD(s)
with a diagnosis of any autoimmune rheumatic disease or other than methotrexate and neither methotrexate nor
inflammatory bowel disease except for RA were excluded. other csDMARD(s) (bDMARD monotherapy). Disease
severity based on DAS28-ESR was not included as a
Statistical and analytical assessment covariate as it led to the exclusion of too many patients.
Propensity score matching was determined on a ratio of
The primary exposure of interest was an initial prescription for one tofacitinib user to two bDMARD users (1:2) using a cal-
tofacitinib or a bDMARD identified during the sample selec- iper width of 0.20. The success of the matching was deter-
tion window. The date of the first prescription after the start of mined by examining the propensity score distribution (density
the sample selection window served as the study index date plot) in both the original sample and the matched sample and
and the beginning of the post-index period. Patients that had comparing the standardized differences (in means and propor-
received a prescription of tofacitinib during the sample selec- tions) between the groups in the matched cohort. A difference
tion window were considered part of the “tofacitinib group” above 0.1 was considered indicative of a substantial difference
even if they had also been prescribed a DMARD. All other in that covariate [16].
patients were considered to be in the bDMARD group.
Patients who discontinued treatment were followed up for a
minimum period of 1 year from their date on index. Data was Study size
evaluated for missing data and to identify implausible values.
Implausible values were adjudicated by two rheumatologists, A planned sample size of 500 and 2500 patients, in the
who were members of OPAL but independent of the sponsor. tofacitinib and bDMARD groups respectively, would ensure
Treatment effectiveness was evaluated from baseline (in- acceptably precise estimates of treatment patterns and clinical
dex date) to 18 months using Disease Activity Score-28 with effectiveness. Data was extracted for 652 patients adminis-
erythrocyte sedimentation rate (DAS28-ESR) and Clinical tered tofacitinib and 2158 patients administered bDMARDs.
Disease Activity Index (CDAI) and Simplified Disease Though the bDMARD sample size was smaller than planned,
Activity Index (SDAI) measures. An exploratory and descrip- this sample size still ensured clinically acceptable precision in
tive analysis of the most common adverse events was per- the estimates of treatment patterns and clinical effectiveness;
formed. Treatment persistence with the index DMARD, in for example, for a proportion of 30%, the confidence interval
part a surrogate for efficacy, was defined as the time (in con- is 28 to 32% for samples of 2500 or 2158.
secutive days) from treatment initiation until treatment discon-
tinuation. Duration of treatment of the index DMARD was
summarized using Kaplan-Meier estimates of treatment per-
sistence. Treatment pattern evaluation included percentage of Results
patients receiving monotherapy or conventional synthetic
DMARD (csDMARD) combination therapy at treatment ini- From March 2015 to September 2018, 2810 patients initiated
tiation and at 19- to 26-month follow-up. No formal compar- tofacitinib or bDMARDs and met the initial study selection
isons were made between patients treated with tofacitinib and criteria. Overall, 2158 (77%) patients received index treatment
bDMARDs. Any numerical differences or trends were de- with bDMARDs and 652 (23%) patients received index treat-
scriptive only. ment with tofacitinib. The matched population included 1950
patients (1300 bDMARD initiators and 650 tofacitinib initia-
Propensity score matching tors). Participant demographics for the matched population are
reported in Table 1. Patients were predominantly aged 55 to
In order to address the observational nature of the data, 74 years (57.8%), and female (81.2%). Baseline comorbidities
propensity score matching was planned between the (not shown) were well matched across bDMARD and
tofacitinib and bDMARD groups. Propensity score tofacitinib groups. The disease status was similar between
matching increases the comparability of the observed the two groups and the treatment combinations were also well
baseline characteristics in patients treated with tofacitinib matched at index between the two groups. At index, in the
and bDMARDs. The propensity score was the condition- matched population, median disease duration was 107 months
al probability of receiving treatment (e.g., tofacitinib ver- and 120 months (p = 0.037), for the bDMARD and tofacitinib
sus other biologic agent), which was estimated using groups, respectively. In the overall population, median disease
logistic regression. Covariates included age, sex, and se- duration was 100 months and 120 months (p = 0.002) for the
lected baseline (where baseline is the index date) treat- bDMARD and tofacitinib groups, respectively (data not
ment combinations. The baseline treatment combination shown).
Clin Rheumatol

Table 1 Patient demographics and disease characteristics at baseline for the propensity score matched population

bDMARD Tofacitinib

N (%) 1300 (66.7) 650 (33.3)


Age at index (years)
Mean (SD) 60.8 (13.1) 61.0 (12.7)
Median (min, max) 62 (21, 92) 63 (23, 89)
Gender, n (% of column)
Female 1056 (81.2%) 528 (81.2%)
Disease duration in months, median 107 [N = 818] 120 [N = 411]
Disease status
DAS28-CRP, n (% of column) N = 542 N = 304
Rem Low Mod High Rem Low Mod High
88 (16.2) 48 (8.9) 185 (34.1) 221 (40.8) 54 (17.8) 26 (8.6) 109 (35.9) 115 (37.8)
CDAI, n (% of column) N = 533 N = 308
Rem Low Mod High Rem Low Mod High
37 (6.9) 82 (15.4) 131 (24.6) 283 (53.1) 20 (6.5) 53 (17.2) 78 (25.3) 157 (51.0)
TJC28, mean (SD) 9.17 (8.65) [N = 783] 8.42 (8.24) [N = 402]
SJC28, mean (SD) 9.03 (8.47) [N = 783] 8.18 (8.09) [N = 402]
RAPID3, mean (SD) 3.58 (2.58) [N = 126] 4.39 (2.25) [N = 54]
Baseline treatment combinationsa
Methotrexate + cDMARD 294 (22.6%) 146 (22.5%)
Methotrexate alone 300 (23.1%) 150 (23.1%)
cDMARD (excluding methotrexate) 142 (10.9%) 72 (11.1%)
bDMARD monotherapy 564 (43.4%) 282 (43.4%)
Missing – –
a
Monotherapy was assigned for patients who were treated with a bDMARD or tsDMARD but had no record of methotrexate or alternative cDMARD at
the time of index. It is not clear, therefore, whether this is the true proportion of patients with monotherapy, or whether it represents missing information.
Missing represents patients with no information in the Medication dataset. These patients received bDMARD or tsDMARD but have no concomitant
medication reported

Treatment effectiveness treatment, 49.1% (n = 157/320) and 49.7% (n = 73/147)


had achieved DAS28-ESR remission and after 18 months
In the matched population, the percentage of patients of treatment, 52.4% (n = 89/170) and 57.8% (n = 48/83)
achieving DAS28-ESR disease remission and low disease of patients had achieved DAS28-ESR remission in the
activity (LDA) are shown in Fig. 1. Only patients with bDMARD and tofacitinib groups, respectively. There
baseline DAS28-ESR data are represented. At index, were no significant differences in remission rates be-
16.1% (n = 87/539) and 17.3% (n = 52/300) of patients tween the bDMARD and tofacitinib groups at any of
were in DAS28-ESR remission for the bDMARD and these time points (p = 0.66, p = 0.90, and p = 0.41 at in-
tofacitinib groups, respectively. After 3 months of dex, 3 months, and 18 months respectively).

Fig. 1 Percentage of patients in the propensity score matched population achieving DAS28-ESR a disease remission and b low disease activity (LDA)
Clin Rheumatol

Fig. 2 Percentage of patients in the propensity score matched population achieving a CDAI and b SDAI disease remission

At index, 7.4% (n = 40/539) and 8.0% (n = 24/300) of pa- Treatment persistence


tients were in DAS28-ESR LDA for the bDMARD and
tofacitinib groups, respectively. After 18 months of treatment The median persistence of treatment for the matched popula-
14.1% (n = 24/170) and 10.8% (n = 9/83) of patients were in tion was 33.8 months (95% CI 28.8 to 40.4) for patients pre-
DAS28-ESR LDA in the bDMARD and tofacitinib groups, scribed bDMARDs and was 34.2 months (95% CI 32.2 to
respectively. There were no significant differences in LDA “not reached”) for patients prescribed tofacitinib (Fig. 3).
rate between the bDMARD and tofacitinib groups at any of The difference in median persistence between groups was
these time points (p = 0.76 and p = 0.47, at index and not significant (p = 0.19, log rank test). In the propensity score
18 months respectively). matched population, the most common reasons for discontin-
The percentage of patients in the matched population uation were completion of treatment (429/1300 (33%), 135/
achieving CDAI and SDAI disease remission activity is 650 (25%)), lack of efficacy (281/1300 (22%), 113/650
shown in Fig. 2. At 18 months, the percentage of pa- (17%)), lack of efficacy, secondary failure (213/1300 (16%),
tients achieving CDAI and SDAI remission was similar 63/650 (10%)), and adverse reaction (211/1300 (16%), 81/650
with 29.0% (n = 51/176) and 29.2% (n = 50/171) (12%)), in the bDMARD and tofacitinib arms, respectively.
bDMARD patients and 30.5% (n = 25/82) and 30.9% Note patients could have more than one reason for discontin-
(n = 25/81) tofacitinib patients reporting CDAI or SDAI uation recorded.
remission, respectively (p = 0.80 and p = 0.79,
respectively). Treatment patterns
No adverse events were reported in more than 5% of the
population for bDMARD or tofacitinib treatment for the over- Monotherapy and combination cDMARD therapy treatment
all or the matched population. patterns are shown in Table 2 for the overall population. At
index, more patients were prescribed tofacitinib as monother-
apy (43.4%, n = 282/650) compared with bDMARD mono-
therapy (33.4%, n = 719/2154; p < 0.001). For patients re-
maining on index tofacitinib or index bDMARD at 19 to
26 months of follow-up, the percentage of patients on mono-
therapy increased for both groups (tofacitinib 49.1% (n = 208/
424); bDMARD 39.0% (n = 518/1329) (p < 0.001)).
Corticosteroid use was similar at index for both groups
(tofacitinib 35.6% (n = 232/652); bDMARD 34.2% (n = 738/
2158)) and similar at 18 months of treatment (tofacitinib
34.3% (n = 185/540); bDMARD 32.1% (n = 567/1764); p =
0.51 and 0.36). (Data not shown).

Discussion

This study of real-world treatment in Australian patients


Fig. 3 Treatment persistence in the propensity score matched population with RA found that tofacitinib demonstrated treatment
for bDMARD and tofacitinib effectiveness and persistence that was similar to
Clin Rheumatol

Table 2 Monotherapy and


combination therapy treatment Treatment combinations At index 19 to 26 months
patterns at treatment initiation and
at 19- to 26-month follow-up bDMARD Tofacitinib bDMARD Tofacitinib
(overall population) (N = 2154) (N = 650) (N = 1329) (N = 424)
n (%) n (%) n (%) n (%)

Methotrexate + cDMARD 589 (27.3) 146 (22.5) 270 (20.3) 78 (18.4)


Methotrexate alone 556 (25.8) 150 (23.1) 386 (29.0) 86 (20.3)
cDMARD (excluding 290 (13.5) 72 (11.1) 155 (11.7) 52 (12.3)
methotrexate)
bDMARD monotherapy 719 (33.4) 282 (43.4) 518 (39.0) 208 (49.1)

This is limited to patients who remained on their index bDMARD or tofacitinib at the time of follow-up

bDMARDs. Similar persistence between tofacitinib and “missing not at random” data occur. We assume, given the
bDMARDs has also been observed in other real-world source of this data however, that data are likely to be missing
studies [17, 18]. The criteria for reimbursement in at random, as important clinical information is likely to have
Australia require that patients demonstrate a response to been recorded within the patient’s notes. In addition, the
treatment within 3 months. The small reduction in per- Pharmaceutical Benefits Scheme only requires a total joint
sistence observed at 3 months in both treatment arms count/swollen joint count along with ESR or CRP, and as such
may be due to those patients who had not responded the patient global VAS is often not clinically recorded, so a
adequately to treatment by 3 months and were no longer DAS-28 score cannot be calculated. Propensity score
eligible for receiving government-subsided treatment. matching was not possible using the DAS28-ESR score due
Patients that initiated tofacitinib at index had longer disease to insufficient numbers.
duration than patients that initiated bDMARD treatment. This
has also been observed for patients in the US Corrona dataset Acknowledgments The authors acknowledge the members of OPAL
Rheumatology Ltd. and their patients for providing clinical data for this
where the mean disease duration was significantly longer for
study, and Software4Specialists Pty Ltd. for providing the Audit4 plat-
initiators of tofacitinib (13.9 years) compared with initiators of form. Medical writing services provided by Theresa Wade from
bDMARDs (9.9 years, p < 0.001) [19]. The longer duration of WriteSource Medical were funded by OPAL in accordance with Good
disease for patients that initiated tofacitinib may reflect that Publication Practice (GPP3) guidelines (http://ismpp.org/gpp3).
tofacitinib has only recently become available compared with
Funding information This study was funded by Pfizer.
other bDMARD therapy. Patients may have been waiting for a
therapy with a new mode of action because they were not
responsive to other bDMARDs. It may also reflect that some Compliance with ethical standards
patients may have been waiting to use orally administered
Conflict of interest PB has attended advisory board meetings for Pfizer,
tofacitinib rather than use an injectable bDMARD. Celgene, Novartis, Janssen, Gilead, and AbbVie; been a consultant for
There was a trend for a greater use of tofacitinib as mono- Roche; and received speaker financial support from Janssen. BB is an
therapy compared with bDMARDs. Observational studies in independent statistical consultant, funded by OPAL. HG has attended
the USA have also observed that tofacitinib was used more advisory board meetings for Pfizer, Novartis, Janssen, and AbbVie; been
a consultant for Roche; and received speaker financial support from
commonly as monotherapy than bDMARDs [17, 19, 20]. Janssen and conference financial support from Bristol-Myers Squibb.
The results from this study suggest that tofacitinib is an GL has attended advisory board meetings for Janssen and AbbVie and
effective and enduring intervention in RA with tofacitinib per- has received speaker financial support from AbbVie and Bristol-Myers
sistence and effectiveness comparable to bDMARDs. Future Squibb. TS has no conflicts to report. DW and CdFP are employees of
Pfizer.
research could focus on trying to identify factors associated
with lack of persistence.

References
Limitations
1. Tymms K, Littlejohn G (2014) OPAL: a clinician driven point of
A number of limitations of this study have been identified. care observational data management consortium. Clin Exp
This was a retrospective study of observational data and as Rheumatol 32(5 Suppl 85):S-150–S-152
would be expected, there was a large proportion of missing 2. Smolen JS, Breedveld FC, Burmester GR, Bykerk V, Dougados M,
Emery P, Kvien TK, Navarro-Compan MV, Oliver S, Schoels M,
data. The sample size, variables, and study duration were se- Scholte-Voshaar M, Stamm T, Stoffer M, Takeuchi T, Aletaha D,
lected to minimize the impact of this. Missing data does im- Andreu JL, Aringer M, Bergman M, Betteridge N, Bijlsma H,
pair statistical analysis and interpretation, especially if Burkhardt H, Cardiel M, Combe B, Durez P, Fonseca JE,
Clin Rheumatol

Gibofsky A, Gomez-Reino JJ, Graninger W, Hannonen P, Haraoui 12. Boyce EG, Vyas D, Rogan EL, Valle-Oseguera CS, O'Dell KM
B, Kouloumas M, Landewe R, Martin-Mola E, Nash P, Ostergaard (2016) Impact of tofacitinib on patient outcomes in rheumatoid
M, Ostor A, Richards P, Sokka-Isler T, Thorne C, Tzioufas AG, van arthritis - review of clinical studies. Patient Relat Outcome Meas
Vollenhoven R, de Wit M, van der Heijde D (2016) Treating rheu- 7:1–12. https://doi.org/10.2147/PROM.S62879
matoid arthritis to target: 2014 update of the recommendations of an 13. Sherman RE, Anderson SA, Dal Pan GJ, Gray GW, Gross T, Hunter
international task force. Ann Rheum Dis 75(1):3–15. https://doi. NL, LaVange L, Marinac-Dabic D, Marks PW, Robb MA, Shuren
org/10.1136/annrheumdis-2015-207524 J, Temple R, Woodcock J, Yue LQ, Califf RM (2016) Real-world
3. Dhillon S (2017) Tofacitinib: a review in rheumatoid arthritis. evidence - what is it and what can it tell us? N Engl J Med 375(23):
Drugs 77(18):1987–2001. https://doi.org/10.1007/s40265-017- 2293–2297. https://doi.org/10.1056/NEJMsb1609216
0835-9 14. Caporali R, Zavaglia D (2019) Real-world experience with
4. Burmester GR, Blanco R, Charles-Schoeman C, Wollenhaupt J, tofacitinib for the treatment of rheumatoid arthritis. Clin Exp
Zerbini C, Benda B, Gruben D, Wallenstein G, Krishnaswami S, Rheumatol 37(3):485–495
Zwillich SH, Koncz T, Soma K, Bradley J, Mebus C (2013) 15. Roberts L, Arnold M, Bird P, Burnet S, De Jager J, Littlejohn G,
Tofacitinib (CP-690,550) in combination with methotrexate in pa- Nicholls D, Tymms K, Zochling J, Griffiths H (2011) ARA-P39
tients with active rheumatoid arthritis with an inadequate response Optimising patient outcomes in Australian rheumatology (OPAL) -
to tumour necrosis factor inhibitors: a randomised phase 3 trial. a quality use of medicines initiative (QUMI) with >7000 rheuma-
Lancet 381(9865):451–460. https://doi.org/10.1016/s0140- toid arthritis (RA) patients. Intern Med J 41:9–38. https://doi.org/
6736(12)61424-x 10.1111/j.1445-5994.2010.02466.x
5. Cohen SB, Tanaka Y, Mariette X, Curtis JR, Lee EB, Nash P, 16. Austin PC (2011) An introduction to propensity score methods for
Winthrop KL, Charles-Schoeman C, Thirunavukkarasu K, reducing the effects of confounding in observational studies.
DeMasi R, Geier J, Kwok K, Wang L, Riese R, Wollenhaupt J Multivariate Behav Res 46(3):399–424. https://doi.org/10.1080/
(2017) Long-term safety of tofacitinib for the treatment of rheuma- 00273171.2011.568786
toid arthritis up to 8.5 years: integrated analysis of data from the
17. Harnett J, Gerber R, Gruben D, Koenig AS, Chen C (2016)
global clinical trials. Annals of the Rheumatic Diseases 76(7):1253.
Evaluation of real-world experience with tofacitinib compared with
https://doi.org/10.1136/annrheumdis-2016-210457
adalimumab, etanercept, and abatacept in RA patients with 1 pre-
6. Fleischmann R, Kremer J, Cush J, Schulze-Koops H, Connell CA,
vious biologic DMARD: data from a U.S. administrative claims
Bradley JD, Gruben D, Wallenstein GV, Zwillich SH, Kanik KS
database. Journal of Managed Care & Specialty Pharmacy
(2012) Placebo-controlled trial of tofacitinib monotherapy in rheu-
22(12):1457–1471. https://doi.org/10.18553/jmcp.2016.22.12.
matoid arthritis. N Engl J Med 367(6):495–507. https://doi.org/10.
1457
1056/NEJMoa1109071
7. Fleischmann R, Mysler E, Hall S, Kivitz AJ, Moots RJ, Luo Z, 18. Finckh A, Herzog L, Scherer A, Dudler J, Moeller B, Ciurea A,
DeMasi R, Soma K, Zhang R, Takiya L, Tatulych S, Mojcik C, Mueller R, Hasler P, Exer P, Iv M, Kyburz D, Gabay C, Zufferey P
Krishnaswami S, Menon S, Smolen JS (2017) Efficacy and safety (2017) THU0174 Drug retention of tofacitinib versus biologic an-
of tofacitinib monotherapy, tofacitinib with methotrexate, and tirheumatic agents in rheumatoid arthritis: observational data from
adalimumab with methotrexate in patients with rheumatoid arthritis the swiss scqm registry. Ann Rheum Dis 76(Suppl 2):267. https://
(ORAL strategy): a phase 3b/4, double-blind, head-to-head, doi.org/10.1136/annrheumdis-2017-eular.6804
randomised controlled trial. Lancet 390(10093):457–468. https:// 19. Kavanaugh A, Reed GW, Saunders KC, Koenig AS, Geier JL,
doi.org/10.1016/s0140-6736(17)31618-5 Kremer JM, Greenberg JD, Bingham CO (2015) AB0439 Clinical
8. Kremer J, Li ZG, Hall S, Fleischmann R, Genovese M, Martin- characteristics of RA patients newly prescribed tofacitinib citrate
Mola E, Isaacs JD, Gruben D, Wallenstein G, Krishnaswami S, (tofacitinib) in the United States after food and drug administration
Zwillich SH, Koncz T, Riese R, Bradley J (2013) Tofacitinib in approval: results from the Corrona us rheumatoid arthritis registry.
combination with nonbiologic disease-modifying antirheumatic Ann Rheum Dis 74(Suppl 2):1041. https://doi.org/10.1136/
drugs in patients with active rheumatoid arthritis: a randomized annrheumdis-2015-eular.1760
trial. Ann Intern Med 159(4):253–261. https://doi.org/10.7326/ 20. Reed GW, Gerber RA, Shan Y, Takiya L, Dandreo KJ, Gruben D,
0003-4819-159-4-201308200-00006 Kremer JM, Wallenstein GV (2017) OP0022 TNFI and
9. Lee EB, Fleischmann R, Hall S, Wilkinson B, Bradley JD, Gruben TOFACITINIB monotherapy and comparative effectiveness in
D, Koncz T, Krishnaswami S, Wallenstein GV, Zang C, Zwillich clinical practice: results from Corrona registry. Ann Rheum Dis
SH, van Vollenhoven RF (2014) Tofacitinib versus methotrexate in 76(Suppl 2):60. https://doi.org/10.1136/annrheumdis-2017-eular.
rheumatoid arthritis. N Engl J Med 370(25):2377–2386. https://doi. 1939
org/10.1056/NEJMoa1310476 21. Bird P, Littlejohn G, Butcher B, Smith T, Pereira CDF, Witcombe
10. van der Heijde D, Tanaka Y, Fleischmann R, Keystone E, Kremer J, D, Griffiths H (2019) FRI0140 Evaluation of effectiveness and
Zerbini C, Cardiel MH, Cohen S, Nash P, Song YW, Tegzova D, usage patterns of tofacitinib in treatment of rheumatoid arthritis in
Wyman BT, Gruben D, Benda B, Wallenstein G, Krishnaswami S, Australia: an analysis from the OPAL-QUMI real world dataset.
Zwillich SH, Bradley JD, Connell CA (2013) Tofacitinib (CP-690, Ann Rheum Dis 78(Suppl 2):740. https://doi.org/10.1136/
550) in patients with rheumatoid arthritis receiving methotrexate: annrheumdis-2019-eular.5216
twelve-month data from a twenty-four-month phase III randomized 22. Bird P, Littlejohn G, Butcher B, Smith T, da Fonseca PC, Witcombe
radiographic study. Arthritis Rheum 65(3):559–570. https://doi.org/ D, Griffiths H (2019) Evaluation of effectiveness and usage patterns
10.1002/art.37816 of tofacitinib in treatment of rheumatoid arthritis in Australia: an
11. Wallenstein GV, Kanik KS, Wilkinson B, Cohen S, Cutolo M, analysis from the OPAL-QUMI real world dataset. Arthritis
Fleischmann RM, Genovese MC, Gomez Reino J, Gruben D, Rheumatol 71:suppl 10
Kremer J, Krishnaswami S, Lee EB, Pascual-Ramos V, Strand V,
Zwillich SH (2016) Effects of the oral Janus kinase inhibitor Publisher’s note Springer Nature remains neutral with regard to jurisdic-
tofacitinib on patient-reported outcomes in patients with active tional claims in published maps and institutional affiliations.
rheumatoid arthritis: results of two phase 2 randomised controlled
trials. Clin Exp Rheumatol 34(3):430–442

You might also like