Skip to main content
BMJ Open logoLink to BMJ Open
. 2022 Sep 21;12(9):e059979. doi: 10.1136/bmjopen-2021-059979

Oral Janus kinase inhibitors and venous thromboembolic events in atopic dermatitis: protocols for a case–time control study and a nested case-control study based on the French national health insurance (SNDS) cohort

Pauline Berthe 1,, Lucie-Marie Scailteux 2,3, Alain Lescoat 3,4, Delphine Staumont 5, Guillaume Coiffier 6,7, Pierre Guéret 8, Alain Dupuy 1,3, Emmanuel Oger 2,3, Catherine Droitcourt 1,3
PMCID: PMC9494565  PMID: 36130766

Abstract

Introduction

Atopic dermatitis (AD) is a highly prevalent, chronic, inflammatory skin disease. Several orally administered Janus kinase inhibitors (JAKis, including baricitinib, upadacitinib and abrocitinib) have received a marketing authorisation for AD.

Clinical trials in rheumatoid arthritis (RA) have flagged up a potential risk of JAKi-induced venous thromboembolic events (VTEs). Accordingly, the summary of product characteristics for a JAKi must mention VTEs as potential adverse drug reactions. In contrast to RA, AD per se is not associated with an elevated risk of VTEs. Assessing this potential risk among patients with AD would shed further light on the putative underlying relationship between JAKis and VTEs.

Our research question is to investigate whether JAKi administration increases the risk of VTEs in adults with AD. Our primary objective is to assess the risk of VTEs in adults with AD exposed to JAKis compared to AD adults not exposed to JAKis, and our secondary objective is to evaluate whether JAKi initiation acts as a trigger of VTEs in adults with AD within 3 months.

Methods and analysis

Hence, we have designed (1) a nested case–control study and (2) a case–time control study in a cohort of adults with AD with data from the French national health insurance system (2017–2025).

Here, we describe the study protocol, our methodological choices and certain novel aspects, including the combined value of the two assumptions and the use of an exhaustive national health insurance database with potentially greater statistical power for studying rare events in the population of patients with AD at a low risk of VTEs (thus limiting the influence of confounding factors).

Ethics and dissemination

The protocol has been approved by an independent ethics committee and registered with the French National Data Protection Commission. The study’s findings will be published in peer-reviewed scientific journals and presented at international conferences.

Keywords: Adult dermatology, Eczema, Thromboembolism, Risk management, CLINICAL PHARMACOLOGY


Strengths and limitations of this study.

  • A population-based study using the exhaustive French national health insurance database would provide additional insight into the risk of venous thromboembolic events (VTEs). Advantageously, this nationwide study should be able to exhaustively identify VTEs, the time of their occurrence, and prescriptions of JAK inhibitors.

  • By studying atopic dermatitis (AD), we hope to avoid a major source of confounding bias; in contrast to rheumatoid arthritis, AD is not associated per se with an elevated risk of VTEs.

  • The limitations of this study protocol (based on the use of French national health insurance database) include a lack of data on certain risk factors for VTEs (including obesity and a family history of thromboembolic disease)

  • A potential lack of statistical power.

Introduction

Atopic dermatitis (AD) is a highly prevalent, pruritic, inflammatory disease skin that occurs in both adults (3%–10%)1–3 and children (15%–20%).1 4 5 Approximately 2%–8% of adults with AD have severe forms; the associated impairments in quality of life make AD a disabling disease. Severe AD is frequently associated with other atopic comorbidities (eg, asthma, allergic rhinitis, allergic conjunctivitis and food allergy) and may be associated with psychiatric disorders.

The European guidelines on the management of AD in adults recommend first-line treatment with topical anti-inflammatory drugs (topical corticosteroids and tacrolimus) and then (if the treatment fails) systemic immunosuppressants.6 7 In late 2017, the management of treatment-refractory AD was revolutionised by the marketing of the first biological drug, dupilumab (a subcutaneously administered monoclonal antibody against interleukin (IL)-4 and IL-13 receptors).8 9 Other systemic treatments have since received (or are awaiting) marketing authorisation: baricitinib (an orally administered Janus kinase (Jak) 1 and 2 inhibitor (Janus kinase inhibitor (JAKi)),10–13 upadacitinib (an orally administered JAK1 inhibitor),14–16 abrocitinib (another orally administered JAK1 inhibitor)17–19 and tralokinumab (a subcutaneously administered anti-IL-13 monoclonal antibody).20 21

JAKis constitute a new family of orally administered molecules that target the JAK signal transducer and activator of transcription (STAT) pathway. Jaks are involved in the transduction of intracellular signals in response to various cytokines and growth factors involved in haematopoiesis, inflammation and immune functions.

In the European Union, baricitinib was approved for the treatment of active, moderate-to-severe rheumatoid arthritis (RA) in adults in 2017 and for moderate-to-severe AD in adults who are candidates for systemic drug treatment in 2021. Upadacitinib was approved for the treatment of adults with moderate-to-severe active RA, psoriatic arthritis (PsA) or ankylosing spondylitis (AS) in 2020 and 2021 and for the treatment of moderate-to-severe AD in adults and adolescents (aged 12 or over) who are candidates for systemic drug treatment in August 2021. Lastly, abrocitinib was approved very recently by the European Medicines Agency (EMA) for the systemic treatment of moderate-to-severe AD in adults and adolescents.

Clinical trials in RA have flagged up a potential risk of JAKi-induced venous thromboembolic events (VTEs, including deep vein thrombosis and pulmonary embolism).22–26 Although the EMA approved low (2 mg) and high (4 mg) doses of baricitinib, the Food and Drug Administration approved only the 2 mg dose because of the VTE risk. On a broader scale, the summary of product characteristics for a JAKi must mention VTEs as potential adverse drug reactions. The safety profiles of baricitinib and upadacitinib in patients with RA have been described in nine and five clinical studies, respectively. The estimated incidence of VTEs ranged from 0.3 to 0.6 per 100 person-years.22 27

Due to the presence of systemic inflammation, RA per se can induce thromboembolic events, and the treatment of RA with anti-inflammatory drugs helps to reduce the cardiovascular and thromboembolic risks.25 28 Furthermore, most patients with RA are aged over 50 at diagnosis and have higher prevalence of obesity and a higher incidence of VTEs. In this case, the interplay between RA, JAKis and thromboembolic risk is particularly difficult to characterise.

The pathogenic links between JAKis and a potentially greater risk of thromboembolic disease are poorly understood, and the literature data are contradictory. The potential thromboembolic risk might be related to an imbalance between prothrombotic and antithrombotic signals, including the inhibition of proinflammatory signals (such as interferon-dependant pathways) and the paradoxical inhibition of JAK–STAT-dependent anti-inflammatory pathways (such as the IL-10 pathway that helps to limit clot formation under normal conditions).29 30 JAKis that influence JAK2-dependent signalling (such as baricitinib) might also promote platelet formation from megakaryocytes, as evidenced by a transient increase in the platelet count following JAKi initiation. Nonetheless, a causal link between transient thrombocytosis and VTE has never been proven.22

The results of meta-analyses of the links between JAKis and the risk of thromboembolic and/or cardiovascular events are summarised in table 1.31–37

Table 1.

List of meta-analyses on the risk of VTEs during treatment with JAKis

First
author
Date of publication JAK inhibitor Indication Studies included (n) Type of studies included Patients included (n) Median follow-up (weeks) Events among exposed participants (n) Events among non-exposed participants (n) Results
OR (95% CI)
Methods used
Xie et al31 2019 Tofacitinib, baricitinib, upadacitinib, peficitinib, decernotinib RA 26 RCT 11 799 Placebo-controlled period: 12
Dose-comparison period: 24
12 3 All JAKis: 1.16 (0.48 to 2.81)
Tofacitinib: 0.17 (0.03 to 1.05)
Baricitinib: 2.33 (0.62 to 8.75)
Upadacitinib: 1.77 (0.20 to 16.00)
Mantel-Haenszel fixed-effect method
Xie et al32 2019 Tofacitinib RA, PsA, CPP, UC, CD, AS 27 RCT 13 611 Placebo-controlled period: 12
Dose-comparison period: 24
1 5 0.03 (0.00 to 0.21) Peto method
Olivera et al33 2020 Tofacitinib, upadacitinib, filgotinib, baricitinib RA, AS, UC, CD, CPP 10 RCT
Cohorts
5143 26 12 3 All JAKis: 0.90 (0.32 to 2.54) Random-effect model
Giménez Poderós et al34 2020 Tofacitinib, baricitinib RA, KT, UC, CPP, CD, PsA, AD, DKD, SLE, JIA, SS 59 RCT
Cohorts
25 947 16 24 23 Tofacitinib: 0.29 (0.10 to 0.84)
Baricitinib: 3.39 (0.82 to 14.04)
Fixed-effect or random-effect model, with application of the most conservative model in each case
Yates et al35 2020 Tofacitinib, baricitinib, upadacitinib, filgotinib RA, PsA, AS, UC, CD, CPP 42 RCT 17 269 Unavailable 15 4 All JAKis: 0.68 (0.36 to 1.29) Mantel-Haenszel fixed-effect method
Wang et al36 2020 Upadacitinib RA 3 RCT 2852 Unavailable 3 1 2.34 (0.15 to 15.02) Random-effect model
Bilal et al37 2021 Abrocitinib, baricitinib, decernotinib, filgotinib, peficitinib, ruxolitinib, tofacitinib RA, AD, SLE, CPP, AS, PsA, UC, pancreatic cancer, breast cancer 29 RCT 13 910 48 50 27 All JAKis: 0.91 (0.57 to 1.47)
Baricitinib: 1.12 (0.27 to 4.69)
Decernotinib: 1.07 (0.18 to 6.43)
Filgotinib: 2.13 (0.22 to 20.64)
Ruxolitinib: (0.31 to 2.29)
Upadacitinib: 2.25 (0.55 to 9.25)
Tofacitinib: 0.27 (0.08 to 0.89)
Random-effects model

AD, atopic dermatitis; AS, ankylosing spondylarthritis; CD, Crohn’s disease; CPP, chronic plaque psoriasis; DKD, diabetic kidney disease; IR, incidence rate; JAKi, Janus kinase inhibitor; JIA, juvenile idiopathic arthritis; KT, kidney transplantation; PsA, psoriatic arthritis; RA, rheumatoid arthritis; RCT, randomised clinical trial; SLE, systemic lupus erythematosus; SS, systemic sclerosis; UC, ulcerative colitis; VTE, venous thromboembolic event.

Most of the meta-analysed data came from clinical trials rather than real-life studies with a longer follow-up period. The meta-analyses concluded that although the JAKi treatment is associated with an elevated risk of VTEs, the association is not statistically significance. Lastly, the meta-analyses did not encompass data on VTEs treated in primary care facilities (ie, on an outpatient basis). Two analyses of US medical–administrative databases did not find a difference in the VTE risk between patients with RA taking tofacitinib and those taking an antitumour necrosis factor agent (HR=1.13 (95% CI 0.77 to 1.65) and HR=1.33 (95% CI 0.78 to 2.24), respectively).38 39 However, the researchers could not rule out such a risk and only considered VTEs leading to hospital admission.38 39

A population-based study of a health insurance database (the Système National des Données de Santé (SNDS)) would provide additional insights by focusing on the VTE risk. The advantages of studying a health insurance database include the precise, national-level identification of JAKi prescriptions, VTEs and the time of occurrence (eg, relative to treatment initiation). Furthermore, studying AD avoids a major source of confounding bias; in contrast to RA and inflammatory bowel disease, AD is not associated with an increased risk of VTE40 and predominantly affects a younger population with a lower prevalence of concomitant cardiovascular comorbidities or obesity.

Here, we describe the protocol for the ‘JAK inhibitors and ThromboEmbolic Risk’ study of the association between JAKis and VTEs in AD using real-world evidence from an exhaustive French medical–administrative database. We also discuss our methodological choices. Our primary objective is to assess the risk of VTEs in adults with AD exposed to JAKis compared with AD adults not exposed to JAKis, and our secondary objective is to evaluate whether JAKi initiation acts as a trigger of VTEs in adults with AD within 3 months, corresponding to two different methodological approaches.

Methods and analysis

Overall study design

The literature data on the temporal relationship between the initiation of treatment with a JAKi and the occurrence of a VTE are contradictory. Some studies suggest that the incidence rates of VTEs are consistent over time,22 whereas other indicate that the incidence rates are clustered soon after the start of exposure.41 The study null hypotheses are formulated as follows: (1) VTE risk is equal in adults with AD exposed or not exposed to JAKis; and (2) JAKi initiation does not trigger VTE. We will therefore use two different methodological approaches to investigate the VTEs and the JAKis prescribed for AD: (1) a nested case–control study in a cohort of adults with AD (analysis 1) and (2) a case–time control study (analysis 2).

The overall study design is summarised in figure 1.

Figure 1.

Figure 1

Overall study design. AD, atopic dermatitis; JAKi, Janus kinase inhibitor.

Place and study time

The analysis period will run from January first, 2017, to August 31st, 2025, in France.

Data sources

We will analyse the French national health insurance database (SNDS), which covers 98% of the 66 million people in France. The SNDS database contains anonymous data on individuals’ demographic characteristics (sex, dates of birth and (if applicable) date of death); all healthcare reimbursements, including drugs (with the prescription filling date, the prescriber’s medical specialty, laboratory tests, outpatient care/visits, all hospital stays, and the associated diagnoses (coded according to the International Classification of Diseases, 10th Revision (ICD-10), all causes of death (classified according to the ICD-10 codes) and the attribution or not of ‘chronic disease’ status (‘affection de longue durée’ (ALD), giving entitlement to the full coverage of related healthcare costs and again coded according to ICD-10 codes). Information on medical procedures or biological results are not available in the SNDS.

Selection criteria and constitution of the target cohort

To avoid indication bias and form a homogeneous group of patients in terms of medical care, we will build up a cohort of adults with AD and who start systemic immunomodulatory treatment for this disease.

In France, AD is a chronic condition that is mostly managed in outpatient settings and not during hospital stays. Furthermore, AD does not give entitlement to ALD chronic disease status. All eligible adults (aged 18 or over) with a priori AD will be identified as follows:

  • Adults (aged 18 or over) with an initial fulfilment of a prescription for dupilumab, cyclosporine, methotrexate, tralokinumab or a JAKi (baricitinib, upadacitinib or abrocitinib), two or more fulfilments of topical corticosteroids, and a consultation with a dermatologist between 1 January 2017 and 31 December 2024.

  • Adults with no fulfilments of dupilumab, cyclosporine, methotrexate, tralokinumab or JAKi (baricitinib, upadacitinib or abrocitinib) prescriptions in the year prior to cohort entry.

  • Adults with no other indications for dupilumab, cyclosporine, methotrexate, tralokinumab or the JAKis baricitinib, upadacitinib or abrocitinib (ie, RA, PsA, AS, ulcerative colitis, lupus, organ or bone marrow transplant, nephrotic syndrome and psoriasis) identified through ‘ALD’ chronic disease status or the hospital discharge ICD-10 codes, between 1 January 2016 and 31 December 2024.

  • Adults with follow-up starting on the date of the first filled prescription of a JAKi (baricitinib, upadacitinib or abrocitinib), dupilumab, tralokinumab, cyclosporine or methotrexate, up to 31 August 2025.

Outcomes

The primary endpoint is VTE; it is a composite endpoint encompassing pulmonary embolism, managed mostly in hospital and identified through hospital discharge ICD-10 code (table 2) and deep vein thrombosis managed mostly in an outpatient setting and identified through a dedicated and validated algorithm (manuscript under review). The cases will be adults with AD and incident deep vein thrombosis or pulmonary embolism, managed in an outpatient setting, a hospital or an emergency department.

Table 2.

List of variables

Variables Registry Code
AD
 AD PMSI ICD-10 code L20
 Topical corticosteroids DCIR ATC codes D07AB01, D07AB02, D07AB03, D07AB04, D07AB05, D07AB06, D07AB07, D07AB08, D07AB09, D07AB10, D07AB11, D07AB19, D07AB21, D07AB30, D07AC01, D07AC02, D07AC03, D07AC04, D07AC05, D07AC06, D07AC07, D07AC08, D07AC09, D07AC10, D07AC11, D07AC12, D07AC13, D07AC14, D07AC15, D07AC16, D07AC17, D07AC18, D07AC19, D07AC20, D07AC21, D07AD01, D07AD02
 Consultation with a dermatologist DCIR PFS_SPE_COD or PFE_SPE_COD code 05
Exposure
 Baricitinib DCIR ATC code L04AA37
 Upadacitinib DCIR ATC code L04AA44
 Abrocitinib DCIR ATC code D11AH08
 Dupilumab DCIR ATC code D11AH05
 Tralokinumab DCIR ATC code D11AH07
 Cyclosporine DCIR ATC code L04AD01
 Methotrexate DCIR ATC code L01BA01
VTEs
 VTEs PMSI, DCIR EPIGETBAM algorithm under submission
Exclusion criteria
 Oral oestroprogestative DCIR ATC codes G03AA01, G03AA02, G03AA03, G03AA04, G03AA05, G03AA06, G03AA07, G03AA08, G03AA09, G03AA10, G03AA11, G03AA12, G03AA13, G03AA14, G03AA15, G03AA16, G03AB01, G03AB02, G03AB03, G03AB04, G03AB05, G03AB06, G03AB07, G03AB08
 Pregnancy PMSI ICD-10 code Z321
 Hospital stay >72 hours, with or without surgery PMSI ICD-10 codes
 Cancer and haematological malignancies PMSI ICD-10 codes C00 to C43 and C45 to C97, D00 to D03, D05 to D09, D37 to D48, or ALD n°30
 Anticoagulant treatment DCIR ATC codes B01AA01, B01AA02, B01AA03, B01AA04, B01AA07, B01AA08, B01AA09, B01AA10, B01AA11, B01AA12, B01AB01, B01AB02, B01AB04, B01AB05, B01AB06, B01AB07, B01AB08, B01AB09, B01AB10, B01AB11, B01AB12, B01AB51, B01AE01, B01AE02, B01AE03, B01AE04, B01AE05, B01AE06, B01AE07, B01AF01, B01AF02, B01AF03, B01A×01, B01A×04, B01A×05
 Rheumatoid arthritis PMSI
DCIR
ICD-10 codes M069, M0690, M0691, M0692, M0693, M0694, M0695, M0696, M0697, M0698, M0699, M06 or ALD n°22
 Psoriatic arthritis PMSI
DCIR
ICD-10 codes M0700, M0701, M0702, M0703, M0704, M0705, M0706, M0707, M0708, M0709, M072, M0720, M0721, M0722, M0723, M0724, M0725, M0726, M0727, M0728, M0729, M073, M0730, M0734, M0732, M0733, M0734, M0735, M0736, M0737, M0738, M0739
 Ulcerative colitis PMSI
DCIR
ICD-10 codes K519 or ALD n°24
 Lupus PMSI
DCIR
ICD-10 codes L93, M32 or ALD n°21
 Organ and bone marrow transplants PMSI
DCIR
ICD-10 codes Z940, Z941, Z942, Z943, Z944, Z945, Z946, Z947, Z948, Z9480, Z94800, Z94801, Z9481, Z9482, Z94802, Z94803, Z94804, Z94809, Z949
 Nephrotic syndrome PMSI
DCIR
ICD-10 code N04 or ALD n°19
 Psoriasis PMSI
DCIR
ICD-10 code L40, L400, L401, L402, L403, L404, L405, L408, L409
 Ankylosing spondylitis PMSI ICD-10 codes M45, M450, M451, M452, M453, M454, M455, M456, M457, M458, M459 or ALD n°27
Covariates
 Charlson Comorbidity Index PMSI Algorithm developed by Bannay et al.47
 Systemic corticosteroids DCIR ATC codes H02A and H02B
 Asthma PMSI
DCIR
ICD-10 codes J45, J450, J451, J458, J459, J46
ATC code R03
 Statins DCIR ATC codes C10AA, C10B

ALD long-term chronic disease status giving entitlement to full coverage of related healthcare costs.

AD, atopic dermatitis; ALD, Affection Longue Durée; ATC, Anatomical Therapeutic Chemical; DCIR, Données de Consommation Inter Régimes; ICD-10, International Classification of Diseases 10th Revision; PMSI, Programme de Médicalisation des Systèmes d’Information; VTE, venous thromboembolic event.

The index date is the date of the VTE.

To study cases of ‘unprovoked’ VTEs, we will exclude the following cases of adults with ‘provoked’ VTEs42:

  • Initiation of oral oestroprogestative contraception in the 3 months before the index date.

  • Pregnancy (including a 2-month postpartum period) before the index date.

  • Surgery (orthopaedic surgery involving long bones or the pelvis, or other major surgery) in the 4 weeks before the index date.

  • Prolonged hospitalisation (>72 hours) in the 4 weeks before the index date.

  • A diagnosis of cancer (including haematological malignancies but not including non-melanoma skin cancer) before the index date.

  • Fulfilment of one or more prescriptions for preventive or curative treatments with anticoagulants, including heparins, antivitamin K agents and direct oral anticoagulant (ensuring the exclusion of patients with a history of VTEs and persistent risk factors for VTE recurrence) before the index date (for VTEs managed in hospital or in an emergency department) or before the index date minus 7 days (for adults starting an anticoagulant treatment before hospitalisation for VTE).

Data analysis

The characteristics of the JAKis-treated population of patients with AD will be described, together with the time interval between JAKi initiation and the occurrence of the VTE. We will explore the risk function and the potential time-varying association.

Analysis 1: a nested case–control study of a cohort of adults with AD

The association between exposure to JAKis and the occurrence of VTEs will be investigated in a nested case–control study of a cohort of adults with AD requiring systemic treatment.

Adults with AD will be considered to have been exposed to JAKis if they have at least one fulfilled prescription for a JAKi prior to the index date. Adults with AD will be assigned to a ‘JAKi user’ category or a ‘JAKi never-user’ category, based on the prior fulfilment closest to the index date. Subgroups of JAKi users will be defined as follows: for current JAKis users, the last prescription will have been fulfilled in the month before the index date: for recent JAKis users, the last prescription will have been fulfilled between 1 and 4 months before the index date; and for past JAKis users, the last prescription will have been fulfilled more than 4 months before the index date. Furthermore, for current JAKi users; the number of JAKi prescription fulfilments and the total cumulative dose of JAKis received before the index date will be calculated.

References will be adults with AD whose most recent prescription fulfilment before the index date (regardless of how long before) will have been for another systemic treatment for AD.

For each case (adults with AD having experienced a VTE), four controls will be selected from the target AD cohort. Controls must not have experienced a VTE at the time of their selection. Cases and controls will be matched for age, sex and length of exposure at the case’s index date. The inclusion and exclusion criteria applied to cases will be applied to the matched controls. It will be possible for a control to become a case after his/her selection (density sampling).43 We will estimate ORs using conditional logistic regression. We will consider systemic treatment of AD as a binary variable: JAKi users (baricitinib, upadacitinib or abrocitinib) versus users of other systemic drugs (dupilumab, tralokinumab, cyclosporine or methotrexate). We will consider drug exposure as a continuous variable. The primary analysis will compare current JAKi users with JAKi never-users. The secondary analyses will cover ‘recent JAKi user’ status, ‘past JAKi user’ status and use of each individual JAKi (baricitinib, upadacitinib and abrocitinib). A Schneeweiss diagram for analysis 1 is shown in figure 2.44

Figure 2.

Figure 2

Schneeweiss diagram for analysis #1 (44). *Censored at the date of the first VTE, death, emigration or the end of the study period. **ED denotes the date of the first VTE (the index date). AD, atopic dermatitis; ED, event date; VTE, venous thromboembolic event.

Analysis 2: a case-only design – a nested case–time control study of a cohort of adults with AD

To evaluate whether or not initiation of a JAKi increases the risk of VTE in the following 3 months (ie, a ‘triggering effect’), we will perform a case–time control analysis.

In the field of pharmacoepidemiology, case–time control studies can be used to study an acute, early-onset adverse event during treatment.45 A VTE is sudden (with a short time interval between the pathophysiological cause and the clinical manifestations) and is easy to date by screening for specific treatments and additional investigations (including Doppler ultrasound). The majority of the VTEs observed in clinical trials22 or reported in pharmacovigilance databases41 occurred within 3–4 months of JAKi initiation.46 Furthermore, the case-only design can control for potential confounding factors (such as obesity and physical activity) not recorded in the French health insurance database.

Only patients with AD exposed to a JAKi and having experienced a VTE (ie, cases) will be analysed. The case–time control design compares the exposure status immediately before the event (the risk period) with exposure during a designated (earlier) reference period. Each VTE case will serve as his/her own control during a comparison of the risk period (0–3 months before occurrence of the VTE) with the reference period (3–6 months before occurrence of the VTE). Each VTE case will be assessed for exposure (yes/no) during the risk period and during the reference period. Only participants whose status differs when comparing the two periods (ie, discordants) will be considered in our estimation of the OR. To take account of the expected increase in JAKi prescription, the case–time control analysis will include a selection of controls matched with VTE cases. Each VTE case will be matched for age and sex with five controls without VTEs and who will be randomly selected from the AD target cohort. The date of the VTE will be used as the index date for the matched controls. The aforementioned defined risk and reference periods will be screened for JAKi initiation among the controls in the same way as among the cases, and a case-crossover OR for controls will be computed. The case–time control OR (95% CI) will be estimated with a conditional logistic model by considering the interaction term between the exposure of interest (JAKi initiation) and the participant’s status (case or control). The case–time control OR will correspond to the ratio between the respective case-crossover ORs obtained in cases and controls.

Sensitivity analyses in which the durations of the risk and reference period are modified will be performed as follows: the risk period will be defined as 0–2 months or 0–4 months before the VTE, and the control period will be defined as 2–4 months or 4–8 months before the VTE. Furthermore, sensitivity analysis will be performed for analyses 1 and 2 by changing the patient selection criteria and excluding patients with asthma. Lastly, we shall exclude patients having initiated oral oestroprogestative contraception in the 6 or 12 months before the date of the VTE in cases or the corresponding date in controls.

Covariates

We used a directed acyclic graph (figure 3) to describe covariates, mediators and potential confounding factors in the relationship between JAKis and VTEs.

Figure 3.

Figure 3

A directed acyclic graph of the relationship between JAKis, AD and VTEs. AD, atopic dermatitis; JAKi, Janus kinase inhibitor; RA, rheumatoid arthritis; UC, ulcerative colitis; VTE, venous thromboembolic event.

The results will be adjusted for several covariates, including the patient’s chronic comorbidities (using Bannay et al’s algorithm for use of the Charlson Comorbidity Index with an electronic healthcare database47 48) and the use of statins49 or systemic corticosteroids.50 Obesity is either not documented or only partially documented in the SNDS database; in Europe, most adults with AD are not obese.51 The case-only design approach (analysis 2) avoids this potential confounding factor, since the patient is his/her own control. The SNDS database does not contain identifiable information on a family history of venous thromboembolic disease.

Asthma (the most important atopic comorbidity in AD) will be assessed and defined as follows: an ICD-10 code J45–J46 and/or at least two fulfilments of a drug for the treatment of obstructive airway diseases (an Anatomical Therapeutic Chemical code of R03). The study variables are listed in table 2.

Sample size

Based on a frequency of exposure to JAKi among the targeted cohort of 25%, a 1:4 case:control ratio, and a statistical significance threshold of 0.05, the sample sizes required for a power of 80% in a comparison of JAKi exposure in cases versus controls are as follows: 1836 participants (306 cases and 1530 controls) for detecting an OR of 1.5, 618 participants (103 cases and 515 controls) for detecting an OR of 2, 354 participants (59 cases and 295 controls) for detecting an OR of 2.5, 246 participants (41 cases and 205 controls) for detecting an OR of 3 and 192 participants (32 cases and 160 controls) for detecting an OR of 3.5. These calculations do not take account of matching, which will tend to increase the power in an unknown manner. The estimated power calculation is given in table 3. A final power calculation will be performed at the end of the study.

Table 3.

Power calculation for analysis 1

Frequency of exposure to JAKis in the targeted cohort OR Nominal power Controls (n) Cases (n) Participants (total n)
0.50 1.5 0.8 1275 255 1530
0.50 2.0 0.8 465 93 558
0.50 3.0 0.8 205 41 246
0.25 1.5 0.8 1530 306 1836
0.25 2.0 0.8 515 103 618
0.25 2.5 0.8 295 59 354
0.25 3.0 0.8 205 41 246
0.25 3.5 0.8 160 32 192

JAKi, Janus kinase inhibitor.

The estimated incidence of thromboembolic diseases in France is one per 1000 per year; approximately 50 000 adults with a follow-up of 3 years are required. The target population for baricitinib/upadacitinib has been estimated between 26 500 and 42 500 by the French High Authority for Health52; this is almost certainly an underestimate, given that courses of treatment with cyclosporine are short.

Patient and public involvement

A patient will join the independent scientific committee and will participate in the discussion of the results. This patient is the director of the French Eczema Association (https://www.associationeczema.fr/). Once the study will be published, patients with AD who are members of the association will be informed of the results in the form of newsletter suitable for a non-specialist audience through the website of the association.

Ethics and dissemination

In accordance with French legislation, the protocol has been approved by an independent ethics committee (Comité éthique et scientifique pour les recherches, les études et les évaluations dans le domaine de la santé, Paris, France; reference: 4523600, dated 17 June 2021) and has been registered with the French National Data Protection Commission (Commission Nationale de l'Informatique et des Libertés, Paris, France; reference: 921265, dated 28 June 2021). The study’s findings will be published in peer-reviewed scientific journals and presented at international conferences.

The data will be consulted via the French national health insurance system’s (Caisse Nationale de l’Assurance Maladie) portal; the investigators’ access is restricted to the scope of the study. The data were not extracted from the main database but were analysed in a dedicated project area on the server. The investigators will comply with the reference framework applicable to the SNDS database (as set out in the government act dated 22 March 2017).

The study protocol has been registered at France’s Health Data Hub (www.health-data-hub.fr). The statistical analysis plan and data management book will now be drafted. The first results are expected in late 2025. The study’s findings will be published in peer-reviewed scientific journals and presented at international conferences.

Discussion

A population-based study of a cohort of adults with AD documented in the SNDS French national health insurance database should provide additional insights on the potential association between VTE and JAKis (baricitinib, upadacitinib and abrocitinib).

There are several possible pathophysiological explanations for an elevated risk of VTE during treatment with a JAKi. First, the leading hypothesis states that the thrombogenic effect is related to the thrombocytosis associated with baricitinib use.22 However, a clear time-domain or quantitative association between the platelet count and the occurrence of VTE has not been observed.22 Furthermore, elevation of the platelet count is not observed in people treated with other JAKis, including upadacitinib.53 Second, the JAK two pathway has an important role in haematopoiesis and might promote VTE. Paradoxically, inhibition of the JAK2 pathway by JAKis does not account for the occurrence of VTE: in Vaquez disease and essential thrombocythemia, an activating mutation in JAK 2 increases the risk of arterial and venous thrombotic events.54 Data from mouse models suggest that JAK V617F expression induces hypersensitivity to fibrinogen, thrombopoietin and other endogenous prothrombogenic factors.55

The literature data on the potential risk are contradictory and do not enable a firm conclusion about the association between JAKis and VTE to be drawn. A false association might result from methodological bias. For example, selection bias occurs when including patients who have received several courses of systemic treatment (and so might have more severe disease and a higher thromboembolic risk) are included in clinical trials (especially in open-label trials in RA).22 24 Confounding bias may occur because the disease treated with JAKi is itself associated with a higher risk of VTE; this is particularly true for RA. Indeed, the thromboembolic risk is known to be two to three times higher in patients with RA25 than in the general population.28 56 The baseline risk also appears to be elevated other systemic inflammatory diseases, including inflammatory bowel disease.57 58 In contrast, adults managed for moderate-to-severe AD are not known to have an elevated thromboembolic risk and are also younger than patients with RA; hence, the baseline risk of VTEs is lower. Published data on this indication are scarce: the only two meta-analyses included data from four randomised clinical trials evaluating the efficacy of baricitinib and abrocitinib in AD.34 The lack of a significant association might have several explanations: (1) a lack of power would apply if the number of JAKi-exposed patients experiencing a VTE is low; meta-analyses have provided inconclusive results due to the rarity of the event and the predominant inclusion of clinical trial data; (2) insufficient follow-up in clinical trials (given the latency between JAKi initiation and VTE occurrence); and (3) a lack of specific detection of VTEs (requiring a targeted initial assessment and follow-up and perhaps a longer follow-up period). Lastly, it is unclear whether the published studies considered only VTEs leading to a hospitalisation or, in contrast, all VTEs. In France, the majority of VTEs are managed in an outpatient setting.59

Our implementation of two complementary methodological approaches should shed more light on this question. The case–control study is carried out on a population of patients with AD with similar disease severity levels and receiving similar intensities of systemic treatment. This design assumes that after initiation of a JAKi, the risk of a VTE is constant. The case–time control design will be applied to address (1) the assumption whereby a JAKi triggers a VTE and (2 the issue of residual confounding factors. This study design is particularly suitable when the outcome is sudden and easily dated, as is the case here.60–62 The hypothetical triggering effect is based on (1) the transient thrombocytosis observed with baricitinib early after treatment initiation63 64; (2) pharmacovigilance data from France and North America,41 46 where more than half of the reported VTEs occurred within 120 days of JAKi initiation46; and (3) the fact that other drugs (such as contraceptives) can trigger VTEs.65–69 An increase over the study period in the prevalence of JAKi use for AD is expected; the case–time control design considers time trends in the prevalence of exposure that might introduce a confounding effect in a case-crossover design. We chose to study unprovoked VTEs by excluding well-known risk factors for thromboembolic disease,70 such as cancer,71 surgery,72 immobilisation (proxy marker: a hospital stay), hospital admission73 and the initiation of hormone therapy.74 Furthermore, we will adjust for the Charlson Comorbidity Index, which includes diabetes.75–78 However, obesity, black ethnicity79 and a family history of thromboembolic disease are not documented in the SNDS database, and so we cannot rule out residual confounding in analysis 1 (the nested case–control study). In analysis 2 (the case-only design), cases serve as their own controls, which can mitigate the potential confounding factors (such as diet, smoking, the level of physical activity and a family history of thromboembolic disease) not documented in healthcare databases.45 80

Our study has several potential strengths, including the exhaustive nationwide coverage of the French population (thereby enabling an assessment of rare events and providing potentially greater statistical power), the theoretical absence of selection bias, given our use of the SNDS database, the quality of the recorded data (enabling estimation of the time of occurrence of VTEs), the implementation of two complementary methodological approaches and the definitions of outcomes that encompass VTEs managed in outpatient and inpatient settings.

The study’s potential limitations include the difficulty of tracking all VTEs (the use of an algorithm for the identification of inpatient and outpatient diagnoses of VTE in the health insurance database is, however, currently being validated); potential information bias on hormone therapy, since a proportion of these treatments are not reimbursed and therefore cannot be detected in the SNDS; a potential lack of statistical power; and the inability to take account of some risk factors for VTEs (including obesity and a family history of thromboembolic disease) in the case–control design, although we believe that these potential confounding factors should affect cases and controls to the same extent.

Supplementary Material

Reviewer comments
Author's manuscript

Acknowledgments

We thank David Fraser for the English language revision and Stéphanie Mehrand, the Director of the French Eczema Association for her contribution to this work.

Footnotes

Twitter: @LM_Scailteux

Contributors: PB and CD wrote the first draft of the manuscript. PB, L-MS, AL, DS, GC, PG, AD, EO and CD conceived the scoping review and developed the research questions and the search strategy. All authors critically reviewed drafts and edited the manuscript.

Funding: The authors have not declared a specific grant for this research from any funding agency in the public, commercial or not-for-profit sectors.

Competing interests: None declared.

Patient and public involvement: Patients and/or the public were not involved in the design, conduct, reporting or dissemination plans of this research.

Provenance and peer review: Not commissioned; externally peer reviewed.

Data availability statement

No data are available.

Ethics statements

Patient consent for publication

Not applicable.

References

  • 1.Bieber T. Atopic dermatitis. N Engl J Med 2008;358:1483–94. 10.1056/NEJMra074081 [DOI] [PubMed] [Google Scholar]
  • 2.Silverberg JI, Hanifin JM. Adult eczema prevalence and associations with asthma and other health and demographic factors: a US population-based study. J Allergy Clin Immunol 2013;132:1132–8. 10.1016/j.jaci.2013.08.031 [DOI] [PubMed] [Google Scholar]
  • 3.Barbarot S, Auziere S, Gadkari A, et al. Epidemiology of atopic dermatitis in adults: results from an international survey. Allergy 2018;73:1284–93. 10.1111/all.13401 [DOI] [PubMed] [Google Scholar]
  • 4.Williams H, Robertson C, Stewart A, et al. Worldwide variations in the prevalence of symptoms of atopic eczema in the International study of asthma and allergies in childhood. J Allergy Clin Immunol 1999;103:125–38. 10.1016/s0091-6749(99)70536-1 [DOI] [PubMed] [Google Scholar]
  • 5.Flohr C, Mann J. New insights into the epidemiology of childhood atopic dermatitis. Allergy 2014;69:3–16. 10.1111/all.12270 [DOI] [PubMed] [Google Scholar]
  • 6.Wollenberg A, Barbarot S, Bieber T, et al. Consensus-based European guidelines for treatment of atopic eczema (atopic dermatitis) in adults and children: Part I. J Eur Acad Dermatol Venereol 2018;32:657–82. 10.1111/jdv.14891 [DOI] [PubMed] [Google Scholar]
  • 7.Wollenberg A, Barbarot S, Bieber T, et al. Consensus-based European guidelines for treatment of atopic eczema (atopic dermatitis) in adults and children: part II. J Eur Acad Dermatol Venereol 2018;32:850–78. 10.1111/jdv.14888 [DOI] [PubMed] [Google Scholar]
  • 8.Blauvelt A, de Bruin-Weller M, Gooderham M, et al. Long-term management of moderate-to-severe atopic dermatitis with dupilumab and concomitant topical corticosteroids (liberty AD CHRONOS): a 1-year, randomised, double-blinded, placebo-controlled, phase 3 trial. Lancet 2017;389:2287–303. 10.1016/S0140-6736(17)31191-1 [DOI] [PubMed] [Google Scholar]
  • 9.Simpson EL, Bieber T, Guttman-Yassky E, et al. Two phase 3 trials of dupilumab versus placebo in atopic dermatitis. N Engl J Med 2016;375:2335–48. 10.1056/NEJMoa1610020 [DOI] [PubMed] [Google Scholar]
  • 10.Simpson EL, Lacour J-P, Spelman L, et al. Baricitinib in patients with moderate-to-severe atopic dermatitis and inadequate response to topical corticosteroids: results from two randomized monotherapy phase III trials. Br J Dermatol 2020;183:242–55. 10.1111/bjd.18898 [DOI] [PubMed] [Google Scholar]
  • 11.Simpson EL, Forman S, Silverberg JI, et al. Baricitinib in patients with moderate-to-severe atopic dermatitis: results from a randomized monotherapy phase 3 trial in the United States and Canada (BREEZE-AD5). J Am Acad Dermatol 2021;85:62–70. 10.1016/j.jaad.2021.02.028 [DOI] [PubMed] [Google Scholar]
  • 12.Reich K, Kabashima K, Peris K, et al. Efficacy and safety of Baricitinib combined with topical corticosteroids for treatment of moderate to severe atopic dermatitis: a randomized clinical trial. JAMA Dermatol 2020;156:1333–43. 10.1001/jamadermatol.2020.3260 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.King B, Maari C, Lain E, et al. Extended safety analysis of Baricitinib 2 Mg in adult patients with atopic dermatitis: an integrated analysis from eight randomized clinical trials. Am J Clin Dermatol 2021;22:395–405. 10.1007/s40257-021-00602-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Guttman-Yassky E, Teixeira HD, Simpson EL, et al. Once-daily upadacitinib versus placebo in adolescents and adults with moderate-to-severe atopic dermatitis (measure up 1 and measure up 2): results from two replicate double-blind, randomised controlled phase 3 trials. Lancet 2021;397:2151–68. 10.1016/S0140-6736(21)00588-2 [DOI] [PubMed] [Google Scholar]
  • 15.Reich K, Teixeira HD, de Bruin-Weller M, et al. Safety and efficacy of upadacitinib in combination with topical corticosteroids in adolescents and adults with moderate-to-severe atopic dermatitis (AD up): results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2021;397:2169–81. 10.1016/S0140-6736(21)00589-4 [DOI] [PubMed] [Google Scholar]
  • 16.AbbVie . A phase 3B multicenter, randomized, double-blind, Double-Dummy, active controlled study comparing the safety and efficacy of Upadacitinib to Dupilumab in adult subjects with moderate to severe atopic dermatitis; 2021. https://clinicaltrials.gov/ct2/show/NCT03738397 [Accessed 05 July 2021].
  • 17.Simpson EL, Sinclair R, Forman S, et al. Efficacy and safety of abrocitinib in adults and adolescents with moderate-to-severe atopic dermatitis (JADE MONO-1): a multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet 2020;396:255–66. 10.1016/S0140-6736(20)30732-7 [DOI] [PubMed] [Google Scholar]
  • 18.Silverberg JI, Simpson EL, Thyssen JP, et al. Efficacy and safety of abrocitinib in patients with moderate-to-severe atopic dermatitis: a randomized clinical trial. JAMA Dermatol 2020;156:863–73. 10.1001/jamadermatol.2020.1406 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bieber T, Simpson EL, Silverberg JI, et al. Abrocitinib versus placebo or dupilumab for atopic dermatitis. N Engl J Med 2021;384:1101–12. 10.1056/NEJMoa2019380 [DOI] [PubMed] [Google Scholar]
  • 20.Wollenberg A, Blauvelt A, Guttman‐Yassky E, et al. Tralokinumab for moderate‐to‐severe atopic dermatitis: results from two 52‐week, randomized, double‐blind, multicentre, placebo‐controlled phase III trials (ECZTRA 1 and ECZTRA 2)*. Br J Dermatol 2021;184:437–49. 10.1111/bjd.19574 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Silverberg JI, Toth D, Bieber T, et al. Tralokinumab plus topical corticosteroids for the treatment of moderate-to-severe atopic dermatitis: results from the double-blind, randomized, multicentre, placebo-controlled phase III ECZTRA 3 trial. Br J Dermatol 2021;184:450–63. 10.1111/bjd.19573 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Taylor PC, Weinblatt ME, Burmester GR, et al. Cardiovascular safety during treatment with Baricitinib in rheumatoid arthritis. Arthritis Rheumatol 2019;71:1042–55. 10.1002/art.40841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.European Medicines Agency . olumiant EPAR risk management plan summary. Available: https://www.ema.europa.eu/en/documents/rmp-summary/olumiant-epar-risk-management-plan-summary_en.pdf [Accessed 22 July 2021].
  • 24.Food and Drug Administation . NDA 207924, Baricitinib, a JAK inhibitor for RA, 2018. Available: https://www.fda.gov/media/112372/download [Accessed 16 May 2021].
  • 25.Scott IC, Hider SL, Scott DL. Thromboembolism with janus kinase (JAK) inhibitors for rheumatoid arthritis: how real is the risk? Drug Saf 2018;41:645–53. 10.1007/s40264-018-0651-5 [DOI] [PubMed] [Google Scholar]
  • 26.Mogul A, Corsi K, McAuliffe L. Baricitinib: the second FDA-approved JAK inhibitor for the treatment of rheumatoid arthritis. Ann Pharmacother 2019;53:947–53. 10.1177/1060028019839650 [DOI] [PubMed] [Google Scholar]
  • 27.Cohen SB, van Vollenhoven RF, Winthrop KL, et al. Safety profile of upadacitinib in rheumatoid arthritis: integrated analysis from the select phase III clinical programme. Ann Rheum Dis 2021;80:304–11. 10.1136/annrheumdis-2020-218510 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.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. 10.1136/annrheumdis-2020-218419 [DOI] [PubMed] [Google Scholar]
  • 29.Downing LJ, Strieter RM, Kadell AM, et al. Il-10 regulates thrombus-induced vein wall inflammation and thrombosis. J Immunol 1998;161:1471–6. [PubMed] [Google Scholar]
  • 30.Riley JK, Takeda K, Akira S, et al. Interleukin-10 receptor signaling through the JAK-STAT pathway. Requirement for two distinct receptor-derived signals for anti-inflammatory action. J Biol Chem 1999;274:16513–21. 10.1074/jbc.274.23.16513 [DOI] [PubMed] [Google Scholar]
  • 31.Xie W, Huang Y, Xiao S, et al. Impact of Janus kinase inhibitors on risk of cardiovascular events in patients with rheumatoid arthritis: systematic review and meta-analysis of randomised controlled trials. Ann Rheum Dis 2019;78:1048–54. 10.1136/annrheumdis-2018-214846 [DOI] [PubMed] [Google Scholar]
  • 32.Xie W, Xiao S, Huang Y, et al. Effect of tofacitinib on cardiovascular events and all-cause mortality in patients with immune-mediated inflammatory diseases: a systematic review and meta-analysis of randomized controlled trials. Ther Adv Musculoskelet Dis 2019;11:1759720X1989549. 10.1177/1759720X19895492 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Olivera PA, Lasa JS, Bonovas S, et al. Safety of Janus kinase inhibitors in patients with inflammatory bowel diseases or other immune-mediated diseases: a systematic review and meta-analysis. Gastroenterology 2020;158:1554–73. 10.1053/j.gastro.2020.01.001 [DOI] [PubMed] [Google Scholar]
  • 34.Giménez Poderós T, Gallardo Borge S, Vazquez-Ferreiro P. Risk of venous thromboembolism associated with tofacitinib and Baricitinib: a systematic review and indirect meta-analysis. Pharmacotherapy 2020;40:1248–64. 10.1002/phar.2472 [DOI] [PubMed] [Google Scholar]
  • 35.Yates M, Mootoo A, Adas M, et al. Venous thromboembolism risk with JAK inhibitors: a meta-analysis. Arthritis Rheumatol 2021;73:779-788. 10.1002/art.41580 [DOI] [PubMed] [Google Scholar]
  • 36.Wang F, Sun L, Wang S, et al. Efficacy and safety of tofacitinib, Baricitinib, and upadacitinib for rheumatoid arthritis: a systematic review and meta-analysis. Mayo Clin Proc 2020;95:1404–19. 10.1016/j.mayocp.2020.01.039 [DOI] [PubMed] [Google Scholar]
  • 37.Bilal J, Riaz IB, Naqvi SAA, et al. Janus kinase inhibitors and risk of venous thromboembolism: a systematic review and meta-analysis. Mayo Clin Proc 2021;96:1861–73. 10.1016/j.mayocp.2020.12.035 [DOI] [PubMed] [Google Scholar]
  • 38.Desai RJ, Pawar A, Weinblatt ME, et al. Comparative risk of venous thromboembolism in rheumatoid arthritis patients receiving tofacitinib versus those receiving tumor necrosis factor inhibitors: an observational cohort study. Arthritis Rheumatol 2019;71:892–900. 10.1002/art.40798 [DOI] [PubMed] [Google Scholar]
  • 39.Desai RJ, Pawar A, Khosrow-Khavar F, et al. Risk of venous thromboembolism associated with tofacitinib in patients with rheumatoid arthritis: a population-based cohort study. Rheumatology 2021;61:121–30. 10.1093/rheumatology/keab294 [DOI] [PubMed] [Google Scholar]
  • 40.Meyers KJ, Silverberg JI, Rueda MJ, et al. Risk of venous thromboembolism among patients with atopic dermatitis: a cohort study in a US administrative claims database. Dermatol Ther 2021;11:1041–52. 10.1007/s13555-021-00538-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Setyawan J, Azimi N, Strand V, et al. Reporting of thromboembolic events with JAK inhibitors: analysis of the FAERS database 2010-2019. Drug Saf 2021;44:889–97. 10.1007/s40264-021-01082-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Di Nisio M, van Es N, Büller HR. Deep vein thrombosis and pulmonary embolism. The Lancet 2016;388:3060–73. 10.1016/S0140-6736(16)30514-1 [DOI] [PubMed] [Google Scholar]
  • 43.Rothman KJ, Greenland S, Lash TL. Modern epidemiology; 2008.
  • 44.Schneeweiss S, Rassen JA, Brown JS, et al. Graphical depiction of longitudinal study designs in health care databases. Ann Intern Med 2019;170:398–406. 10.7326/M18-3079 [DOI] [PubMed] [Google Scholar]
  • 45.Consiglio GP, Burden AM, Maclure M, et al. Case-crossover study design in pharmacoepidemiology: systematic review and recommendations. Pharmacoepidemiol Drug Saf 2013;22:1146–53. 10.1002/pds.3508 [DOI] [PubMed] [Google Scholar]
  • 46.Comité scientifique permanent Surveillance et pharmacovigilance du 24/09/2019 - Formation restreinte Expertise - Compte-rendu (07/02/2020). Available: https://archiveansm.integra.fr/Mediatheque/Publications/Ordres-du-jour-comptes-rendus-des-commissions-comites-groupes-de-travail-Comites-scientifiques-permanents [Accessed 20 May 2021].
  • 47.Bannay A, Chaignot C, Blotière P-O, et al. The best use of the charlson comorbidity index with electronic health care database to predict mortality. Med Care 2016;54:188–94. 10.1097/MLR.0000000000000471 [DOI] [PubMed] [Google Scholar]
  • 48.Charlson M, Szatrowski TP, Peterson J, et al. Validation of a combined comorbidity index. J Clin Epidemiol 1994;47:1245–51. 10.1016/0895-4356(94)90129-5 [DOI] [PubMed] [Google Scholar]
  • 49.Kunutsor SK, Seidu S, Khunti K. Statins and primary prevention of venous thromboembolism: a systematic review and meta-analysis. Lancet Haematol 2017;4:e83–93. 10.1016/S2352-3026(16)30184-3 [DOI] [PubMed] [Google Scholar]
  • 50.Johannesdottir SA, Horváth-Puhó E, Dekkers OM, et al. Use of glucocorticoids and risk of venous thromboembolism: a nationwide population-based case-control study. JAMA Intern Med 2013;173:743–52. 10.1001/jamainternmed.2013.122 [DOI] [PubMed] [Google Scholar]
  • 51.Zhang A, Silverberg JI. Association of atopic dermatitis with being overweight and obese: a systematic review and metaanalysis. J Am Acad Dermatol 2015;72:606–16. 10.1016/j.jaad.2014.12.013 [DOI] [PubMed] [Google Scholar]
  • 52.Haute Autorité de Santé . Olumiant et dermatite atopique, avis de la Commission de transparence, 2021. Available: https://www.has-sante.fr/upload/docs/evamed/CT-18912_OLUMIANT_PIC_EI_DA_AvisDef_CT18912.pdf [Accessed 23 July 2021].
  • 53.European Medicines Agency . Rinvoq-résumé des charactéristiques du produit, 2019. Available: https://www.ema.europa.eu/en/medicines/human/EPAR/rinvoq [Accessed 20 May 2021].
  • 54.Trifan G, Shafi N, Testai FD. Implications of Janus kinase 2 mutation in embolic stroke of unknown source. J Stroke Cerebrovasc Dis 2018;27:2572–8. 10.1016/j.jstrokecerebrovasdis.2018.05.052 [DOI] [PubMed] [Google Scholar]
  • 55.Hobbs CM, Manning H, Bennett C, et al. Jak2V617F leads to intrinsic changes in platelet formation and reactivity in a knock-in mouse model of essential thrombocythemia. Blood 2013;122:3787–97. 10.1182/blood-2013-06-501452 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Matta F, Singala R, Yaekoub AY, et al. Risk of venous thromboembolism with rheumatoid arthritis. Thromb Haemost 2009;101:134–8. [PubMed] [Google Scholar]
  • 57.Zöller B, Li X, Sundquist J, et al. Autoimmune diseases and venous thromboembolism: a review of the literature. Am J Cardiovasc Dis 2012;2:171–83. [PMC free article] [PubMed] [Google Scholar]
  • 58.Galloway J, Barrett K, Irving P, et al. Risk of venous thromboembolism in immune-mediated inflammatory diseases: a UK matched cohort study. RMD Open 2020;6:e001392. 10.1136/rmdopen-2020-001392 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Schmidt C. Traitement ambulatoire des thromboses veineuses profondes des membres inférieurs la phase aiguë Outpatient treatment of deep vein thrombosis. 6, 2002. [DOI] [PubMed] [Google Scholar]
  • 60.Maclure M. The case-crossover design: a method for studying transient effects on the risk of acute events. Am J Epidemiol 1991;133:144–53. 10.1093/oxfordjournals.aje.a115853 [DOI] [PubMed] [Google Scholar]
  • 61.Maclure M, Fireman B, Nelson JC, et al. When should case-only designs be used for safety monitoring of medical products? Pharmacoepidemiol Drug Saf 2012;21 Suppl 1:50–61. 10.1002/pds.2330 [DOI] [PubMed] [Google Scholar]
  • 62.Bykov K, Mittleman MA, Glynn RJ, et al. The case-crossover design for drug-drug interactions: considerations for implementation. Epidemiology 2019;30:204–11. 10.1097/EDE.0000000000000944 [DOI] [PubMed] [Google Scholar]
  • 63.Kremer J, Huizinga TWJ, Chen L. FRI0090 analysis of neutrophils, lymphocytes, and platelets in pooled phase 2 and phase 3 studies of baricitinib for rheumatoid arthritis. Ann Rheum Dis 2017;76:512. 10.1136/annrheumdis-2017-eular.1325 [DOI] [Google Scholar]
  • 64.European Medicines Agency . Olumiant. Available: https://www.ema.europa.eu/en/medicines/human/EPAR/olumiant
  • 65.Norris LA, Bonnar J. Haemostatic changes and the oral contraceptive pill. Baillieres Clin Obstet Gynaecol 1997;11:545–64. 10.1016/s0950-3552(97)80027-2 [DOI] [PubMed] [Google Scholar]
  • 66.Norris LA, Bonnar J. Effect of oestrogen dose on whole blood platelet activation in women taking new low dose oral contraceptives. Thromb Haemost 1994;72:926–30. [PubMed] [Google Scholar]
  • 67.Quehenberger P, Loner U, Kapiotis S, et al. Increased levels of activated factor VII and decreased plasma protein S activity and circulating thrombomodulin during use of oral contraceptives. Thromb Haemost 1996;76:729–34. [PubMed] [Google Scholar]
  • 68.Kelleher CC. Clinical aspects of the relationship between oral contraceptives and abnormalities of the hemostatic system: relation to the development of cardiovascular disease. Am J Obstet Gynecol 1990;163:392–5. 10.1016/0002-9378(90)90589-Y [DOI] [PubMed] [Google Scholar]
  • 69.Contraceptifs hormonaux combinés (pilules, anneau vaginal et patch) : Position finale du Comité des médicaments usage humain (CHMP) - Point d’information - ANSM : Agence nationale de sécurité du médicament et des produits de santé. Available: https://archiveansm.integra.fr/S-informer/Travaux-de-l-Agence-Europeenne-des-Medicaments-EMA-Comite-des-medicaments-a-usage-humain-CHMP/Contraceptifs-hormonaux-combines-pilules-anneau-vaginal-et-patch-Position-finale-du-Comite-des-medicaments-a-usage-humain-CHMP-Point-d-information [Accessed 22 Sep 2021].
  • 70.Heit JA, Spencer FA, White RH. The epidemiology of venous thromboembolism. J Thromb Thrombolysis 2016;41:3–14. 10.1007/s11239-015-1311-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Falanga A, Russo L, Milesi V, et al. Mechanisms and risk factors of thrombosis in cancer. Crit Rev Oncol Hematol 2017;118:79–83. 10.1016/j.critrevonc.2017.08.003 [DOI] [PubMed] [Google Scholar]
  • 72.Sweetland S, Parkin L, Balkwill A, et al. Smoking, surgery, and venous thromboembolism risk in women: United Kingdom cohort study. Circulation 2013;127:1276–82. 10.1161/CIRCULATIONAHA.113.001428 [DOI] [PubMed] [Google Scholar]
  • 73.Bjøri E, Johnsen HS, Hansen J-B, et al. Hospitalization as a trigger for venous thromboembolism - results from a population-based case-crossover study. Thromb Res 2019;176:115–9. 10.1016/j.thromres.2019.02.024 [DOI] [PubMed] [Google Scholar]
  • 74.Rott H. Prevention and treatment of venous thromboembolism during HRT: current perspectives. Int J Gen Med 2014;7:433–40. 10.2147/IJGM.S46310 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Aksu K, Donmez A, Keser G. Inflammation-Induced thrombosis: mechanisms, disease associations and management. Curr Pharm Des 2012;18:1478–93. 10.2174/138161212799504731 [DOI] [PubMed] [Google Scholar]
  • 76.van den Oever IAM, Sattar N, Nurmohamed MT. Thromboembolic and cardiovascular risk in rheumatoid arthritis: role of the haemostatic system. Ann Rheum Dis 2014;73:954–7. 10.1136/annrheumdis-2013-204767 [DOI] [PubMed] [Google Scholar]
  • 77.Gaertner S, Cordeanu E-M, Mirea C, et al. Increased risk and severity of unprovoked venous thromboembolism with clustering cardiovascular risk factors for atherosclerosis: results of the REMOTEV registry. Int J Cardiol 2018;252:169–74. 10.1016/j.ijcard.2017.11.055 [DOI] [PubMed] [Google Scholar]
  • 78.Ageno W, Becattini C, Brighton T, et al. Cardiovascular risk factors and venous thromboembolism: a meta-analysis. Circulation 2008;117:93–102. 10.1161/CIRCULATIONAHA.107.709204 [DOI] [PubMed] [Google Scholar]
  • 79.Zakai NA, McClure LA, Judd SE, et al. Racial and regional differences in venous thromboembolism in the United States in 3 cohorts. Circulation 2014;129:1502–9. 10.1161/CIRCULATIONAHA.113.006472 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hallas J, Pottegård A. Use of self-controlled designs in pharmacoepidemiology. J Intern Med 2014;275:581–9. 10.1111/joim.12186 [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Reviewer comments
Author's manuscript

Data Availability Statement

No data are available.


Articles from BMJ Open are provided here courtesy of BMJ Publishing Group

RESOURCES