ArticlePDF Available

Identification of two tofacitinib subpopulations with different relative risk versus TNF inhibitors: an analysis of the open label, randomised controlled study ORAL Surveillance

Authors:

Abstract and Figures

Objectives: Based on primary results from ORAL Surveillance, an event-driven clinical trial of risk-enriched patients, identify subpopulations with different relative risk (ie, 'high-risk' and 'low-risk') with tofacitinib versus tumour necrosis factor inhibitors (TNFi). Methods: Patients with rheumatoid arthritis aged ≥50 years with ≥1 additional cardiovascular risk factor received tofacitinib 5 or 10 mg two times a day or TNFi. Prior analyses had identified age and smoking as risk factors of particular interest across safety outcomes. Hazard ratios (HRs) and incidence rates were evaluated by age and smoking individually and in combination. Results were validated across tofacitinib development programmes. Results: 'Age ≥65 years or ever smoker' defined a group ('high-risk') with increased risk of malignancies (excluding non-melanoma skin cancer), major adverse cardiovascular events, myocardial infarction, venous thromboembolism and all-cause death with tofacitinib (combined doses) versus TNFi (HRs 1.41-5.19). In patients 'aged <65 years and never smokers' ('low-risk'), there was no detectable risk increase with tofacitinib versus TNFi (HRs ≈1.0) up to 6 years of follow-up, and absolute risk remained low and was corroborated across tofacitinib rheumatoid arthritis, psoriatic arthritis and ulcerative colitis programmes with up to 10 years of observation. Conclusions: This posthoc analysis of ORAL Surveillance identified two tofacitinib subpopulations with different relative risk versus TNFi. High risk was confined to patients defined by distinct risk factors age ≥65 years or smoking, and these differentiating risk factors accounted for the excess risk observed with tofacitinib versus TNFi. These findings can guide individualised benefit/risk assessment and clinical decision-making on treatment with tofacitinib. Trial registration numbers: NCT02092467, NCT01262118, NCT01484561, NCT00147498, NCT00413660, NCT00550446, NCT00603512, NCT00687193, NCT01164579, NCT00976599, NCT01059864, NCT01359150, NCT02147587, NCT00960440, NCT00847613, NCT00814307, NCT00856544, NCT00853385, NCT01039688, NCT02281552, NCT02187055, NCT02831855, NCT00413699, NCT00661661, NCT00787202, NCT01465763, NCT01458951, NCT01458574, NCT01470612, NCT01877668, NCT01882439, NCT01976364.
Content may be subject to copyright.
901
Kristensen LE, etal. Ann Rheum Dis 2023;82:901–910. doi:10.1136/ard-2022-223715
Rheumatoid arthritis
CLINICAL SCIENCE
Identification of two tofacitinib subpopulations with
different relative risk versus TNF inhibitors: an analysis of
the open label, randomised controlled study ORAL
Surveillance
Lars Erik Kristensen,1 Silvio Danese,2 Arne Yndestad,3 Cunshan Wang,4 Edward Nagy,5
Irene Modesto,6 Jose Rivas,6 Birgitta Benda7
To cite: KristensenLE,
DaneseS, YndestadA,
etal. Ann Rheum Dis
2023;82:901–910.
Handling editor Josef S
Smolen
Additional supplemental
material is published online
only. To view, please visit
the journal online (http:// dx.
doi. org/ 10. 1136/ ard- 2022-
223715).
1The Parker Institute, Bispebjerg
and Frederiksberg Hospital,
University of Copenhagen,
Copenhagen, Denmark
2Gastroenterology and
Endoscopy, IRCCS San Raffaele
Hospital and Vita-Salute San
Raffaele University, Milan, Italy
3Pfizer Inc, Oslo, Norway
4Pfizer Inc, Groton, Connecticut,
USA
5Pfizer Ltd, Tadworth, UK
6Pfizer SLU, Madrid, Spain
7Pfizer Inc, New York, New
York, USA
Correspondence to
Dr Jose Rivas, Pfizer SLU, Madrid
28108, Spain;
joseluis. rivas@ pfizer. com
Received 2 December 2022
Accepted 8 March 2023
Published Online First
17March2023
Watch Video
ard. bmj. com
© Author(s) (or their
employer(s)) 2023. Re- use
permitted under CC BY- NC. No
commercial re- use. See rights
and permissions. Published
by BMJ.
ABSTRACT
Objectives Based on primary results from ORAL
Surveillance, an event- driven clinical trial of risk- enriched
patients, identify subpopulations with different relative
risk (ie, ’high- risk’ and ’low- risk’) with tofacitinib versus
tumour necrosis factor inhibitors (TNFi).
Methods Patients with rheumatoid arthritis aged
≥50 years with ≥1 additional cardiovascular risk factor
received tofacitinib 5 or 10 mg two times a day or TNFi.
Prior analyses had identified age and smoking as risk
factors of particular interest across safety outcomes.
Hazard ratios (HRs) and incidence rates were evaluated
by age and smoking individually and in combination.
Results were validated across tofacitinib development
programmes.
Results ’Age ≥65 years or ever smoker’ defined a
group (’high- risk’) with increased risk of malignancies
(excluding non- melanoma skin cancer), major adverse
cardiovascular events, myocardial infarction, venous
thromboembolism and all- cause death with tofacitinib
(combined doses) versus TNFi (HRs 1.41–5.19). In
patients ’aged <65 years and never smokers’ (’low- risk’),
there was no detectable risk increase with tofacitinib
versus TNFi (HRs ≈1.0) up to 6 years of follow- up, and
absolute risk remained low and was corroborated across
tofacitinib rheumatoid arthritis, psoriatic arthritis and
ulcerative colitis programmes with up to 10 years of
observation.
Conclusions This posthoc analysis of ORAL Surveillance
identified two tofacitinib subpopulations with different
relative risk versus TNFi. High risk was confined to
patients defined by distinct risk factors age ≥65 years or
smoking, and these differentiating risk factors accounted
for the excess risk observed with tofacitinib versus TNFi.
These findings can guide individualised benefit/risk
assessment and clinical decision- making on treatment
with tofacitinib.
Trial registration numbers NCT02092467,
NCT01262118, NCT01484561, NCT00147498,
NCT00413660, NCT00550446, NCT00603512,
NCT00687193, NCT01164579, NCT00976599,
NCT01059864, NCT01359150, NCT02147587,
NCT00960440, NCT00847613, NCT00814307,
NCT00856544, NCT00853385, NCT01039688,
NCT02281552, NCT02187055, NCT02831855,
NCT00413699, NCT00661661, NCT00787202,
NCT01465763, NCT01458951, NCT01458574,
NCT01470612, NCT01877668, NCT01882439,
NCT01976364.
INTRODUCTION
ORAL Surveillance was a large, randomised,
open- label, event- driven clinical trial in patients
WHAT IS ALREADY KNOWN ON THIS TOPIC
Primary findings from ORAL Surveillance
indicated that patients with rheumatoid
arthritis aged ≥50 years with ≥1 additional
cardiovascular risk factor have an increased risk
of major adverse cardiovascular events (MACE)
and malignancies (excluding non- melanoma
skin cancer) with tofacitinib compared with
tumour necrosis factor inhibitors (TNFi).
Increased risk of MACE with tofacitinib versus
TNFi was primarily observed in patients with a
history of atherosclerotic cardiovascular disease.
Previous analyses from ORAL Surveillance
identified age and smoking as independent (ie,
across treatment groups) risk factors of interest
across safety outcomes.
WHAT THIS STUDY ADDS
This posthoc analysis of ORAL Surveillance
identified high- risk and low- risk populations
with different relative risk with tofacitinib
versus TNFi.
Higher risk versus TNFi was confined to a
subgroup of patients defined by distinct, readily
identifiable risk factors, age 65 years or older
and long- time smoking (current or past).
In ‘low- risk’ patients who were younger than
65 years and had never smoked, there was no
detectable risk increase versus TNFi with up
to 6 years of follow- up in ORAL Surveillance,
and absolute risk remained low and was
corroborated across tofacitinib rheumatoid
arthritis, psoriatic arthritis and ulcerative colitis
development programmes with up to 10 years
of observations.
HOW THIS STUDY MIGHT AFFECT RESEARCH,
PRACTICE OR POLICY
These easily identifiable and clinically practical
subpopulations with different relative risk
versus TNFi (ie, ‘high- risk’ and ‘low- risk’)
can better guide individualised benefit/risk
assessment and clinical decision- making on
treatment with tofacitinib.
on July 4, 2023 by guest. Protected by copyright.http://ard.bmj.com/Ann Rheum Dis: first published as 10.1136/ard-2022-223715 on 17 March 2023. Downloaded from
902 Kristensen LE, etal. Ann Rheum Dis 2023;82:901–910. doi:10.1136/ard-2022-223715
Rheumatoid arthritis
with rheumatoid arthritis (RA) designed to demonstrate non-
inferiority of tofacitinib versus TNF inhibitors (TNFi) for the
coprimary endpoints of adjudicated major adverse cardiovas-
cular events (MACE) and adjudicated malignancies (excluding
non- melanoma skin cancer (NMSC)).1 The U.S. FDA required
the study to be of sufficient size and duration to evaluate long-
latency and rare events.2 For the study to be declared complete,
≥1500 patients had to be followed for ≥3 years, and ≥103
MACE, and ≥138 malignancies (excluding NMSC), had to
accrue.1 The study started in 2014, enrolled 4362 patients and
completed 6 years later.
To ensure enough cardiovascular (CV) and malignancy events
accrued in a reasonable timeframe, ORAL Surveillance enrolled
patients with RA with higher- than- average risk; patients had to
be at least 50 years old and have at least one additional CV risk
factor.1 Previous studies have shown a relationship and shared
risk factors between CV disease and malignancy, for example,
data from Dutch and US cohorts found an association between
the 10- year atherosclerotic cardiovascular disease (ASCVD) risk
scores and risk of future cancer.3 4 Importantly, a wide range of
CV risk factors were applied as eligibility criteria, therefore, a
52- year- old with mild hypertension and a 71- year- old with prior
myocardial infarction (MI) would both have been eligible despite
very different risk levels. Indeed, enrolled patients were found to
be distributed across a continuum of risk.5 For example, close to
one- third of the overall ORAL Surveillance study population had
only low- to- borderline predicted 10- year risk of ASCVD.5
Guided by initial thematic analyses of clinically meaningful
factors in ORAL Surveillance, in which age and smoking were
consistently identified as independent (ie, across treatment
groups) risk factors of particular interest across safety outcomes,
we aimed to find easily identifiable and clinically practical
subpopulations with different relative risk vs TNFi (ie, ‘high- risk’
and ‘low- risk’) to better guide individualised benefit/risk assess-
ment and clinical decision- making. For risk minimisation and
product labelling purposes, it was important to identify the risk
factors (individual or combinations thereof) accounting for the
increased risk observed with tofacitinib versus TNFi (ie, differ-
entiating risk factors) and, equally important, where the reverse
was true, that is, the absence of these risk factors produced risk
estimates with no apparent risk difference between treatments
(ie, HR at or below 1). Analyses were repeated in the larger and
longer tofacitinib RA development programme, and tofacitinib
psoriatic arthritis (PsA) and ulcerative colitis (UC) development
programmes to assess consistency and validate results.
METHODS
Study design and patients
ORAL Surveillance
ORAL Surveillance (NCT02092467) was a phase IIIb/IV
randomised, open- label, non- inferiority, safety endpoint study
conducted from March 2014 to July 2020 in patients with active
moderate- to- severe RA despite methotrexate treatment who
were aged ≥50 years with ≥1 additional CV risk factor (current
smoking, hypertension, high- density lipoprotein cholesterol
<40 mg/dL, diabetes mellitus, family history of premature coro-
nary heart disease, RA- associated extra- articular disease and/or
history of coronary artery disease).1
Patients were randomised 1:1:1 to receive oral tofacitinib 5
or 10 mg two times a day or subcutaneous TNFi (adalimumab
40 mg every 2 weeks (North America) or etanercept 50 mg
once a week (rest of the world)). All patients continued their
pre- study stable dose of methotrexate unless modification was
clinically indicated. In February 2019, the tofacitinib 10 mg two
times a day dose was reduced to 5 mg two times a day after the
Data Safety Monitoring Board noted an increased frequency of
pulmonary embolism in patients receiving tofacitinib 10 mg two
times a day versus TNFi and an increase in overall mortality with
tofacitinib 10 vs 5 mg two times a day and TNFi.
RA, PsA and UC development programmes
This exploratory analysis includes data for patients who
received ≥1 dose of tofacitinib in clinical trials and open- label
LTE studies across RA (excluding ORAL Surveillance), PsA and
UC. Full details of the individual studies can be found in online
supplemental table S3.
All cohorts included patients aged ≥18 years who received
tofacitinib as monotherapy (RA and UC) or with background
conventional synthetic disease- modifying antirheumatic drugs
(RA, PsA). All studies have been completed. Final data for the
RA, PsA and UC cohorts are from 18 January 2019, 31 July
2019 and 24 August 2020, respectively.
Patient and public involvement
Patients and/or the public were not involved in the design,
conduct, reporting or dissemination plans of this research.
Outcomes
This analysis focused on events of malignancies (excluding
NMSC), MACE, MI (fatal and non- fatal), venous thromboem-
bolism (VTE) and all- cause death. In ORAL Surveillance, these
events were adjudicated by an external, independent adjudi-
cation committee. MACE was defined as the composite of CV
death, non- fatal MI and non- fatal stroke. Similarly, events of
malignancies (excluding NMSC), MACE and MI were adjudi-
cated in the tofacitinib development programmes. See online
supplemental table S1) for definition of outcomes.
Statistical analyses
ORAL Surveillance was powered to assess non- inferiority for risk
of MACE and malignancies (excluding NMSC) with combined
tofacitinib doses versus TNFi. To maximise statistical precision
and power in these posthoc analyses, data for the combined
doses of tofacitinib were prioritised. HRs and two- sided 95%
CIs comparing tofacitinib and TNFi were estimated using Cox
proportional hazard regression models. Incidence rates (IRs) and
two- sided 95% CIs for malignancies (excluding NMSC), MACE,
MI, VTE and all- cause death were reported as the number of
unique patients with events per 100 patient- years. See online
supplemental table S1 for definition of censoring times for the
different analyses. Exact Poisson, adjusted for exposure, was
used to calculate 95% CIs for the crude IR.
Prior analyses had consistently identified age and smoking
as independent risk factors of particular interest across safety
outcomes and were therefore assessed here, individually and
in combination.1 6 7 Informed by the definition of the geriatric
population from the ICH Topic E7 (Studies in Support of Special
Populations: Geriatrics), a cut- off of 65 years of age was a pre-
specified analysis in ORAL Surveillance and also applied in these
analyses.1 8 IRs and HRs with tofacitinib versus TNFi were also
determined by 5- year intervals of age.
Further analyses were conducted to identify risk factors
accounting for the increased risk observed with tofacitinib
versus TNFi (ie, differentiating risk factors) while when absent
produced tofacitinib risk estimates with no difference versus
TNFi (ie, HR≈1.0). HRs (95% CIs) comparing tofacitinib and
on July 4, 2023 by guest. Protected by copyright.http://ard.bmj.com/Ann Rheum Dis: first published as 10.1136/ard-2022-223715 on 17 March 2023. Downloaded from
903
Kristensen LE, etal. Ann Rheum Dis 2023;82:901–910. doi:10.1136/ard-2022-223715
Rheumatoid arthritis
TNFi (ORAL Surveillance) and IRs (95% CIs) for each treatment
group were calculated for subgroups of patients defined by age
(≥ or <65 years of age), or by smoking status (current, past or
never smoking), or by composites thereof.
For the subgroups defined by ‘age ≥65 years or ever smoker’
(‘high- risk’) and ‘age <65 years and never smoker’ (‘low- risk’),
number needed to harm (NNH) was calculated as the reciprocal
of the difference in IRs between tofacitinib and TNFi. Positive
NNH was defined as patient- years of tofacitinib exposure needed
for one more patient to report an additional event versus TNFi.
Negative NNH was defined as the reverse. The differential or
interaction effect of treatment group (tofacitinib vs TNFi) and
high- risk versus low- risk was assessed using the difference of the
incidence rate difference (IRD, comparing tofacitinib and TNFi)
between high- risk and low- risk and its SE. The two- sided inter-
action p value was calculated assuming normal approximation to
the difference of IRD. In published criteria for how to evaluate
the credibility of subgroup analyses, it is advised to not use a
specified p value threshold, but p<0.1 are generally supportive
of the hypothesis.9 Cumulative probability plots using Kaplan-
Meier estimates were generated for analysis of time to events.
IRs (95% CI) of all outcomes were determined by high- risk
and low- risk in the tofacitinib RA, PsA and UC development
programmes.
All analyses were posthoc. Across these exploratory analyses,
no multiplicity adjustments were applied.
RESULTS
Patients
In ORAL Surveillance, 4362 patients were randomised and
treated (tofacitinib 5 mg two times a day, n=1455; tofacitinib
10 mg two times a day, n=1456; TNFi, n=1451).1 In addi-
tion, this analysis includes 9904 tofacitinib- exposed patients
from completed studies in the development programmes:
7964 with RA (excluding ORAL Surveillance), 783 with PsA
and 1157 with UC.10 Table 1 (online supplemental table S2
by low- risk/high- risk) summarises demographics and base-
line characteristics for patients in ORAL Surveillance and
in all patients who received ≥1 dose of tofacitinib in the
RA, PsA and UC development programmes. Compared with
the tofacitinib development programmes, patients included
in ORAL Surveillance represented a risk- enriched popu-
lation as reflected in a higher proportion of patients aged
≥65 years, current smokers and patients with a history of
diabetes, hypertension and ASCVD (table 1).
Tofacitinib relative risk versus TNFi and role of age and
smoking as individual risk factors
Age ≥65 years (figure 1A) and ever (ie, current or past)
smoking (figure 1B) were both independent overall risk
factors with associations with absolute (IRs) and relative
(HRs) risk with tofacitinib versus TNFi of malignancies
(excluding NMSC), MACE, MI, VTE and all- cause death
in ORAL Surveillance.1 6 7 Even though less pronounced,
an increased risk versus TNFi (HRs >1) was observed in
patients who were <65 years as well as in never smokers for
certain outcomes.
Analyses of risk with tofacitinib by 5- year age intervals indi-
cated a risk continuum with increasing age where the inflection
point seemed to appear at or around ≥65 years of age (online
supplemental figure S1).
An analysis of smoking duration showed that the majority
of current and past smokers in ORAL Surveillance were
long- time smokers. More than 90% of patients treated with
tofacitinib in ORAL Surveillance that had ever smoked (ie,
current or past smokers) had a smoking duration of more
Table 1 Demographic and baseline disease characteristics of patients in ORAL Surveillance and the tofacitinib RA, PsA and UC development
programmes
ORAL Surveillance Tofacitinib development programme
N=4362
RA*
N=7964
PsA
N=783
UC
N=1157
Female, % (n) 78.2% (3410) 81.9% (6522) 54.7% (428) 41.3% (478)
Duration of disease (years), mean/median 10.4/8.0 8.1/5.6 7.7/5.5 8.2/6.3
Age, mean (SD) 61.2 (7.1) 52.6 (12.1) 48.7 (12.0) 41.3 (13.9)
≥65 years of age, % (n) 31.0% (1353) 15.9% (1270) 9.2% (72) 6.7% (77)
Smoking status†, % (n)
Current 26.7% (1166) 17.2% (1366) 17.9% (140) 5.1% (59)
Past 21.5% (937) 17.4% (1388) 20.2% (158) 30.9% (357)
Never 51.8% (2259) 62.7% (4996) 61.9% (485) 64.0% (740)
History of other CV risk factors, % (n)
Diabetes mellitus 17.4% (759) 8.2% (651) 13.7% (107) 4.1% (48)‡
Hyperlipidaemia 35.2% (1534) 19.3% (1534) 21.3% (107) NA
Hypertension 66.0% (2878) 35.4% (2818) 39.1% (306) 13.9% (161)‡
Coronary artery disease 11.4% (497) 1.6% (126) 5.6% (44) 1.6% (18)
ASCVD 14.7% (640) 3.4% (274) 6.5% (51) 3.9% (45)
Treatment history, % (n)
Prior TNFi 7.6% (330) 15.6% (1245) 48.1% (377) 54.4% (1124)
Statin at baseline‡ 23.4% (1020) 7.8% (620) 12.8% (100) 6.4% (74)
Aspirin at baseline‡ 15.3% (667) 6.9% (551) 6.4% (50) NA
*Excluding ORAL Surveillance.
†In the tofacitinib RA development programme, 2.7% (N=214) of patients had unknown smoking status. Patients <65 years old with unknown smoking status were not included in the low- risk
group. 25 patients in the high- risk group had unknown smoking status.
‡Based on day 1 of treatment.
ASCVD, atherosclerotic cardiovascular disease; CV, cardiovascular; n, number of patients with characteristic; NA, not available; RA, rheumatoid arthritis; TNFi, tumour necrosis factor inhibitor; UC,
ulcerative colitis.
on July 4, 2023 by guest. Protected by copyright.http://ard.bmj.com/Ann Rheum Dis: first published as 10.1136/ard-2022-223715 on 17 March 2023. Downloaded from
904 Kristensen LE, etal. Ann Rheum Dis 2023;82:901–910. doi:10.1136/ard-2022-223715
Rheumatoid arthritis
Figure 1 Risk of malignancies (excluding NMSC), MACE, MI, VTE and all- cause death with tofacitinib versus TNFi in ORAL Surveillance by (A) age
(≥ or <65 years) or (B) history of smoking (current, past or never) or (C) composite of age and smoking (‘Age ≥65 years and Ever smoked’ or ‘Age <65
years or Never smoked’). HRs (95% CIs), shown on a logarithmic scale, are based on a simple Cox proportional hazard model comparing combined
tofacitinib doses versus TNFi. Arrow heads indicate that CI extends beyond the graph axis. IRs express the number of patients with first events per 100
PY. IRs, n and PY are for combined tofacitinib doses. *Results previously reported in Ytterberg et al.1 †Results reported in Curtis et al.7 IR, incidence
rate; MACE, major adverse cardiovascular events; n, number of evaluable patients; N, number of patients with events; NMSC, non- melanoma skin
cancer; PY, patient- years; TNFi, tumour necrosis factor inhibitor; VTE, venous thromboembolism.
on July 4, 2023 by guest. Protected by copyright.http://ard.bmj.com/Ann Rheum Dis: first published as 10.1136/ard-2022-223715 on 17 March 2023. Downloaded from
905
Kristensen LE, etal. Ann Rheum Dis 2023;82:901–910. doi:10.1136/ard-2022-223715
Rheumatoid arthritis
than 10 years. Median duration of smoking in current and
past smokers was 35.0 and 39.0 years, respectively. Majority
of past smokers had been smoke- free for 10 years or longer
(table 2).
Tofacitinib relative risk versus TNFi and composites of age
and smoking as differentiating risk factors
Figures 1C and 2 show the relative risk (HRs) for malig-
nancies (excluding NMSC), MACE, MI, VTE and all- cause
death with tofacitinib versus TNFi in subgroups defined by
composites of age and smoking. In patients with both risk
factors, that is, ‘age≥65 years and ever smoker’ (figure 1C),
or one risk factor, that is, ‘age ≥65 years or ever smoker’
(figure 2; online supplemental figure S2 by tofacitinib dose),
the risk was higher with tofacitinib versus TNFi, and for
some endpoints, the 95% CI for the HR excluded 1. It was
only in patients with neither risk factor, that is, ‘age <65
years and never smoker’ group that we could not detect a
higher risk with tofacitinib vs TNFi (ie, HRs≈1.0) for any
of these events (figure 2). Based on IRD, the p values for
the treatment- by- risk high/low interaction for these events
with combined doses of tofacitinib versus TNFi ranged from
0.002 to 0.173 (figure 2), supporting that this composite of
age ≥65 years or ever smoker represents a differentiating
risk factor for these events.9
Table 2 Smoking duration in current and past smokers and time since smoking cessation in past smokers in ORAL Surveillance
Current smokers Past smokers
Tofacitinib
N=811
TNFi
N=352
Tofacitinib
N=605
TNFi
N=322
Smoking Duration (years)
Mean (SD) 32.7 (13.1) 31.9 (13.2) 37.2 (13.0) 39.9 (12.2)
Median (Range) 35.0 (0.02, 67.00) 34.2 (0.02, 69.00) 39.0 (0.00, 69.00) 41.0 (1.00, 70.00)
Smoking duration levels*, % (n)
>10 years 91.4% (741) 91.2% (321) 96.2% (582) 98.4% (317)
>5–10 years 4.4% (36) 3.7% (13) 2.3% (14) 0.9% (3)
>0–5 years 4.2% (34) 5.1% (18) 1.5% (9) 0.6% (2)
Time since smoking cessation levels, % (n)
≥10 years 61.5% (372) 71.7% (231)
<10 years 38.5% (233) 28.3% (91)
<5 years 21.8% (132) 15.2% (49)
<1 year 5.6% (34) 3.1% (10)
*Information on smoking duration was missing on eight patients (two current and six past smokers) treated with tofacitinib and five patients (one current and four past smokers) treated with TNFi.
n, number of patients with characteristic; TNFi, tumour necrosis factor inhibitor.
Figure 2 Risk of malignancies (excluding NMSC), MACE, MI, VTE and all- cause death with tofacitinib versus TNFi in ORAL Surveillance by subgroups
of high- risk and low- risk patients. HRs (95% CIs), shown on a logarithmic scale, are based on a simple Cox proportional hazard model comparing
combined tofacitinib doses versus TNFi. Arrow heads indicate that CI extends beyond the graph axis. IRs express the number of patients with first
events per 100 PY. Treatment- by- risk interaction p values were calculated based on IR differences (two- sided, normal approximation of difference in
IR). NNH (PY) should be interpreted as the number of patient- years of exposure to tofacitinib required to have one additional event versus TNFi. All
data are for combined tofacitinib doses. *Results reported in Curtis et al.7 IR, incidence rate; MACE, major adverse cardiovascular events; N, number
of evaluable patients; n, number of patients with events; NMSC, non- melanoma skin cancer; NNH, numbers needed to harm; PY, patient- years; TNFi,
tumour necrosis factor inhibitor; VTE, venous thromboembolism.
on July 4, 2023 by guest. Protected by copyright.http://ard.bmj.com/Ann Rheum Dis: first published as 10.1136/ard-2022-223715 on 17 March 2023. Downloaded from
906 Kristensen LE, etal. Ann Rheum Dis 2023;82:901–910. doi:10.1136/ard-2022-223715
Rheumatoid arthritis
Cumulative probability of events with tofacitinib and TNFi in
high-risk and low-risk subpopulations
Age ≥65 years or ever smokerand ‘age <65 years and never
smoker’ are hereafter referred to as ‘high- risk’ and ‘low- risk’,
respectively. To further assess absolute and relative risk of malig-
nancies (excluding NMSC), MACE, MI, VTE and all- cause
death with tofacitinib versus TNFi over time in high- risk and
low- risk patients, cumulative probability curves were gener-
ated (figure 3). These curves confirmed that there were two
tofacitinib subpopulations with different relative risk versus
TNFi: one subpopulation (‘high- risk’) of patients which had
higher risk with tofacitinib versus TNFi for all these outcomes
and one subpopulation (‘low- risk’) for which the curves repre-
senting treatment with tofacitinib and TNFi overlapped and/or
Figure 3 Cumulative probability of patients with malignancies (excluding NMSC), MACE, MI, VTE and all- cause death in ORAL Surveillance overall
population and by subgroups of high- risk and low- risk patients. Overall population received tofacitinib 5 mg or 10 mg two times a day (N=2911)
or TNFi (N=1451). High- risk patients were ≥65 years of age or ever smoker (tofacitinib, N=1895; TNFi, N=926). Low- risk patients were <65 years
of age and never smoker (tofacitinib, N=1016; TNFi, N=525). Cumulative probabilities of events were calculated based on Kaplan- Meier estimates.
Cumulative probability plots for malignancies (excluding NMSC) and MACE in overall population have been reported in Ytterberg et al1 and are
included for reference. MACE, major adverse cardiovascular events; MI, myocardial infarction; NMSC, non- melanoma skin cancer; TNFi, tumour
necrosis factor inhibitor; VTE, venous thromboembolism.
on July 4, 2023 by guest. Protected by copyright.http://ard.bmj.com/Ann Rheum Dis: first published as 10.1136/ard-2022-223715 on 17 March 2023. Downloaded from
907
Kristensen LE, etal. Ann Rheum Dis 2023;82:901–910. doi:10.1136/ard-2022-223715
Rheumatoid arthritis
crossed, and we could not detect a difference between tofacitinib
and TNFi up to 6 years of follow- up in ORAL Surveillance.
Absolute risk in low-risk patients in ORAL Surveillance
Absolute risk of malignancies (excluding NMSC), MACE, MI,
VTE and all- cause death in low- risk patients (figure 2) was low
as reflected in low event rates and IRs. NNH based on the IR
differences with tofacitinib versus TNFi indicated that 1485 and
9898 low- risk patients would need to be treated with tofacitinib
for 1 year to have one additional event of, respectively, malig-
nancies (excluding NMSC) and all- cause mortality versus TNFi.
For MACE, MI and VTE in low- risk patients, IRD and NNH
were negative and in favour of tofacitinib versus TNFi. The
relatively large NNH (positive or negative numbers) principally
reflect similar absolute risk with tofacitinib versus TNFi in low-
risk patients.
Absolute risk in low-risk patients in tofacitinib RA, PsA and
UC clinical development programmes
ORAL Surveillance was a substantial RA study, in terms of
number of patients and exposure time. However, it makes up
only a third of the overall tofacitinib experience in RA. Specifi-
cally, 34.1% (1016/2911) of patients treated with tofacitinib in
ORAL Surveillance were <65 years of age and never smokers,
and this population had low absolute risk and no detectable
excessive risk of malignancies (excluding NMSC), MACE, MI,
VTE and all- cause death versus TNFi. To increase the precision
of absolute low- risk estimates, we conducted additional anal-
yses in the tofacitinib development programmes of RA, PsA and
UC, collectively representing 25 437 patient years of tofacitinib
exposure and safety observations extending up to 10.5 years.10
52.7% (4198/7964) of patients in the tofacitinib RA develop-
ment programme were <65 years of age and never smokers,
representing more than 4000 patients and 13 497 patient years
of tofacitinib exposure in the low- risk group of interest (table 3).
In the tofacitinib PsA and UC development programmes, 56.4%
and 59.6% of patients were <65 years of age and never smokers.
Figure 4 shows absolute risk (ie, IRs (95% CIs)) of malig-
nancies (excluding NMSC), MACE, MI, VTE and all- cause
death in the overall population and in low- risk patients treated
with tofacitinib in the overall RA, PsA and UC development
programmes compared with absolute risk with TNFi and tofac-
itinib in ORAL Surveillance (RA controlled phase and high- risk
patient data in online supplemental figure S3). Consistently, IRs
for malignancies (excluding NMSC), MACE, MI, VTE and all-
cause death in low- risk patients treated with tofacitinib in the
RA development programme were similar to those observed in
low- risk patients in ORAL Surveillance; however, the precision
of the estimate was higher, that is, the 95% CIs were narrower.
Similarly, IRs of malignancies (excluding NMSC), MACE, MI,
VTE and all- cause death in the PsA and UC tofacitinib develop-
ment programmes in overall populations and low- risk patients
were similar to those observed in tofacitinib- treated RA low-
risk patients in ORAL Surveillance and the RA development
programme (figure 4).
DISCUSSION
Primary findings in ORAL Surveillance demonstrated a higher
incidence of MACE and malignancies with tofacitinib versus
TNFi.1 5 In this posthoc analysis, two clinically practical and
easily identifiable subpopulations were found that demarcated
tofacitinib- treated patients with increased risk of these events
from patients with similar risk relative to TNFi- treated patients.
Given the design of ORAL Surveillance, enough events accrued
to allow for posthoc identification of patients at higher risk
with tofacitinib versus TNFi. Initial analyses identified age and
smoking as potential independent risk factors, and more than
80% of tofacitinib- treated patients with MACE, MI, malignan-
cies (excluding NMSC), VTE or all- cause death were accounted
for by the combination of these two risk factors. Of note, ‘having
ever smoked’ was found to largely correspond to substantial
smoking history. Specifically, more than 90% of current or past
smokers in ORAL Surveillance were long- time smokers with
more than 10 years of smoking and a median smoking history
over 30 years on study entry.
We found an exacerbation in risk with the combination of
risk factors age 65 years or older and having ever smoked, and
patients with at least one of these risk factors had a dispropor-
tionately larger risk increase with tofacitinib versus TNFi, while
in patients without these two differentiating risk factors, we
could not detect a risk difference for malignancies (excluding
NMSC), MACE, MI, VTE or all- cause death between tofacitinib
and TNFi.
The risk difference for long- latency events between tofaci-
tinib and TNFi observed at the overall study population level
emerged after approximately two years of follow- up in ORAL
Surveillance. Since JAK inhibitors are used chronically, patients
without risk factors could potentially be exposed for prolonged
periods of time during which a small risk increase could become
clinically relevant. Analyses of absolute and relative risk over
time found that the excess risk with tofacitinib versus TNFi
was confined to patients defined by the composite of age ≥65
years or ever smoking (‘high- risk’), and these differentiating risk
factors accounted for the excess risk of malignancies (excluding
NMSC), MACE, MI, VTE or all- cause death observed with
tofacitinib versus TNFi. On the other hand, in patients who were
younger than 65 and had never smoked (‘low- risk’), but who
all had prevalent other CV risk factors per ORAL Surveillance
eligibility criteria, we could not detect a difference between
tofacitinib and TNFi even up to 6 years of follow- up which is
longer than the median drug survival in RA.11 This observa-
tion was consistent across outcomes including malignancies
(excluding NMSC), MACE, MI, VTE and all- cause death, and
Table 3 Tofacitinib treatment exposure in high- risk and low- risk
populations in ORAL Surveillance and RA, PsA, UC development
programmes
ORAL
Surveillance (RA)
Tofacitinib development programme
RA* PsA UC
Overall population
N 2911 7964 783 1157
Exposure (PY) 10 922 23 497 2038 2814
Follow- up; mean, max (years) 3.8, 6.1 3.0, 10.5 2.6, 4.8 2.4, 7.8
High- risk: ≥65 years or ever smoked
N 1895 3577 341 444
% 65.1% 44.9% 43.6% 38.4%
Exposure (PY) 6986 9961 837 1113
Low- risk: <65 years and never smoked
N 1016 4198 442 713
% 34.9% 52.7% 56.4% 61.6%
Exposure (PY) 3937 13 168 1201 1702
PY was calculated from the first dose of tofacitinib to the last contact date in ORAL Surveillance, and
from the first dose of tofacitinib to the last dose of tofacitinib for all other development programmes.
*Excluding ORAL Surveillance.
N, number of patients treated with tofacitinib; PsA, psoriatic arthritis; PY, patient- years; RA, rheumatoid
arthritis; UC, ulcerative colitis.
on July 4, 2023 by guest. Protected by copyright.http://ard.bmj.com/Ann Rheum Dis: first published as 10.1136/ard-2022-223715 on 17 March 2023. Downloaded from
908 Kristensen LE, etal. Ann Rheum Dis 2023;82:901–910. doi:10.1136/ard-2022-223715
Rheumatoid arthritis
the magnitude of absolute risk in the low- risk group remained
low over time and was similar to TNFi with large NNH.
The composite of age ≥65 years or ever smoker was strongly
associated with absolute risk of malignancies (excluding NMSC),
MACE, MI, VTE and all- cause death in ORAL Surveillance,
and, as mentioned above, with treatment with tofacitinib, most
events (>82%) occurred in the 65% of patients in this high-
risk subgroup. There were accordingly few events in the low- risk
Figure 4 Risk of malignancies (excluding NMSC), MACE, MI, VTE and all- cause death in low- risk populations in ORAL Surveillance and tofacitinib
clinical development programmes. Low- risk patients were <65 years of age and never smoker. Horizontal dotted line represents IR in low- risk
patients treated with tofacitinib in ORAL Surveillance. IRs express the number of patients with first events per 100 PY. All data are for combined
tofacitinib doses. *Excluding ORAL Surveillance. Data from ORAL Surveillance overall populations have previously been published and are included
for reference; malignancies (excluding NMSC) and MACE (Ytterberg et al1), MI (Charles- Schoeman et al5). Also, previously published are data from
the tofacitinib RA and PsA development programmes on MACE (Burmester et al25) and VTE (Mease et al26). IR, incidence rate; MACE, major adverse
cardiovascular events; MI, myocardial infarction; N, number of evaluable patients; n, number of patients with events; NMSC, non- melanoma skin
cancer; PsA, psoriatic arthritis; PY, patient- years; RA, rheumatoid arthritis; TNFi, tumour necrosis factor inhibitor; UC, ulcerative colitis; VTE, venous
thromboembolism.
on July 4, 2023 by guest. Protected by copyright.http://ard.bmj.com/Ann Rheum Dis: first published as 10.1136/ard-2022-223715 on 17 March 2023. Downloaded from
909
Kristensen LE, etal. Ann Rheum Dis 2023;82:901–910. doi:10.1136/ard-2022-223715
Rheumatoid arthritis
group, patients <65 years of age that had never smoked, which
made up 35% of the overall study population. Consequently, it
can be argued that there is not sufficient precision in the absolute
and relative risk estimates in this low- risk population in ORAL
Surveillance. Even though ORAL Surveillance was a large and
long study, it makes up only a third of the overall tofacitinib
experience (excluding ORAL Surveillance) in RA and therefore,
it was important to validate the absolute risk estimate in ORAL
Surveillance within the larger tofacitinib RA clinical programme
which extended up to 10 years and included more than 4000
patients in the low- risk group of interest. Importantly, we corrob-
orated the absolute risk estimates from ORAL Surveillance and
with higher precision, with data from the overall population
and the low- risk population of the more extensive tofacitinib
RA development programme. Moreover, the magnitude of the
absolute risk in the low- risk group was low also in relation to
published rates in randomised controlled trials and their LTEs in
RA populations treated with TNFi and other biologics,12–18 and
this finding was confirmed also in the other tofacitinib devel-
opment programmes of PsA and UC. A limitation of these anal-
yses is that the average follow- up time in the tofacitinib clinical
development programmes (3.0, 2.6 and 2.4 years in the RA, PsA
and UC programmes, respectively) was shorter than in ORAL
Surveillance (3.8 years).
Recognising limitations associated with the posthoc nature
of these results, multiple aspects were considered to improve
robustness and confirm validity of findings. First, only data for
the combined doses of tofacitinib (with larger number of patients
and events vs per individual dose) were used for increased preci-
sion. In ORAL Surveillance, p values (0.002–0.173 for the
outcomes) for the interactions between treatment groups and
subgroups of high- risk/low- risk patients lent support to the
presence of a differential treatment effect for the two subgroups
of patients. Second, the analyses are based on risk factors (ie,
older age and smoking) already identified as major risk factors
of malignancies, MACE and VTE in the general and RA popula-
tions.3 19–24 Finally, we cannot, based on these posthoc analyses,
rule out that there is an increased relative risk of safety outcomes
with tofacitinib versus TNFi in the low- risk group. However, if
such an increased risk is present, we show that the absolute risk
is low, as indicated by low or no difference in IRs over time and
high NNH. Moreover, we have recently reported on history of
ASCVD as another differentiating risk factor.5 However, whereas
the combination of age and smoking is capable of differentiating
risk across major outcomes, history of ASCVD is specific for
MACE. These are all factors that need to be considered in an
individualised benefit/risk assessment.
The analyses presented herein had a particular focus on the
identification of a high- risk and a low- risk population. Future
investigations should aim at assessing whether the high- risk
population can be further segmented into different relative risk
levels.
In summary, ORAL Surveillance identified a high- risk and
low- risk tofacitinib population with different relative risk vs
TNFi. Higher risk versus TNFi was confined to a subgroup of
patients defined by distinct, readily identifiable risk factors, age
65 years or older and long- time smoking (current or past), and
these differentiating risk factors accounted for the excess risk
observed with tofacitinib versus TNFi. In ‘low- risk’ patients who
were younger than 65 and had never smoked, but who all had
prevalent other CV risk factors per ORAL Surveillance inclusion
criteria, there was no detectable risk increase vs TNFi with up
to 6 years of follow- up in ORAL Surveillance and the magni-
tude of absolute risk remained low and was corroborated across
tofacitinib programmes with up to 10 years of observations. It
is acknowledged that the findings are posthoc, nevertheless, the
results are clinically important and appear generalizable. These
findings can guide individualised benefit/risk assessment and
clinical decision- making on treatment with tofacitinib.
Acknowledgements Select data in this manuscript were previously presented at
ACR Convergence 2021.6 The authors would like to thank the patients, investigators
and study teams involved in the study. The authors would like to thank Hernan
Valdez for invaluable scientific discussions and guidance and Joseph Wu and
Kenneth Kwok, employees and shareholders of Pfizer Inc, for their contribution to
the statistical analyses.
Contributors LEK, SD, AY, CW, EN, IM, JR and BB conceived or designed the study
and data analyses. CW analysed the data. All authors had access to the data, were
involved in interpretation of data and reviewed and approved the manuscript’s
content before submission. LEK accepts final responsibility for this work and
controlled the decision to publish.
Funding This study was sponsored by Pfizer Inc.
Competing interests LEK has received fees for speaking and consultancy from
Pfizer, AbbVie, Amgen, Galapagos, UCB, Gilead, Biogen, BMS, MSD, Novartis, Eli Lilly
and Janssen pharmaceuticals. LEK has received IIT research grants from Novo, UCB,
Eli Lilly, Novartis and AbbVie. SD reports consultancy fees from AbbVie, Alimentiv,
Allergan, Amgen, Applied Molecular Transport, AstraZeneca, Athos Therapeutics,
Biogen, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Celltrion, Dr Falk
Pharma, Eli Lilly, Enthera, Ferring Pharmaceuticals Inc., Gilead, Hospira, Inotrem,
Janssen, Johnson & Johnson, Morphic, MSD, Mundipharma, Mylan, Pfizer, Roche,
Sandoz, Sublimity Therapeutics, Takeda, Teladoc Health, TiGenix, UCB Inc., Vial,
Vifor. SD reports lecture fees from AbbVie, Amgen, Ferring Pharmaceuticals Inc.,
Gilead, Janssen, Mylan, Pfizer, Takeda. AY, CW, EN, IM, JR and BB are employees and
stockholders of Pfizer Inc.
Patient and public involvement Patients and/or the public were not involved in
the design, or conduct, or reporting, or dissemination plans of this research.
Patient consent for publication Not applicable.
Ethics approval All studies included in this manuscript were conducted in
accordance with the Declaration of Helsinki, International Council on Harmonisation
Guidelines for Good Clinical Practice and local country regulations and were
approved by each centre’s Institutional Review Board or Independent Ethics
Committee. Participants gave informed consent to participate in the study before
taking part.
Provenance and peer review Not commissioned; externally peer reviewed.
Data availability statement Data are available on reasonable request. On
request, and subject to review, Pfizer will provide the data that support the findings
of this study. Subject to certain criteria, conditions and exceptions, Pfizer may also
provide access to the related individual deidentified participant data. See https://
www.pfizer.com/science/clinical-trials/trial-data-and-results for more information.
Supplemental material This content has been supplied by the author(s). It
has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have
been peer- reviewed. Any opinions or recommendations discussed are solely those
of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and
responsibility arising from any reliance placed on the content. Where the content
includes any translated material, BMJ does not warrant the accuracy and reliability
of the translations (including but not limited to local regulations, clinical guidelines,
terminology, drug names and drug dosages), and is not responsible for any error
and/or omissions arising from translation and adaptation or otherwise.
Open access This is an open access article distributed in accordance with the
Creative Commons Attribution Non Commercial (CC BY- NC 4.0) license, which
permits others to distribute, remix, adapt, build upon this work non- commercially,
and license their derivative works on different terms, provided the original work is
properly cited, appropriate credit is given, any changes made indicated, and the use
is non- commercial. See:http://creativecommons.org/licenses/by-nc/4.0/.
REFERENCES
1 Ytterberg SR, Bhatt DL, Mikuls TR, etal. Cardiovascular and cancer risk with
tofacitinib in rheumatoid arthritis. N Engl J Med 2022;386:316–26.
2 FDA. Approval letter - xeljanz (tofacitinib); 2012. Available: https://www.accessdata.
fda.gov/drugsatfda_docs/nda/2012/203214Orig1s000Approv.pdf
3 Koene RJ, Prizment AE, Blaes A, etal. Shared risk factors in cardiovascular disease and
cancer. Circulation 2016;133:1104–14.
4 Lau ES, Paniagua SM, Liu E, etal. Cardiovascular risk factors are associated with
future cancer. JACC CardioOncol 2021;3:48–58.
5 Charles- Schoeman C, Buch MH, Dougados M, etal. Risk of major adverse
cardiovascular events with tofacitinib versus tumour necrosis factor inhibitors
on July 4, 2023 by guest. Protected by copyright.http://ard.bmj.com/Ann Rheum Dis: first published as 10.1136/ard-2022-223715 on 17 March 2023. Downloaded from
910 Kristensen LE, etal. Ann Rheum Dis 2023;82:901–910. doi:10.1136/ard-2022-223715
Rheumatoid arthritis
in patients with rheumatoid arthritis with or without a history of atherosclerotic
cardiovascular disease: a post hoc analysis from oral surveillance. Ann Rheum Dis
2023;82:119–29.
6 Charles- Schoeman C, Buch M, Dougados M, etal. Risk factors for major adverse
cardiovascular events in patients aged ≥ 50 years with RA and ≥ 1 additional
cardiovascular risk factor: results from a phase 3b/4 randomized safety study of
tofacitinib vs TNF inhibitors [abstract]. Arthritis Rheumatol 2021;73 (suppl 9).
7 Curtis JR, Yamaoka K, Chen Y- H, etal. Malignancy risk with tofacitinib versus TNF
inhibitors in rheumatoid arthritis: results from the open- label, randomised controlled
oral surveillance trial. Ann Rheum Dis 2023;82:331–43.
8 European medicines agency. ICH topic E 7. studies in support of special populations:
geriatrics; Available: https://www.ema.europa.eu/en/ich-e7-studies-support-special-
populations-geriatrics-scientific-guideline#current-effective-version-section1994
[Accessed 27 Mar 2021].
9 Sun X, Briel M, Walter SD, etal. Is a subgroup effect believable? updating criteria to
evaluate the credibility of subgroup analyses. BMJ 2010;340:bmj.c117.
10 Burmester GR, Nash P, Sands BE, etal. Adverse events of special interest in clinical
trials of rheumatoid arthritis, psoriatic arthritis, ulcerative colitis and psoriasis with 37
066 patient- years of tofacitinib exposure. RMD Open 2021;7:e001595.
11 Pope JE, Keystone E, Jamal S, etal. Persistence of tofacitinib in the treatment of
rheumatoid arthritis in open- label, long- term extension studies up to 9.5 years. ACR
Open Rheumatol 2019;1:73–82.
12 Burmester GR, Gordon KB, Rosenbaum JT, etal. Long- term safety of adalimumab
in 29,967 adult patients from global clinical trials across multiple indications: an
updated analysis. Adv Ther 2020;37:364–80.
13 Curtis JR, Mariette X, Gaujoux- Viala C, etal. Long- term safety of certolizumab
pegol in rheumatoid arthritis, axial spondyloarthritis, psoriatic arthritis, psoriasis and
Crohn’s disease: a pooled analysis of 11 317 patients across clinical trials. RMD Open
2019;5:e000942.
14 Schiff MH, Kremer JM, Jahreis A, etal. Integrated safety in tocilizumab clinical trials.
Arthritis Res Ther 2011;13:R141.
15 Rubbert- Roth A, Sebba A, Brockwell L, etal. Malignancy rates in patients with
rheumatoid arthritis treated with tocilizumab. RMD Open 2016;2:e000213.
16 Weinblatt ME, Moreland LW, Westhovens R, etal. Safety of abatacept administered
intravenously in treatment of rheumatoid arthritis: integrated analyses of up
to 8 years of treatment from the abatacept clinical trial program. J Rheumatol
2013;40:787–97.
17 Emery P, Furst DE, Kirchner P, etal. Risk of malignancies in patients with rheumatoid
arthritis treated with rituximab: analyses of global postmarketing safety data and
long- term clinical trial data. Rheumatol Ther 2020;7:121–31.
18 van Vollenhoven RF, Emery P, Bingham CO 3rd, etal. Long- term safety of rituximab
in rheumatoid arthritis: 9.5- year follow- up of the global clinical trial programme
with a focus on adverse events of interest in RA patients. Ann Rheum Dis
2013;72:1496–502.
19 De Cock D, Hyrich K. Malignancy and rheumatoid arthritis: epidemiology, risk factors
and management. Best Pract Res Clin Rheumatol 2018;32:869–86.
20 Gregson J, Kaptoge S, Bolton T, etal. Cardiovascular risk factors associated with
venous thromboembolism. JAMA Cardiol 2019;4:163–73.
21 Molander V, Bower H, Frisell T, etal. Risk of venous thromboembolism in rheumatoid
arthritis, and its association with disease activity: a nationwide cohort study from
Sweden. Ann Rheum Dis 2021;80:169–75.
22 Omair MA, Alkhelb SA, Ezzat SE, etal. Venous thromboembolism in rheumatoid
arthritis: the added effect of disease activity to traditional risk factors. Open Access
Rheumatol 2022;14:231–42.
23 Sepriano A, Kerschbaumer A, Bergstra SA, etal. Safety of synthetic and biological
dmards: a systematic literature review Informing the 2022 update of the EULAR
recommendations for the management of rheumatoid arthritis. Ann Rheum Dis
2023;82:107–18.
24 Agca R, Heslinga SC, Rollefstad S, etal. EULAR recommendations for
cardiovascular disease risk management in patients with rheumatoid arthritis and
other forms of inflammatory joint disorders: 2015/2016 update. Ann Rheum Dis
2017;76:17–28.
25 Burmester GR, Curtis JR, Yun H, etal. An integrated analysis of the safety of tofacitinib
in psoriatic arthritis across phase III and long- term extension studies with comparison
to real- world observational data. Drug Saf 2020;43:379–92.
26 Mease P, Charles- Schoeman C, Cohen S, etal. Incidence of venous and arterial
thromboembolic events reported in the tofacitinib rheumatoid arthritis, psoriasis and
psoriatic arthritis development programmes and from real- world data. Ann Rheum Dis
2020;79:1400–13.
on July 4, 2023 by guest. Protected by copyright.http://ard.bmj.com/Ann Rheum Dis: first published as 10.1136/ard-2022-223715 on 17 March 2023. Downloaded from
... Analyses of longerterm data showed similar incidence rates of MACE, VTE, and malignancy in patients with PsA and plaque psoriasis treated with JAKis or biologic drugs [110,113,129,130]. The risk of cardiovascular events, but also of malignancy, is increased in patients who have an increased cardiovascular risk at baseline, a history of prior cardiovascular events, diabetes, high blood pressure, and obesity [131][132][133]. More data from long-term real-world studies are needed in order to establish a more a reliable safety profile. ...
... More events were seen with the 30 mg than with the 15 mg dose of upadacitinib, but the 5 and 10 mg twice daily doses of tofacitinib seemed to have similar safety profiles [110,113,129,130]. However, both drugs were generally well tolerated and showed a similar safety profile with biologic or conventional DMARDs already in use in psoriasis and PsA [110,113,[129][130][131][132][133][134]. ...
... Several open-label studies for JAKis are ongoing, and results from these studies will help to better understand the real safety profile of this new class of drugs. Even though in psoriasis and PsA patients treated with JAKis, the risk of major cardiovascular events and malignancy seems similar to the global population with these diseases [131][132][133], data from a 4-year open label study in rheumatoid arthritis showed increased rates of major cardiovascular events and malignancy with tofacitinib compared to TNF-α inhibitors [140]. ...
Article
Full-text available
Psoriasis is a highly prevalent dermatological disease associated with an increased systemic inflammatory response. In addition, joint involvement is also present in around 20% of patients. Therefore, treatment modalities used in this condition should be simultaneously effective at improving skin manifestations, reducing inflammation, and addressing psoriatic arthritis when present. Twenty years ago, the introduction of biologic treatments for psoriasis was a turning point in the management of this condition, offering an effective and reasonably safe option for patients whose disease could not be adeqsuately controlled with conventional therapies. At the moment, Janus Kinase inhibitors (JAKis) are a new class of promising molecules in the management of psoriasis. They are orally administered and can show benefits in patients who failed biologic therapy. We conducted a scoping review in order to identify randomized-controlled trials that investigated different JAKis in patients with plaque psoriasis and psoriatic arthritis, with an emphasis on molecules that have been approved by the European Medicines Agency and the Food and Drug Administration. The added value of this study is that it collected information about JAKis approved for two different indications, plaque psoriasis and psoriatic arthritis, in order to provide an integrated understanding of the range of effects that JAKis have on the whole spectrum of psoriasis manifestations.
... 44 Post hoc analysis of the ORAL Surveillance study indicated that the increased risk of MACE with tofacitinib was mainly observed in the sub-populations with a history of atherosclerotic CVD 45 and those who were aged ≥65 years or who smoked. 46 On the other hand, data from STAR-RA, 47 French, 48 and Hong Kong biologics (tofacitinib) 33 registries did not indicate increased frequency of MACE when compared with TNFi. 43 On the subject of VTEs, results are also conflicting. ...
... In ORAL Surveillance, the risk of DVT and PE were comparable between tofacitinib 5 mg recipients and TNFi-treated individuals, but VTE risk (mostly PE) was increased for the 10 mg BD tofacitinib dose, 44 again mainly in those aged ≥65 years or who smoked. 46 Data from the Swedish Registry reported a hazard ratio of 1.73 (1.24 to 2.42) for VTE in JAKi-treated RA patients compared to anti-TNFs, 49 whereas real world data also suggested that there is an augmented risk for VTEs in RA patients treated with baricitinib [IR ratio compared to TNFi: 1.51 (95% CI 1.10, 2.08)]. 49 Similarly, to MACE, there were also studies reporting comparable risk for VTEs between JAKi and bDMARDS, such as those deriving from French and Corona registries 43 and a population-based study in Korean patients with RA treated with JAKi versus TNFi. ...
Article
Full-text available
As our research interest and knowledge increases in the field of Spondyloarthritis, new aspects also emerge as regards to their therapeutic approach. JAK inhibitors (JAKi) are a relatively new treatment option, aiming molecules in the JAK-STAT pathway, which has a leading role in the pathophysiology of both Psoriatic Arthritis and Axial Spondyloarthritis. JAKi exhibit different selectivity towards the four different members of the JAK family (namely JAK1, JAK2, JAK3, and TYK2), possibly reflecting different efficacy and safety profile. Although knowledge is more consolidated for rheumatoid arthritis in which JAKi are being used for more than 10 years, data are still accumulating for PsA/SpA. In this review we aim to present and assess current knowledge about the efficacy of JAKi (with a focus on selective JAKi) in the treatment of patients with SpA and evaluate their safety profile as some concerns may arise around this therapeutic option.
... In ORAL Surveillance, cardiovascular risk-enriched adult patients with rheumatoid arthritis demonstrated a higher rate of MACE and cancers with tofacitinib, compared with tumour necrosis factor inhibitors 43 ; risk differences of these outcomes were confined to patients who were ≥65 years of age, and/or long-time current/past smokers, 44 and those who had a history of atherosclerotic cardiovascular disease (MACE only). 45 While the current LTE study did not observe any cases of MACE or malignancies in a population of patients with JIA, and indeed, these differentiating risk factors are less applicable to the JIA population, the safety findings of ORAL Surveillance warrant a precautionary approach to apply these findings across all JAK inhibitors and all approved disease states, until data from additional dedicated safety studies (of sufficient size and duration) establish that this is not appropriate. ...
Article
Full-text available
Objectives We report the safety, tolerability and efficacy of tofacitinib in patients with juvenile idiopathic arthritis (JIA) in an ongoing long-term extension (LTE) study. Methods Patients (2–<18 years) with JIA who completed phase 1/3 index studies or discontinued for reasons excluding treatment-related serious adverse events (AEs) entered the LTE study and received tofacitinib 5 mg two times per day or equivalent weight-based doses. Safety outcomes included AEs, serious AEs and AEs of special interest. Efficacy outcomes included improvement since tofacitinib initiation per the JIA-American College of Rheumatology (ACR)70/90 criteria, JIA flare rate and disease activity measured by Juvenile Arthritis Disease Activity Score (JADAS)27, with inactive disease corresponding to JADAS ≤1.0. Results Of 225 patients with JIA (median (range) duration of treatment, 41.6 (1–103) months), 201 (89.3%) had AEs; 34 (15.1%) had serious AEs. 10 patients developed serious infections; three had herpes zoster. Two patients newly developed uveitis. Among patients with polyarticular course JIA, JIA-ACR70/90 response rates were 60.0% (78 of 130) and 33.6% (47 of 140), respectively, at month 1, and generally improved over time. JIA flare events generally occurred in <5% of patients through to month 48. Observed mean (SE) JADAS27 was 22.0 (0.6) at baseline, 6.2 (0.7) at month 1 and 2.8 (0.5) at month 48, with inactive disease in 28.8% (36 of 125) of patients at month 1 and 46.8% (29 of 82) at month 48. Conclusions In this interim analysis of LTE study data in patients with JIA, safety findings were consistent with the known profile of tofacitinib, and efficacy was maintained up to month 48. Trial registration number NCT01500551 .
... The findings that women and young adults may warrant closer monitoring for tofacitinib-associated dyslipidemia could inform more tailored RA treatment approaches. No differences in lipid response to tofacitinib versus adalimumab were observed by race, contrasting with some cardiovascular studies showing higher event risks in minorities (Khosrow-Khavar et al., 2022;Kristensen et al., 2023a;Kristensen et al., 2023b;Schreiber et al., 2023). Kaplan-Meier curve of lipidemia disorders incidence. ...
Article
Full-text available
Objective To compare the effects of tofacitinib and adalimumab on the risk of adverse lipidaemia outcomes in patients with newly diagnosed rheumatoid arthritis (RA). Methods Data of adult patients newly diagnosed with RA who were treated with tofacitinib or adalimumab at least twice during a 3-year period from 1 January 2018 to 31 December 2020, were enrolled in the TriNetX US Collaborative Network. Patient demographics, comorbidities, medications, and laboratory data were matched by propensity score at baseline. Outcome measurements include incidental risk of dyslipidemia, major adverse cardiac events (MACE) and all-cause mortality. Results A total of 7,580 newly diagnosed patients with RA (1998 receiving tofacitinib, 5,582 receiving adalimumab) were screened. After propensity score matching, the risk of dyslipidaemia outcomes were higher in the tofacitinib cohort, compared with adalimumab cohort (hazard ratio [HR] with 95% confidence interval [CI], 1.250 [1.076–1.453]). However, there is no statistically significant differences between two cohorts on MACE (HR, 0.995 [0.760–1.303]) and all-cause mortality (HR, 1.402 [0.887–2.215]). Conclusion Tofacitinib use in patients with RA may increase the risk of dyslipidaemia to some extent compared to adalimumab. However, there is no differences on MACE and all-cause mortality.
... 22 Notably, a recent post hoc analysis of ORAL Surveillance suggested that the higher risk of VTE with tofacitinib versus TNFi was restricted to patients ≥65 years of age and/or with a history of long-term current/past smoking. 23 Future studies will provide more clarity on specific patient characteristics associated with increased risk of MI and VTE with tofacitinib vs TNFi. ...
Article
Full-text available
Objectives Compare the risk of extended major adverse cardiovascular (CV) event (MACE) composite outcomes and component events in patients with rheumatoid arthritis (RA) treated with tofacitinib versus tumour necrosis factor inhibitors (TNFi) in Oral Rheumatoid Arthritis Trial (ORAL) Surveillance. Methods Patients with RA aged ≥50 years and with ≥1 additional CV risk factor received tofacitinib 5 mg or 10 mg two times per day or TNFi. MACE (non-fatal myocardial infarction (MI), non-fatal stroke or CV death (MACE-3)) was extended by sequential addition of CV events (hospitalisation for unstable angina (MACE-4), coronary revascularisation (MACE-5), transient ischaemic attack (MACE-6), peripheral vascular disease (MACE-7)), heart failure (HF) hospitalisation (MACE-8) and venous thromboembolism (VTE; (MACE-8 plus VTE)). HRs (tofacitinib vs TNFi) were evaluated for MACE and individual components. Results HRs for MACE-4 to MACE-8 with combined and individual tofacitinib doses versus TNFi were similar. Risk of MACE-8 plus VTE appeared similar with tofacitinib 5 mg two times per day versus TNFi (HR 1.12 (0.82 to 1.52)), but higher with tofacitinib 10 mg two times per day versus TNFi (HR 1.38 (1.02 to 1.85)). Risk of MI was higher with tofacitinib versus TNFi, but difference in risk of other individual CV events was not suggested. Across extended MACE definitions, risk appeared higher with tofacitinib versus TNFi in those with atherosclerotic CV disease or age ≥65 years. Conclusion In ORAL Surveillance, risk of composite CV endpoints combining all ischaemic CV events and HF did not appear different with tofacitinib versus TNFi. The totality of CV risk was higher with tofacitinib 10 mg two times per day versus TNFi, driven by an increase in VTE. Trial registration number NCT02092467.
Article
Objective: Janus kinase inhibitors (JAKi) or targeted synthetic (ts) disease-modifying antirheumatic drugs (DMARDs) effectively treat rheumatoid arthritis (RA). However, due to safety concerns, the European Medicines Agency (EMA) published risk minimisation measures limiting JAKi prescription to certain at-risk patients unless no suitable alternative is available. This analysis included patients who had started their first-ever JAKi before EMA measures were published in a large national cohort study to investigate the potential impact of these measures on JAKi prescribing and utilisation in UK. Method: RA patients starting first-ever JAK inhibitor therapy in BSRBR-RA between 13-February-2017 and 31-May-2022 were included. Percentages of patients meeting EMA risk criteria were presented. For at-risk patients, previous number of distinct biological (b) DMARD classes were described. Result: A total of 1341 patients were included, and 80% (N = 1075) met ≥1 EMA risk criterion. Of those who met ≥1 risk criterion, 529 patients (49%) had received JAKi as their first or second b/tsDMARD class, whereas 299 (28%) had received ≥3 prior bDMARD classes. Conclusion: Four-in-five RA patients commencing JAKi before the EMA advisory were considered 'at-risk' with prescribing only advised if there was no suitable alternative. Almost a third of those patients had already received ≥3 bDMARDs classes, and alternative therapies would be very limited for them; meanwhile, suitable alternatives might have existed for the remaining proportion, especially for those who received JAKi as their first or second b/tsDMARD, and re-evaluation of the suitability of their treatment may be needed.
Article
Full-text available
In this commentary, we review clinical data which helps inform individualized benefit–risk assessment for tofacitinib in patients with psoriatic arthritis (PsA) and ankylosing spondylitis (AS). ORAL Surveillance, a safety trial of patients ≥ 50 years of age with rheumatoid arthritis (RA) and cardiovascular risk factors, found increased rates of safety outcomes (including major adverse cardiovascular events [MACE], malignancies excluding non-melanoma skin cancer, and venous thromboembolism) with tofacitinib versus tumor necrosis factor inhibitors (TNFi). Post hoc analyses of ORAL Surveillance have identified subpopulations with different relative risk versus TNFi; higher risk with tofacitinib was confined to patients ≥ 65 years of age and/or long-time current/past smokers, and specifically for MACE, patients with a history of atherosclerotic cardiovascular disease (ASCVD). In patients without these risk factors, risk differences between tofacitinib and TNFi could not be detected. Given differences in demographics, pathophysiology, and comorbidities, we sought to examine whether the risk stratification observed in RA is also appropriate for PsA and AS. Data from the PsA tofacitinib development program show low absolute risk of safety outcomes in patients < 65 years of age and never smokers, and low MACE risk in patients with no history of ASCVD, consistent with results from ORAL Surveillance. No MACE, malignancies, or venous thromboembolism were reported in the tofacitinib AS development program. The mechanism of the ORAL Surveillance safety findings is unknown, and there are no similar prospective studies of sufficient size and duration. Accordingly, it is appropriate to use a precautionary approach and extrapolate differentiating risk factors identified from ORAL Surveillance (age ≥ 65 years, long-time current/past smoking, and history of ASCVD) to PsA and AS. We recommend an individualized approach to treatment decisions based on these readily identifiable risk factors, in line with updated labeling for Janus kinase inhibitors and international guidelines for the treatment of PsA and AS. Trial Registration: NCT02092467, NCT01262118, NCT01484561, NCT00147498, NCT00413660, NCT00550446, NCT00603512, NCT00687193, NCT01164579, NCT00976599, NCT01059864, NCT01359150, NCT02147587, NCT00960440, NCT00847613, NCT00814307, NCT00856544, NCT00853385, NCT01039688, NCT02281552, NCT02187055, NCT02831855, NCT00413699, NCT00661661, NCT01877668, NCT01882439, NCT01976364, NCT00678210, NCT01710046, NCT01241591, NCT01186744, NCT01276639, NCT01309737, NCT01163253, NCT01786668, NCT03502616.
Article
Full-text available
Janus Kinases (JAKs) play a crucial role as therapeutic targets for various cancers. However, the current JAK inhibitors (JAKi) available have limited therapeutic benefits due to their lack of selectivity. This review focuses on the structural analysis to elucidate the molecular determinants of JAKs specificity and the discovery and design of selective JAKi. It includes descriptions and comparison of different JAK structures and their binding sites, a comparative analysis of JAKi and their binding modes, detailed interaction fingerprints (IFPs), and an extensive structure‐selectivity relationship (SSRs). Moreover, the review also explores the challenges and possibilities of using computational structure‐based methods for discovering and designing selective JAKi. Other structure‐based approaches, such as targeting the pseudokinase domain, as well as covalent and allosteric designs, are also covered. Based on this analysis, key determinants corresponding to JAK specificity and rational medicinal chemistry strategies are proposed to facilitate the development of highly selective JAKi. Overall, we aim to enhance the understanding of JAK specificity and explore strategies that can lead to the discovery of effective and selective JAKi in cancer therapy, thus improving the prognosis for cancer patients.
Article
Full-text available
Objectives To evaluate malignancies and their associations with baseline risk factors and cardiovascular risk scores with tofacitinib versus tumour necrosis factor inhibitors (TNFi) in patients with rheumatoid arthritis (RA). Methods In an open-label, randomised controlled trial (ORAL Surveillance; NCT02092467 ), 4362 patients with RA aged ≥50 years with ≥1 additional cardiovascular risk factor received tofacitinib 5 (N=1455) or 10 mg two times per day (N=1456) or TNFi (N=1451). Incidence rates (IRs; patients with first events/100 patient-years) and HRs were calculated for adjudicated malignancies excluding non-melanoma skin cancer (NMSC), NMSC and subtypes. Post hoc analyses for malignancies excluding NMSC, lung cancer and NMSC included risk factors identified via simple/multivariable Cox models and IRs/HRs categorised by baseline risk factors, history of atherosclerotic cardiovascular disease (HxASCVD) and cardiovascular risk scores. Results IRs for malignancies excluding NMSC and NMSC were higher with tofacitinib (combined and individual doses) versus TNFi. Risk of lung cancer (most common subtype with tofacitinib) was higher with tofacitinib 10 mg two times per day versus TNFi. In the overall study population, the risk of malignancies excluding NMSC was similar between both tofacitinib doses and TNFi until month 18 and diverged from month 18 onwards (HR (95% CIs) for combined tofacitinib doses: 0.93 (0.53 to 1.62) from baseline to month 18 vs 1.93 (1.22 to 3.06) from month 18 onwards, interaction p=0.0469). Cox analyses identified baseline risk factors across treatment groups for malignancies excluding NMSC, lung cancer and NMSC; interaction analyses generally did not show statistical evidence of interaction between treatment groups and risk factors. HxASCVD or increasing cardiovascular risk scores were associated with higher malignancy IRs across treatments. Conclusions Risk of malignancies was increased with tofacitinib versus TNFi, and incidence was highest in patients with HxASCVD or increasing cardiovascular risk. This may be due to shared risk factors for cardiovascular risk and cancer. Trial registration numbers NCT02092467 , NCT01262118 , NCT01484561 , NCT00147498 , NCT00413660 , NCT00550446 , NCT00603512 , NCT00687193 , NCT01164579 , NCT00976599 , NCT01059864 , NCT01359150 , NCT02147587 , NCT00960440 , NCT00847613 , NCT00814307 , NCT00856544 , NCT00853385 , NCT01039688 , NCT02281552 , NCT02187055 , NCT02831855 , NCT00413699 , NCT00661661
Article
Full-text available
Many epidemiological studies have shown an increased risk of venous thromboembolism (VTE) in patients with rheumatoid arthritis (RA). RA and VTE share some background factors, such as increasing age, smoking, and obesity. At the same time, other VTE factors, such as knee replacement and oral contraceptive pills, occur commonly in RA patients. In addition, the chronic inflammatory state of RA might hypothetically lead to endothelial injury and a hypercoagulable state. Two critical pathophysiological pathways lead to VTE. Recently, concerns increased about the increased risk of VTE in patients using Janus Kinase inhibitors. This review aims at reviewing the risk of VTE in RA and the role of traditional risk factors and disease-related inflammation and develops a conceptual framework that describes the interaction between these factors.
Article
Full-text available
Objectives Evaluate risk of major adverse cardiovascular events (MACE) with tofacitinib versus tumour necrosis factor inhibitors (TNFi) in patients with rheumatoid arthritis (RA) with or without a history of atherosclerotic cardiovascular disease (ASCVD) in ORAL Surveillance. Methods Patients with RA aged ≥50 years with ≥1 additional CV risk factor received tofacitinib 5 mg or 10 mg two times per day or TNFi. Hazard rations (HRs) were evaluated for the overall population and by history of ASCVD (exploratory analysis). Results Risk of MACE, myocardial infarction and sudden cardiac death were increased with tofacitinib versus TNFi in ORAL Surveillance. In patients with history of ASCVD (14.7%; 640/4362), MACE incidence was higher with tofacitinib 5 mg two times per day (8.3%; 17/204) and 10 mg two times per day (7.7%; 17/222) versus TNFi (4.2%; 9/214). HR (combined tofacitinib doses vs TNFi) was 1.98 (95% confidence interval (CI) 0.95 to 4.14; interaction p values: 0.196 (for HR)/0.059 (for incidence rate difference)). In patients without history of ASCVD, MACE HRs for tofacitinib 5 mg two times per day (2.4%; 30/1251) and 10 mg two times per day (2.8%; 34/1234) versus TNFi (2.3%; 28/1237) were, respectively, 1.03 (0.62 to 1.73) and 1.25 (0.76 to 2.07). Conclusions This post hoc analysis observed higher MACE risk with tofacitinib versus TNFi in patients with RA and history of ASCVD. Among patients without history of ASCVD, all with prevalent CV risk factors, MACE risk did not appear different with tofacitinib 5 mg two times per day versus TNFi. Due to the exploratory nature of this analysis and low statistical power, we cannot exclude differential MACE risk for tofacitinib 5 mg two times per day versus TNFi among patients without history of ASCVD, but any absolute risk excess is likely low. Trial registration number NCT02092467 .
Article
Full-text available
Objectives To analyse adverse events (AEs) of special interest across tofacitinib clinical programmes in rheumatoid arthritis (RA), psoriatic arthritis (PsA), ulcerative colitis (UC) and psoriasis (PsO), and to determine whether the incidence rates (IRs; unique patients with events per 100 patient-years) of these events are consistent across diseases. Methods The analysis included data from patients exposed to ≥1 dose of tofacitinib in phase 1, 2, 3 or 3b/4 clinical trials and long-term extension (LTE) studies (38 trials) in RA (23 trials), PsA (3 trials), UC (5 trials) and PsO (7 trials). All studies were completed by or before July 2019, except for one ongoing UC LTE study (data cut-off May 2019). IRs were obtained for AEs of special interest. Results 13 567 patients were included in the analysis (RA: n=7964; PsA: n=783; UC: n=1157; PsO: n=3663), representing 37 066 patient-years of exposure. Maximum duration of exposure was 10.5 years (RA). AEs within the ‘infections and infestations’ System Organ Class were the most common in all diseases. Among AEs of special interest, IRs were highest for herpes zoster (non-serious and serious; 3.6, 1.8, 3.5 and 2.4 for RA, PsA, UC and PsO, respectively) and serious infections (2.5, 1.2, 1.7 and 1.3 for RA, PsA, UC and PsO, respectively). Age-adjusted and sex-adjusted mortality ratios (weighted for country) were ≤0.2 across cohorts. Conclusions The tofacitinib safety profile in this analysis was generally consistent across diseases and with longer term follow-up compared with previous analyses.
Article
Full-text available
Background The extent to which co-occurrence of cardiovascular disease (CVD) and cancer is due to shared risk factors or other mechanisms is unknown. Objectives This study investigated the association of standard CVD risk factors, CVD biomarkers, pre-existing CVD, and ideal cardiovascular (CV) health metrics with the development of future cancer. Methods This study prospectively followed Framingham Heart Study and PREVEND (Prevention of Renal and Vascular End-Stage Disease) study participants free of cancer at baseline and ascertained histology-proven cancer. This study assessed the association of baseline CV risk factors, 10-year atherosclerotic (ASCVD) risk score, established CVD biomarkers, prevalent CVD, and the American Heart Association (AHA) Life’s Simple 7 CV health score with incident cancer using multivariable Cox models. Analyses of interim CVD events with incident cancer used time-dependent covariates. Results Among 20,305 participants (mean age 50 ± 14 years; 54% women), 2,548 incident cancer cases occurred over a median follow-up of 15.0 years (quartile 1 to 3: 13.3 to 15.0 years). Traditional CVD risk factors, including age, sex, and smoking status, were independently associated with cancer (p < 0.001 for all). Estimated 10-year ASCVD risk was also associated with future cancer (hazard ratio [HR]: 1.16 per 5% increase in risk; 95% confidence interval [CI] 1.14 to 1.17; p < 0.001). The study found that natriuretic peptides (tertile 3 vs. tertile 1; HR: 1.40; 95% CI: 1.03 to 1.91; p = 0.035) were associated with incident cancer but not high-sensitivity troponin (p = 0.47). Prevalent CVD and the development of interim CV events were not associated with higher risk of subsequent cancer. However, ideal CV health was associated with lower future cancer risk (HR: 0.95 per 1-point increase in the AHA health score; 95% CI: 0.92 to 0.99; p = 0.009). Conclusions CVD risk, as captured by traditional CVD risk factors, 10-year ASCVD risk score, and natriuretic peptide concentrations are associated with increased risk of future cancer. Conversely, a heart healthy lifestyle is associated with a lower risk of future cancer. These data suggest that the association between CVD and future cancer is attributable to shared risk factors.
Article
Full-text available
Objectives Tofacitinib is a Janus kinase inhibitor for the treatment of rheumatoid arthritis (RA), psoriatic arthritis (PsA) and ulcerative colitis, and has been investigated in psoriasis (PsO). Routine pharmacovigilance of an ongoing, open-label, blinded-endpoint, tofacitinib RA trial (Study A3921133; NCT02092467 ) in patients aged ≥50 years and with ≥1 cardiovascular risk factor identified a higher frequency of pulmonary embolism (PE) and all-cause mortality for patients receiving tofacitinib 10 mg twice daily versus those receiving tumour necrosis factor inhibitors and resulted in identification of a safety signal for tofacitinib. Here, we report the incidence of deep vein thrombosis (DVT), PE, venous thromboembolism (VTE; DVT or PE) and arterial thromboembolism (ATE) from the tofacitinib RA (excluding Study A3921133), PsA and PsO development programmes and observational studies. Data from an ad hoc safety analysis of Study A3921133 are reported separately within. Methods This post-hoc analysis used data from separate tofacitinib RA, PsO and PsA programmes. Incidence rates (IRs; patients with events per 100 patient-years’ exposure) were calculated for DVT, PE, VTE and ATE, including for populations stratified by defined baseline cardiovascular or VTE risk factors. Observational data from the US Corrona registries (including cardiovascular risk factor stratification), IBM MarketScan research database and the US FDA Adverse Event Reporting System (FAERS) database were analysed. Results 12 410 tofacitinib-treated patients from the development programmes (RA: n=7964; PsO: n=3663; PsA: n=783) were included. IRs (95% CI) of thromboembolic events among the all tofacitinib cohorts’ average tofacitinib 5 mg and 10 mg twice daily treated patients for RA, respectively, were: DVT (0.17 (0.09–0.27) and 0.15 (0.09–0.22)); PE (0.12 (0.06–0.22) and 0.13 (0.08–0.21)); ATE (0.32 (0.22–0.46) and 0.38 (0.28–0.49)). Among PsO patients, IRs were: DVT (0.06 (0.00–0.36) and 0.06 (0.02–0.15)); PE (0.13 (0.02–0.47) and 0.09 (0.04–0.19)); ATE (0.52 (0.22–1.02) and 0.22 (0.13–0.35)). Among PsA patients, IRs were: DVT (0.00 (0.00–0.28) and 0.13 (0.00–0.70)); PE (0.08 (0.00–0.43) and 0.00 (0.00–0.46)); ATE (0.31 (0.08–0.79) and 0.38 (0.08–1.11)). IRs were similar between tofacitinib doses and generally higher in patients with baseline cardiovascular or VTE risk factors. IRs from the overall Corrona populations and in Corrona RA patients (including tofacitinib-naïve/biologic disease-modifying antirheumatic drug-treated and tofacitinib-treated) with baseline cardiovascular risk factors were similar to IRs observed among the corresponding patients in the tofacitinib development programme. No signals of disproportionate reporting of DVT, PE or ATE with tofacitinib were identified in the FAERS database. Conclusions DVT, PE and ATE IRs in the tofacitinib RA, PsO and PsA programmes were similar across tofacitinib doses, and generally consistent with observational data and published IRs of other treatments. As expected, IRs of thromboembolic events were elevated in patients with versus without baseline cardiovascular or VTE risk factors, and were broadly consistent with those observed in the Study A3921133 ad hoc safety analysis data, although the IR (95% CI) for PE was greater in patients treated with tofacitinib 10 mg twice daily in Study A3921133 (0.54 (0.32–0.87)), versus patients with baseline cardiovascular risk factors treated with tofacitinib 10 mg twice daily in the RA programme (0.24 (0.13–0.41)).
Article
Objectives: To perform a systematic literature review (SLR) concerning the safety of synthetic(s) and biological (b) disease-modifying antirheumatic drugs (DMARDs) to inform the 2022 update of the EULAR recommendations for the management of rheumatoid arthritis (RA). Methods: SLR of observational studies comparing safety outcomes of any DMARD with another intervention in RA. A comparator group was required for inclusion. For treatments yet without, or limited, registry data, randomised controlled trials (RCTs) were used. Results: Fifty-nine observational studies addressed the safety of DMARDs. Two studies (unclear risk of bias (RoB)) showed an increased risk of serious infections with bDMARDs compared with conventional synthetic (cs)DMARDs. Herpes zoster infections occurred more with JAKi than csDMARDs (adjusted HR (aHR): 3.66) and bDMARDs (aHR: 1.9-2.3) (four studies, two low RoB). The risk of malignancies was similar across bDMARDs (five studies) and with tofacitinib compared with bDMARDs (one study, low RoB). The risk of major adverse cardiovascular events (MACE) was similar with bDMARDs and tofacitinib (two studies, one low RoB). Thirty studies reported safety from RCTs, with one, designed to evaluate safety, showing that malignancies (HR (95% CI): 1.48 (1.04 to 2.09)) and MACE (HR (95% CI): 1.33 (0.91 to 1.94)) occurred numerically more frequently with tofacitinib (5 mg and 10 mg doses combined) than with TNFi in patients with cardiovascular risk factors. In this study, the risk of venous thromboembolism (VTE) was higher with tofacitinib 10 mg than with TNFi. Conclusion: The safety profile of bDMARDs was further demonstrated. Whether the difference in incidence of malignancies, MACE and VTE between tofacitinib and TNFi applies to other JAKi needs further evaluation.
Article
Objective To assess the incidence of venous thromboembolism (VTE) in rheumatoid arthritis (RA) relative to individuals without RA, and to investigate the relationship between aspects of clinical disease activity in RA and the risk of VTE. Methods We conducted a nationwide register-based cohort study 2006 through 2018 using the Swedish Rheumatology Quality Register linked to other national patient registers to identify all patients with RA with at least one registered rheumatologist visit during the study period (n=46 316 patients, 322 601 visits). The Disease Activity Score 28 erythrocyte sedimentation rate (ESR) (DAS28 ESR) and its components served as the exposure, and a VTE event within the year following the visit was the main outcome. We also included general population referents (1:5) matched on age, sex and residential area. Results Based on 2241 incident VTE events within 1 year of each included visit, and 5301 VTE events in the general population cohort, the risk ratio for VTE in RA was 1.88 (95% CI 1.65 to 2.15). Among patients with RA, the risk (and risk ratio) increased with increasing RA disease activity, from 0.52% following visits in remission to 1.08% following visits with DAS28 ESR high disease activity, RR compared with remission=2.03, 95% CI 1.73 to 2.38. Compared with the general population, also patients with RA in DAS28 ESR remission were at elevated VTE risk. Conclusions This study demonstrates a strong association between clinical RA disease activity measured by DAS28 ESR and the risk of VTE. RA disease activity can be used as an additional tool for VTE risk stratification in patients with RA.