Skip to main content
Erschienen in:

Open Access 28.02.2024 | Original Research

Use of Apremilast to Achieve Psoriatic Arthritis Treatment Goals and Satisfaction at 1 Year in the Canadian Real-World APPRAISE Study

verfasst von: Vinod Chandran, Louis Bessette, Carter Thorne, Maqbool Sheriff, Proton Rahman, Dafna D. Gladman, Sabeen Anwar, Jennifer Jelley, Anne-Julie Gaudreau, Manprit Chohan, John S. Sampalis

Erschienen in: Rheumatology and Therapy | Ausgabe 2/2024

Abstract

Introduction

The APPRAISE study was conducted to better understand the 12-month effectiveness, tolerability, and patient satisfaction with apremilast treatment for patients with psoriatic arthritis (PsA) in real-world settings.

Methods

APPRAISE (NCT03608657), a prospective, multicenter, observational study, enrolled adults with active PsA prescribed apremilast per routine care between July 2018 and March 2020. Patients were followed for 12 months with visits suggested every 4 months. The primary outcome measure was achievement of remission (REM) or low disease activity (LDA), defined as a Clinical Disease Activity Index for Psoriatic Arthritis (cDAPSA) score ≤ 13.

Results

Of the 102 patients who enrolled, 45 (44.1%) discontinued the study by 12 months. Most patients (75.5%) had moderate or high disease activity, and 24.5% were in REM/LDA at baseline based on cDAPSA score. Achievement of cDAPSA REM/LDA was 63.7%, 67.2%, and 53.8% at months 4, 8, and 12, respectively. In those continuing in the study, significant improvements were seen in swollen and tender joint counts, pain visual analog scale, psoriasis body surface area, and complete dactylitis resolution. Enthesitis reduction was also observed. Improvements in treatment satisfaction and patient-reported outcomes, including Health Assessment Questionnaire-Disability Index and the 36-item Short Form physical and mental component scores, were observed over 12 months. The proportion of patients achieving a Patient-Acceptable Symptom State (PASS) increased significantly from baseline at months 4, 8, and 12 (P < 0.001). Apremilast was well tolerated; the most frequent adverse events (AEs) leading to discontinuation were diarrhea (9/102 [8.8%]), nausea (4/102 [3.9%]), and migraine (4/102 [3.9%]).

Conclusion

In this real-world study conducted in Canadian rheumatology clinics, apremilast demonstrated clinical effectiveness in patients with active PsA, along with patient satisfaction with treatment. Safety findings were consistent with previously reported clinical data.

Trial Registration

ClinicalTrials.gov identifier, NCT03608657.
Hinweise
Prior Presentation These results were reported in part in a poster (abstract 57) at CRA 2020, 26–29 February in Victoria, British Columbia, Canada; an e-poster (abstract P22.08) at CDA 2020, 24–27 June in Ottawa, Ontario, Canada; an abstract (AB0557) at EULAR 2021, 2–5 June in Paris, France; and presented virtually in a poster at CRA 2021 (abstract 124), 24–26 February and CRA 2022 (abstract 109), 2–5 February.
Key Summary Points
Why carry out this study?
There are few real-world studies of apremilast use in patients with psoriatic arthritis, and none in a Canadian population.
APPRAISE sought to assess clinical disease activity, patient-reported outcomes, and safety of apremilast use in routine clinical care in Canada.
What was learned from the study
Over half of all patients reached remission or low disease activity after 4, 8, and 12 months of apremilast treatment.
In real-world clinical practice in Canada, apremilast was safe, and significant improvements were observed in multiple physician- and patient-reported outcomes, including Clinical Disease Activity Index for Psoriatic Arthritis (cDAPSA), swollen joint count (SJC), tender joint count (TJC), pain visual analog scale (VAS), body surface area (BSA), and Health Assessment Questionnaire-Disability Index (HAQ-DI).

Introduction

Patients with psoriatic arthritis (PsA) experience substantial disease burden and reduced quality of life [1]. PsA is associated with comorbidities such as obesity, diabetes, and cardiovascular disease [2]. Joint damage occurs in many patients, impairing physical function [3, 4]. Even in patients with oligoarticular PsA (≤ 4 involved joints), many patients report impairment of quality of life despite having few active joints involved [5]. Several studies have indicated that beginning treatment early in disease course is important for achieving optimal outcomes [69]. The goal of PsA treatment is to improve the signs and symptoms of the disease by achieving remission (REM) or low disease activity (LDA), and improving quality of life [10].
Many biologic therapies used for the treatment of PsA directly target inflammatory cytokines like tumor necrosis factor (TNF), interleukin (IL)-12/IL-23, IL-23, and IL-17 [1]. However, these treatments must be administered by injection and can be associated with significant adverse events, particularly infections [11]. Apremilast is an oral phosphodiesterase 4 (PDE4) inhibitor with a unique immunomodulating mechanism of action. Inhibition of PDE4 elevates intracellular cyclic adenosine monophosphate (cAMP) levels, thereby modulating the inflammatory response by reducing the expression of TNF, IL-23, IL-17, and increasing the pro-inflammatory activity of IL-10 [12]. In phase 3 studies, treatment with apremilast was associated with achievement of Clinical Disease Activity Index for Psoriatic Arthritis (cDAPSA) treatment targets at week 52 in patients with PsA, particularly for patients with moderate disease activity [13, 14]. In addition, there is evidence for real-world effectiveness of apremilast in studies of patients with PsA and psoriasis (PsO) [15, 16].
Apremilast is approved in Canada for the treatment of adults with PsA who have had an inadequate response to, were intolerant to, or had a contraindication to a prior disease-modifying antirheumatic drug (DMARD) [12]. No prospective real-world studies with apremilast in the Canadian population for treatment of PsA have been reported. To better understand the 12-month effectiveness, tolerability, and sustainability of apremilast treatment in real-world settings, the APPRAISE study evaluated clinical and patient-reported outcomes (PROs) in patients with PsA receiving routine clinical care in Canada for up to 12 months.

Methods

Study Design

APPRAISE (NCT03608657) was a prospective, multicenter, observational study that enrolled adult patients with active PsA who were prescribed apremilast per routine care between July 2018 and March 2020 in Ontario, Quebec, British Columbia, Saskatchewan, and Nova Scotia. A maximum of 30 days was permitted from apremilast treatment initiation to the screening/baseline visit. Patients were treated as per regional policies and the product monograph. Patients were followed from treatment initiation to 12 months, with suggested follow-up assessment visits every 4 months and a follow-up call 12 months after the end of the study. Sites were selected on the basis of their expertise in treating patients with PsA, access to apremilast through local reimbursement options, and their ability to collect and report data for this study at the required quality standards. APPRAISE was conducted in accordance with the Declaration of Helsinki’s general ethical principles with the study protocol approved by either regional or institutional ethics boards. The study protocol was approved by the Canadian Shields Ethics Review Board (#18-05-00) and the University Health Network Research Ethics Board (18-6022). Informed written consent was obtained from each patient prior to any study-related procedure.

Eligibility Criteria

This study enrolled adult patients (≥ 18 years of age) with active PsA who satisfied the Classification Criteria for Psoriatic Arthritis (CASPAR). The decision to initiate apremilast was made by the prescribing physician prior to and independent of patient enrollment in the study. There were no limitations on concurrent treatment. Patients were excluded if they were pregnant/planning to become pregnant or breastfeeding.

Endpoints

The primary endpoint was achievement of REM or LDA at month 12, defined as a cDAPSA score ≤ 13. cDAPSA is calculated as the sum of swollen joint count (SJC [0–66]), tender joint count (TJC [0–68]), and Patient’s Global Assessment of PsA (PtGA); visual analog scale (VAS [0–10 cm]), and pain (VAS [0–10 cm]). cDAPSA categories are defined as high disease activity (HDA), > 27; moderate disease activity (ModDA), > 13 to ≤ 27; LDA, > 4 to ≤ 13; and REM, ≤ 4.
Clinical assessments at months 4, 8, and 12 included change from baseline in cDAPSA, SJC, TJC, patient assessment of pain (0–100 mm VAS), body surface area (BSA), and Physician Global Assessment of PsA (PhGA) (0–100 mm VAS); and presence of dactylitis (assessed by individual physician assessment) and enthesitis (assessed by the physician using the Leeds enthesitis index).
PROs at months 4, 8, and 12 included PtGA (0–100 mm VAS), achievement of Patient-Acceptable Symptom State (PASS), Treatment Satisfaction Questionnaire for Medication (TSQM) total score, Health Assessment Questionnaire-Disability Index (HAQ-DI), 36-item Short-Form Health Survey version 2 (SF-36v2) physical component summary score (PCS) and mental component summary score (MCS), and work productivity index scores. To determine PASS, patients responded to this single question: “If you were to remain for the next few months as you were during the last week, would this be acceptable or unacceptable to you?” with a binary (Yes or No) response [17]. The TSQM (version 1.4) is a validated 14-item self-administered patient questionnaire that assesses patient satisfaction with treatment; scores range from 0 to 100, with a higher TSQM score indicating greater patient satisfaction [18]. There are four domains that make up the TSQM: effectiveness, side effects, convenience, and overall satisfaction. A TSQM license was obtained for this study (Opportunity #2806168). For information regarding or permission to use the TSQM, please refer to IQVIA at www.​iqvia.​com/​TSQM or TSQM@iqvia.com.
Common treatment-emergent adverse events (TEAEs) and AEs leading to discontinuation were also assessed.

Statistical Analysis

Baseline patient demographics were summarized descriptively. Outcomes at months 4, 8, and 12 were analyzed by paired Student’s t test or McNemar’s test. Data are presented as observed. SF-36 domain scores were transformed to T scores using a mean of 50 and standard deviation of 10.

Results

Patient Population

In total, 102 patients were enrolled across 18 rheumatology clinics. The majority of the population (96.1%) were white, women (56.9%), and had a mean age of 51.8 years (Table 1). Mean duration of PsA was 5.5 years. The most common PsA subtypes were oligoarticular arthritis (41.4%) and polyarticular arthritis (34.3%). Of the 92 patients who reported psoriasis, the most common subtype of psoriasis was plaque psoriasis (62.0%), followed by scalp (38.0%) and nail (33.7%).
Table 1
Demographics and baseline characteristics
Characteristic
N = 102
Age, mean (SD), years
51.8 (11.7)
Male, n (%)
44 (43.1)
Race, n (%)
 White
98 (96.1)
 Asian
2 (2.0)
 Other
2 (2.0)
Duration of PsA, mean (SD), years
5.5 (7.9)
PsA subtypea, n (%)
n = 99
 Oligoarticular
41 (41.4)
 Polyarticular
34 (34.3)
 Distal interphalangeal joint arthritis
26 (26.3)
 Spondylitis with peripheral arthritis
8 (8.1)
 Other
1 (1.0)
Psoriasis subtypea, n (%)
n = 92
 Plaque
57 (62.0)
 Scalp
35 (38.0)
 Nail
31 (33.7)
 Palmoplantar
8 (8.7)
Psoriasis-involved BSA (%), n (%)
n = 66
 < 3%
47 (71.2)
 ≥ 3% to ≤ 10%
12 (18.2)
 > 10%
7 (10.6)
SJC (0–66), mean (SD)
5.4 (5.4)
TJC (0–68), mean (SD)
7.5 (6.7)
Enthesitis, n/N (%)
33/97 (34.0)
Dactylitis, n/N (%)
18/99 (18.2)
 Dactylitis count, mean (SD)
2.8 (2.8)
Comorbidity, n (%)
 Anxiety/depressionb
26 (25.5)
  Anxiety
12 (11.8)
  Depression
20 (19.6)
 Cardiometabolic diseasec
50 (49.0)
Presence of comorbidities, n (%)
 ≥ 1
76 (74.5)
 > 2
46 (45.1)
 > 3
28 (27.5)
Concomitant PsA medications, n (%)
84 (82.4)
 bDMARDs
13 (12.7)
 cDMARDs
59 (57.8)
 Corticosteroids
18 (17.6)
 NSAIDs
31 (30.4)
Prior PsA medication, n (%)
 bDMARDs
18 (17.6)
 cDMARDs
74 (72.5)
  Methotrexate
59 (57.8)
  Sulfasalazine
21 (20.6)
  Hydroxychloroquine
14 (13.7)
  Leflunomide
11 (10.8)
  Chloroquine
1 (1.0)
Prior methotrexate dose, mean (SD), mg/week
17.8 (6.1)
n represents the number of patients; the number of patients with data available for each parameter may vary
bDMARD biologic DMARD, BSA body surface area, cDMARD conventional DMARD, DMARD disease-modifying antirheumatic drug, NSAID nonsteroidal anti-inflammatory drug, PsA psoriatic arthritis, SD standard deviation, SJC swollen joint count, TJC tender joint count
aPatients may be included in more than one category
bIncluding any of the following: anxiety, depressive disorders, depression
cIncluding any of the following: aortic valve incompetence, aortic valve replacement, arrhythmia, atherosclerosis, blood cholesterol increased, cerebrovascular accident, coronary artery disease, diabetes mellitus, dyslipidemia, hypercholesterolemia, hypertension, myocardial infarction, obesity, stent placement, tachycardia
There was a high prevalence of comorbidities in the population; 74.5% had at least one comorbidity and 45.1% had more than two comorbidities. Cardiometabolic disease was present in half of all patients and anxiety and/or depression were reported by the investigator in a quarter of the patients. Three-quarters had moderate or high disease activity at baseline based on cDAPSA score. Prior PsA medication use included biologic DMARDs (bDMARDs) in 17.6% of patients and conventional DMARDs (cDMARDs) in 72.5%. One patient had previously used a Janus kinase inhibitor. Most (57.8%) of the population was concurrently taking cDMARDs for PsA, 12.7% were concurrently taking bDMARDs, and 17.6% were concurrently taking corticosteroids.
Of the 102 patients who enrolled, the number of patients reaching 4, 8, and 12 months follow-up were 93, 71, and 57, respectively; 45 (44.1%) patients discontinued the study by 12 months (Fig. 1). Common reasons for discontinuation included lack or loss of effectiveness (20 [19.6%]) and AEs (19 [18.6%]). Median time to discontinuation including the 24-month follow-up call was 13.6 months.

Primary Endpoint

The proportion of patients in cDAPSA REM or LDA increased from 24.5% at baseline to 63.7% at month 4, 67.2% at month 8, and 53.8% at month 12 with apremilast treatment (Fig. 2a).

Treatment Satisfaction

Treatment satisfaction, as measured by TSQM, increased over time with apremilast, although the changes were not significant (Fig. 2b). Overall satisfaction increased by a mean of 7.1 from baseline to month 12. Improvements from baseline were observed in all subdomains; at 12 months, satisfaction with effectiveness improved by a mean of 9.3, side effects by 8.3, and convenience by 4.4.
At baseline, 26.3% of patients reported PASS. After 12 months of apremilast treatment, the proportion of patients reporting PASS increased to 68.0% (P < 0.001) (Fig. 2c).

Improvements in Clinical Variables

From baseline to month 12, significant improvements were observed in cDAPSA, SJC, TJC, pain VAS, and BSA (Table 2, Fig. 2d). Significant improvements were seen in these parameters at the earliest time point assessed (month 4). Mean (SD) change from baseline in cDAPSA scores at month 12 was − 8.5 (14.2). Mean (SD) change from baseline to month 12 in SJC was − 3.4 (5.2) and that in TJC was − 3.2 (7.5). Mean (SD) change from baseline in pain VAS was − 10.2 (26.0) and that in BSA was − 3.0 (7.3) at month 12. Decreases were also seen in PtGA, PhGA, and the percentage of patients reporting enthesitis and dactylitis at month 12, although these were not significant (Table 2). Among patients with enthesitis at baseline and available enthesitis data during follow-up, resolution of enthesitis was reported by 55.6%, 70.6%, and 60.0% at months 4, 8, and 12, respectively. Among patients with dactylitis at baseline and available dactylitis data during follow-up, complete resolution of dactylitis was reported by 41.2%, 66.7%, and 100% at months 4, 8, and 12, respectively.
Table 2
Disease/clinical parameters and PROs over time
 
Baseline
N = 102
Month 4
N = 93
Month 8
N = 71
Month 12
N = 56
Disease/clinical parameters
 cDAPSA score, mean (SD)
22.3 (13.2)
13.3 (11.0)
11.1 (9.0)
12.6 (9.9)
 SJC (0–66), mean (SD)
5.4 (5.4)
2.8 (1.5)
2.8 (1.3)
2.8 (1.0)
 TJC (0–68), mean (SD)
7.5 (6.7)
4.3 (6.5)
2.7 (3.3)
3.2 (5.6)
 PtGA VAS (0–100 mm), mean (SD)
50.2 (24.6)
38.3 (25.7)
38.3 (26.9)
44.1 (28.6)
 Pain VAS (0–100 mm), mean (SD)
48.6 (25.4)
37.7 (24.9)
35.9 (25.7)
38.1 (25.5)
 PhGA VAS (0–100 mm), mean (SD)
43.1 (19.1)
24.6 (17.5)
21.6 (18.0)
18.8 (16.0)
 Enthesitis, n/m (%)
33/97 (34.0)
21/86 (24.4)
10/55 (18.2)
9/48 (18.8)
 Resolution of enthesitisa, n/m (%)
15/27 (55.6)
12/17 (70.6)
6/10 (60.0)
 Dactylitis, n/m (%)
18/99 (18.2)
10/86 (11.6)
5/63 (7.9)
1/51 (2.0)
 Resolution of dactylitisb, n/m (%)
7/17 (41.2)
10/15 (66.7)
9/9 (100.0)
 BSA (%), mean (SD)
3.0 (6.0)
1.4 (3.0)
1.1 (2.5)
0.9 (1.8)
PROs
 HAQ-DI (0–3), mean (SD)
0.9 (0.7)
0.7 (0.7)
0.7 (0.6)
0.6 (0.6)
 SF-36v2, mean (SD)
  PCS score
34.4 (11.1)
37.5 (11.2)
38.1 (10.2)
39.0 (10.7)
  MCS score
45.5 (11.7)
49.0 (11.7)
50.8 (10.0)
49.6 (10.5)
The number of patients with data available for each parameter may vary
N the number of patients in the study at the time point, n/m the number of patients with the condition/the number of patients with data available for the assessment
BSA psoriasis-involved body surface area, cDAPSA Clinical Disease Activity Index for PsA, HAQ-DI Health Assessment Questionnaire-Disability Index, MCS mental component score, PCS physical component score, PhGA Physician’s Global Assessment of PsA, PROs patient-reported outcomes, PtGA Patient’s Global Assessment of PsA, SD standard deviation, SF-36v2 36-item Short Form Health Survey version 2.0, SJC swollen joint count, TJC tender joint count, VAS visual analog scale
aPercentages based on patients with enthesitis at baseline (N = 33) and available enthesitis data during follow-up
bPercentages based on patients with dactylitis at baseline (N = 18) and available dactylitis data during follow-up

Improvements in PROs

In addition to clinical parameters, significant improvements were observed in PROs. Mean HAQ-DI score significantly decreased at months 4 and 8 (Table 2), with a mean change from baseline of − 0.13 and − 0.15, respectively. A decrease from baseline of − 0.16 was seen at month 12, although this was not significant. Significant decreases were seen in SF-36 PCS score at all time points and in SF-36 MCS score at months 8 and 12 (Table 2). Mean changes in SF-36 MCS and PCS were clinically meaningful at months 8 and 12, as defined by a minimal clinically important difference of ≥ 2.5 [19, 20]. Improvements were also observed in SF-36 subdomains, particularly bodily pain, vitality, social functioning, and emotional role (Fig. 3).

Safety

Overall, more than 80% of patients experienced any TEAE (Table 3). Most TEAEs were mild to moderate in severity. Common TEAEs included diarrhea, headache, nausea, and upper respiratory tract infection. Diarrhea (9/102 [8.8%]), nausea (4/102 [3.9%]), and migraine (4/102 [3.9%]) were the most common AEs in patients who discontinued as a result of AEs. A total of five serious AEs (SAEs) occurred in four patients. Abdominal pain was reported by two patients, seizure and gastric cancer by one patient, and a fall by one patient. None were determined to be related to apremilast treatment and all but one resolved. There were no deaths, major adverse cardiac events (MACE), or COVID-19 cases during the study.
Table 3
Overview and common TEAEs
Overview of TEAEs, n (%)
N = 102
Any TEAEs
83 (81.4)
SAEsa
4 (3.9)
 Serious infections
1 (1.0)
TEAEs occurring in ≥ 5% of patients
 Diarrhea
27 (26.5)
 Nausea
14 (13.7)
 Headache
10 (9.8)
 Upper respiratory tract infection
7 (6.9)
TEAEs leading to discontinuation
21 (20.6)
 Diarrhea
9 (8.8)
 Nausea
4 (3.9)
 Migraine
4 (3.9)
 Abdominal discomfort
2 (2.0)
 Depression
2 (2.0)
 Drug ineffective
1 (1.0)
 Headache
1 (1.0)
 Labile hypertension
1 (1.0)
 Mood altered
1 (1.0)
 Pulmonary mass
1 (1.0)
 Upper respiratory tract infection
1 (1.0)
 Vomiting
1 (1.0)
SAE serious adverse event, TEAE treatment-emergent adverse event
aTwo patients experienced adverse events requiring hospitalization

Discussion

In this real-world study, apremilast demonstrated effectiveness for the treatment of PsA. After 12 months of treatment, significant improvements were observed in multiple measures of disease activity, including cDAPSA, SJC, TJC, pain VAS, and BSA. Of patients with enthesitis and dactylitis at baseline, 60% and 100% reported enthesitis and dactylitis to be resolved, respectively, at 12 months. Treatment satisfaction was high, with two-thirds of patients reporting overall satisfaction at month 12 based on the validated TSQM. This is consistent with results from the randomized, placebo-controlled UNVEIL study that demonstrated mean global satisfaction scores of approximately 60% with apremilast at 1 year in patients with chronic plaque PsO [21]. The proportion of patients who reported an acceptable disease state increased from 26.3% at baseline to 68.0% at month 12 with apremilast treatment. Improvements were also seen in physical function, as measured by HAQ-DI and SF-36. Changes in HAQ-DI were not, on average, clinically meaningful (as defined by a minimal clinically important difference of ≥ 0.35) [22]. However, it should be noted that the HAQ-DI is associated with several limitations in evaluating patients with PsA and establishing a standard for clinically meaningful improvement is difficult [23]. Despite this effectiveness and treatment satisfaction, approximately 40% of patients discontinued by month 12. Discontinuations were mainly due to lack or loss of effectiveness and AEs. Most TEAEs were mild to moderate in severity (> 95%) and the most common TEAEs were diarrhea, headache, and nausea, consistent with the known safety profile of apremilast [2427]. Collectively, these results show many practical benefits of apremilast treatment in clinical practice.
APPRAISE complements phase 3 clinical trial data of apremilast [13, 28]. Pooled data from the PALACE 1–3 studies showed that 46.9% of patients with ModDA and 24.9% with HDA at baseline achieved cDAPSA treatment targets of REM or LDA at 52 weeks [13]. In APPRAISE, the proportions of patients achieving REM or LDA increased from a quarter (24.5%) at baseline to half (53.8%) at month 12. Achievement of cDAPSA treatment targets was associated with improvements in other PsA manifestations in PALACE 1–3 for up to 5 years as well [13, 28], in line with the improvements in SJC, TJC, enthesitis, dactylitis, and skin involvement seen at month 12 in APPRAISE. Notably, the proportions of patients in REM/LDA increased from week 16 to 52 in PALACE 1–3 [13] but decreased from month 4 to 12 in APPRAISE. The high rate of discontinuations in APPRAISE may have been a factor in this observation.
APPRAISE is one of few real-world evidence studies of apremilast, particularly apremilast treatment of PsA. The results of APPRAISE are aligned with the findings from the real-world APOLO study. APOLO was an observational, prospective study of apremilast in 106 patients with PsA in Belgium (NCT03096990) [15]. In APOLO, improvements were seen in enthesitis, dactylitis, SJC, and TJC over 12 months of treatment. Reported TEAEs were also similar between APOLO and APPRAISE; 45.3% of patients experienced any TEAE in APOLO and the most common TEAEs were diarrhea, nausea, and headache. The mean time since PsA diagnosis was 7.3 years in APOLO compared to 5.5 years PsA duration in APPRAISE. The shorter duration of PsA in APPRAISE, as well as the higher prevalence of oligoarticular arthritis, supports apremilast use earlier in disease progression. Despite the lower number of joints involved with oligoarthritis compared to polyarthritis, patients report impaired quality of life. The collective efficacy results from APPRAISE support the potential for apremilast to improve quality of life in these patients through the improvement of clinical outcome measures as well as patient satisfaction measures.
Additional real-world studies have supported the safety and effectiveness of apremilast for the treatment of psoriasis [29, 30]. High rates of comorbidities have been reported in these studies, including hypertension and other cardiovascular diseases, depression, diabetes, and cancer [2931]. Similarly, comorbidities were high in the APPRAISE population at baseline, particularly cardiometabolic comorbidities, and approximately 20% of patients reported depression at baseline. Despite this, there were no new reports of TEAEs of MACE in APPRAISE. The real-world safety profile of apremilast in patients with psoriasis was similar to the profile seen in patients with PsA in APPRAISE, supporting consistent drug safety across patient populations [29]. In addition to safety and effectiveness, one study of apremilast use in Quebec found that treatment persistence was comparable between apremilast and biologics and use of apremilast resulted in significant healthcare savings [32].

Limitations

The study was limited by a lack of racial diversity. COVID-19 restrictions impacted in-office assessment visits, necessitating reliance on virtual visits. However, all sites were encouraged to have patients return to clinic for an in-person visit at month 12 for a full assessment. In addition to limiting clinical assessments, the COVID-19 pandemic may have impacted PROs. Real-world evidence studies do not have the rigor of randomized, placebo-controlled studies. Although reflective of a real-world treatment environment, the lack of limitations on concurrent treatment may confound the results. Reasons for apremilast initiation were not captured. Fatigue is an important aspect of PsA that was not directly captured in this study. Additionally, fibromyalgia and chronic pain syndrome were not evaluated, which may have impacted patient pain scores. Effectiveness assessments in this study were limited to 12 months, limiting the interpretation of long-term effectiveness and safety. The study also is limited by as-observed aggregate analyses and loss of patients over time. Apremilast was initiated at the clinical discretion of the treating physician, and there were no limitations for threshold of disease activity. As a result, some patients were in LDA at the beginning of the study. Because of this, some patients may not have shown a higher degree of disease improvement that is observed in phase 3 studies that include patients with moderate to high disease activity.

Conclusion

In this real-world analysis, patients with PsA who continued apremilast treatment in Canada achieved improvements in clinical and PRO parameters after 4 months, and the majority reported being satisfied with their achieved disease state. Consistent rates of improvement in meaningful clinical disease measures, such as pain, tender and swollen joints, and dactylitis resolution were observed at 8 and 12 months as well. Apremilast demonstrated effectiveness over 12 months in patients with PsA in Canadian clinical practices, with an efficacy and safety profile consistent with previously reported clinical data.

Acknowledgements

Medical Writing/Editorial Assistance.

Writing support was funded by Amgen Inc. and provided by Rebecca Lane, PhD, of Peloton Advantage, LLC, an OPEN Health company. JSS Medical Research Inc., the study management CRO, performed the statistical analyses for the study.

Declarations

Conflict of Interest

Vinod Chandran has received grant/research support from and/or served as a consultant for AbbVie, Amgen Inc., Bristol Myers Squibb, Celgene, Eli Lilly, Janssen, Novartis, Pfizer, and UCB Pharma. His spouse is an employee of AstraZeneca. Louis Bessette has received grant/research support from and/or served as a consultant for AbbVie, Amgen Inc., Bristol Myers Squibb, Celgene, Eli Lilly, Janssen, Merck, Pfizer, Sanofi, Novartis, and UCB Pharma. Carter Thorne has served on an advisory board for AbbVie, Amgen Inc., Celgene, Eli Lilly, Medexus/Medac, Merck, Novartis, Pfizer, Sandoz, and Sanofi; and has served as a consultant for Centocor, Medexus/Medac, and Merck. Maqbool Sheriff has served on an advisory board and received honoraria from AbbVie, Celgene, Eli Lilly, Janssen, Merck, Pfizer, and Sandoz. Proton Rahman has served as a speaker for and received honoraria from AbbVie, Amgen Inc., Celgene, Eli Lilly, Janssen, Merck, Novartis, Pfizer, and UCB Pharma. Dafna D. Gladman has received grant/research support from or served as a consultant for AbbVie, Amgen, Bristol Myers Squibb, Celgene, Eli Lilly, Galapagos, Gilead, Janssen, Novartis, Pfizer, and UCB. Sabeen Anwar has received grant/research support from AbbVie, Amgen Inc., Eli Lilly, Janssen, and Pfizer; and has served as a speaker or consultant for AbbVie, Amgen Inc., Bristol Myers Squibb, Novartis, and Pfizer. Jennifer Jelley, Anne-Julie Gaudreau, and Manprit Chohan are employees of and stockholders in Amgen Inc. John S. Sampalis is an employee of JSS Medical Research.

Ethical Approval

APPRAISE was conducted in accordance with the Declaration of Helsinki’s general ethical principles with the study protocol approved by the Canadian Shields Ethics Review Board (#18-05-00) and the University Health Network Research Ethics Board (18-6022). Informed written consent was obtained from each patient prior to any study-related procedure.
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.
Literatur
1.
Zurück zum Zitat Ritchlin CT, Colbert RA, Gladman DD. Psoriatic arthritis. N Engl J Med. 2017;376(10):957–70.CrossRefPubMed Ritchlin CT, Colbert RA, Gladman DD. Psoriatic arthritis. N Engl J Med. 2017;376(10):957–70.CrossRefPubMed
2.
Zurück zum Zitat Ogdie A, Schwartzman S, Husni ME. Recognizing and managing comorbidities in psoriatic arthritis. Curr Opin Rheumatol. 2015;27(2):118–26.CrossRefPubMed Ogdie A, Schwartzman S, Husni ME. Recognizing and managing comorbidities in psoriatic arthritis. Curr Opin Rheumatol. 2015;27(2):118–26.CrossRefPubMed
3.
Zurück zum Zitat Gladman DD, Stafford-Brady F, Chang CH, Lewandowski K, Russell ML. Longitudinal study of clinical and radiological progression in psoriatic arthritis. J Rheumatol. 1990;17(6):809–12.PubMed Gladman DD, Stafford-Brady F, Chang CH, Lewandowski K, Russell ML. Longitudinal study of clinical and radiological progression in psoriatic arthritis. J Rheumatol. 1990;17(6):809–12.PubMed
4.
Zurück zum Zitat Husted JA, Gladman DD, Farewell VT, Cook RJ. Health-related quality of life of patients with psoriatic arthritis: a comparison with patients with rheumatoid arthritis. Arthritis Rheum. 2001;45(2):151–8.CrossRefPubMed Husted JA, Gladman DD, Farewell VT, Cook RJ. Health-related quality of life of patients with psoriatic arthritis: a comparison with patients with rheumatoid arthritis. Arthritis Rheum. 2001;45(2):151–8.CrossRefPubMed
5.
Zurück zum Zitat Kasiem FR, Luime JJ, Vis M, et al. Lessons learned from clinical phenotypes in early psoriatic arthritis: the real-world Dutch south west Early Psoriatic ARthritis study. Scand J Rheumatol. 2021;50(2):124–31.CrossRefPubMed Kasiem FR, Luime JJ, Vis M, et al. Lessons learned from clinical phenotypes in early psoriatic arthritis: the real-world Dutch south west Early Psoriatic ARthritis study. Scand J Rheumatol. 2021;50(2):124–31.CrossRefPubMed
6.
Zurück zum Zitat Coates LC, Moverley AR, McParland L, et al. Effect of tight control of inflammation in early psoriatic arthritis (TICOPA): a UK multicentre, open-label, randomised controlled trial. Lancet. 2015;386(10012):2489–98.CrossRefPubMedPubMedCentral Coates LC, Moverley AR, McParland L, et al. Effect of tight control of inflammation in early psoriatic arthritis (TICOPA): a UK multicentre, open-label, randomised controlled trial. Lancet. 2015;386(10012):2489–98.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Gladman DD, Thavaneswaran A, Chandran V, Cook RJ. Do patients with psoriatic arthritis who present early fare better than those presenting later in the disease? Ann Rheum Dis. 2011;70(12):2152–4.CrossRefPubMed Gladman DD, Thavaneswaran A, Chandran V, Cook RJ. Do patients with psoriatic arthritis who present early fare better than those presenting later in the disease? Ann Rheum Dis. 2011;70(12):2152–4.CrossRefPubMed
8.
Zurück zum Zitat Haroon M, Gallagher P, FitzGerald O. Diagnostic delay of more than 6 months contributes to poor radiographic and functional outcome in psoriatic arthritis. Ann Rheum Dis. 2015;74(6):1045–50.CrossRefPubMed Haroon M, Gallagher P, FitzGerald O. Diagnostic delay of more than 6 months contributes to poor radiographic and functional outcome in psoriatic arthritis. Ann Rheum Dis. 2015;74(6):1045–50.CrossRefPubMed
9.
Zurück zum Zitat Theander E, Husmark T, Alenius GM, et al. Early psoriatic arthritis: short symptom duration, male gender and preserved physical functioning at presentation predict favourable outcome at 5-year follow-up. Results from the Swedish Early Psoriatic Arthritis Register (SwePsA). Ann Rheum Dis. 2014;73(2):407–13.CrossRefPubMed Theander E, Husmark T, Alenius GM, et al. Early psoriatic arthritis: short symptom duration, male gender and preserved physical functioning at presentation predict favourable outcome at 5-year follow-up. Results from the Swedish Early Psoriatic Arthritis Register (SwePsA). Ann Rheum Dis. 2014;73(2):407–13.CrossRefPubMed
10.
Zurück zum Zitat Gossec L, Baraliakos X, Kerschbaumer A, et al. EULAR recommendations for the management of psoriatic arthritis with pharmacological therapies: 2019 update. Ann Rheum Dis. 2020;79(6):700–12.CrossRefPubMed Gossec L, Baraliakos X, Kerschbaumer A, et al. EULAR recommendations for the management of psoriatic arthritis with pharmacological therapies: 2019 update. Ann Rheum Dis. 2020;79(6):700–12.CrossRefPubMed
11.
Zurück zum Zitat Kamata M, Tada Y. Efficacy and safety of biologics for psoriasis and psoriatic arthritis and their impact on comorbidities: a literature review. Int J Mol Sci. 2020;21(5):1690. Kamata M, Tada Y. Efficacy and safety of biologics for psoriasis and psoriatic arthritis and their impact on comorbidities: a literature review. Int J Mol Sci. 2020;21(5):1690.
12.
Zurück zum Zitat Otezla [product monograph]. Mississauga, ON, Canada: Amgen Canada Inc.; August 2020. Otezla [product monograph]. Mississauga, ON, Canada: Amgen Canada Inc.; August 2020.
13.
Zurück zum Zitat Mease PJ, Gladman DD, Ogdie A, et al. Treatment-to-target with apremilast in psoriatic arthritis: the probability of achieving targets and comprehensive control of disease manifestations. Arthritis Care Res (Hoboken). 2020;72(6):814–21.CrossRefPubMed Mease PJ, Gladman DD, Ogdie A, et al. Treatment-to-target with apremilast in psoriatic arthritis: the probability of achieving targets and comprehensive control of disease manifestations. Arthritis Care Res (Hoboken). 2020;72(6):814–21.CrossRefPubMed
14.
Zurück zum Zitat Mease PJ, Kavanaugh A, Ogdie A, et al. Baseline disease activity predicts achievement of cDAPSA treatment targets with apremilast: phase 3 results in DMARD-naive patients with psoriatic arthritis. J Rheumatol. 2022;49:694–9.CrossRefPubMed Mease PJ, Kavanaugh A, Ogdie A, et al. Baseline disease activity predicts achievement of cDAPSA treatment targets with apremilast: phase 3 results in DMARD-naive patients with psoriatic arthritis. J Rheumatol. 2022;49:694–9.CrossRefPubMed
15.
Zurück zum Zitat de Vlam K, Toukap AN, Kaiser MJ, et al. Real-world efficacy and safety of apremilast in Belgian patients with psoriatic arthritis: results from the prospective observational APOLO study. Adv Ther. 2022;39(2):1055–67.CrossRefPubMedPubMedCentral de Vlam K, Toukap AN, Kaiser MJ, et al. Real-world efficacy and safety of apremilast in Belgian patients with psoriatic arthritis: results from the prospective observational APOLO study. Adv Ther. 2022;39(2):1055–67.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Augustin M, Kleyn CE, Conrad C, et al. Characteristics and outcomes of patients treated with apremilast in the real world: results from the APPRECIATE study. J Eur Acad Dermatol Venereol. 2021;35(1):123–34.CrossRefPubMed Augustin M, Kleyn CE, Conrad C, et al. Characteristics and outcomes of patients treated with apremilast in the real world: results from the APPRECIATE study. J Eur Acad Dermatol Venereol. 2021;35(1):123–34.CrossRefPubMed
17.
Zurück zum Zitat Lubrano E, Scriffignano S, Azuaga AB, Ramirez J, Cañete JD, Perrotta FM. Assessment of the Patient Acceptable Symptom State (PASS) in psoriatic arthritis: association with disease activity and quality of life indices. RMD Open. 2020;6(1):e001170.CrossRefPubMedPubMedCentral Lubrano E, Scriffignano S, Azuaga AB, Ramirez J, Cañete JD, Perrotta FM. Assessment of the Patient Acceptable Symptom State (PASS) in psoriatic arthritis: association with disease activity and quality of life indices. RMD Open. 2020;6(1):e001170.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Atkinson MJ, Sinha A, Hass SL, et al. Validation of a general measure of treatment satisfaction, the Treatment Satisfaction Questionnaire for Medication (TSQM), using a national panel study of chronic disease. Health Qual Life Outcomes. 2004;2:12.CrossRefPubMedPubMedCentral Atkinson MJ, Sinha A, Hass SL, et al. Validation of a general measure of treatment satisfaction, the Treatment Satisfaction Questionnaire for Medication (TSQM), using a national panel study of chronic disease. Health Qual Life Outcomes. 2004;2:12.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Strand V, Mease P, Burmester GR, et al. Rapid and sustained improvements in health-related quality of life, fatigue, and other patient-reported outcomes in rheumatoid arthritis patients treated with certolizumab pegol plus methotrexate over 1 year: results from the RAPID 1 randomized controlled trial. Arthritis Res Ther. 2009;11(6):R170.CrossRefPubMedPubMedCentral Strand V, Mease P, Burmester GR, et al. Rapid and sustained improvements in health-related quality of life, fatigue, and other patient-reported outcomes in rheumatoid arthritis patients treated with certolizumab pegol plus methotrexate over 1 year: results from the RAPID 1 randomized controlled trial. Arthritis Res Ther. 2009;11(6):R170.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Gladman DD, Mease PJ, Cifaldi MA, Perdok RJ, Sasso E, Medich J. Adalimumab improves joint-related and skin-related functional impairment in patients with psoriatic arthritis: patient-reported outcomes of the Adalimumab Effectiveness in Psoriatic Arthritis Trial. Ann Rheum Dis. 2007;66(2):163–8.CrossRefPubMed Gladman DD, Mease PJ, Cifaldi MA, Perdok RJ, Sasso E, Medich J. Adalimumab improves joint-related and skin-related functional impairment in patients with psoriatic arthritis: patient-reported outcomes of the Adalimumab Effectiveness in Psoriatic Arthritis Trial. Ann Rheum Dis. 2007;66(2):163–8.CrossRefPubMed
21.
Zurück zum Zitat Stein Gold L, Papp K, Leonardi C, et al. Efficacy and safety of apremilast in patients with mild to moderate plaque psoriasis: results of a phase 3, multicenter, randomized, double-blind, placebo-controlled trial. J Am Acad Dermatol. 2022;86(1):77–85.CrossRefPubMed Stein Gold L, Papp K, Leonardi C, et al. Efficacy and safety of apremilast in patients with mild to moderate plaque psoriasis: results of a phase 3, multicenter, randomized, double-blind, placebo-controlled trial. J Am Acad Dermatol. 2022;86(1):77–85.CrossRefPubMed
22.
Zurück zum Zitat Mease PJ, Woolley JM, Bitman B, Wang BC, Globe DR, Singh A. Minimally important difference of Health Assessment Questionnaire in psoriatic arthritis: relating thresholds of improvement in functional ability to patient-rated importance and satisfaction. J Rheumatol. 2011;38(11):2461–5.CrossRefPubMed Mease PJ, Woolley JM, Bitman B, Wang BC, Globe DR, Singh A. Minimally important difference of Health Assessment Questionnaire in psoriatic arthritis: relating thresholds of improvement in functional ability to patient-rated importance and satisfaction. J Rheumatol. 2011;38(11):2461–5.CrossRefPubMed
23.
Zurück zum Zitat Mease P, Strand V, Gladman D. Functional impairment measurement in psoriatic arthritis: importance and challenges. Semin Arthritis Rheum. 2018;48(3):436–48.CrossRefPubMed Mease P, Strand V, Gladman D. Functional impairment measurement in psoriatic arthritis: importance and challenges. Semin Arthritis Rheum. 2018;48(3):436–48.CrossRefPubMed
24.
Zurück zum Zitat Kavanaugh A, Mease PJ, Gomez-Reino JJ, et al. Treatment of psoriatic arthritis in a phase 3 randomized, placebo-controlled trial with apremilast, an oral phosphodiesterase 4 inhibitor. Ann Rheum Dis. 2014;73(6):1020–6.CrossRefPubMed Kavanaugh A, Mease PJ, Gomez-Reino JJ, et al. Treatment of psoriatic arthritis in a phase 3 randomized, placebo-controlled trial with apremilast, an oral phosphodiesterase 4 inhibitor. Ann Rheum Dis. 2014;73(6):1020–6.CrossRefPubMed
25.
Zurück zum Zitat Cutolo M, Myerson GE, Fleischmann R, et al. A phase III, randomized, controlled trial of apremilast in patients with psoriatic arthritis: results of the PALACE 2 trial. J Rheumatol. 2016;43(9):1724–34.CrossRefPubMed Cutolo M, Myerson GE, Fleischmann R, et al. A phase III, randomized, controlled trial of apremilast in patients with psoriatic arthritis: results of the PALACE 2 trial. J Rheumatol. 2016;43(9):1724–34.CrossRefPubMed
26.
Zurück zum Zitat Edwards CJ, Blanco FJ, Crowley J, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with psoriatic arthritis and current skin involvement: a phase III, randomised, controlled trial (PALACE 3). Ann Rheum Dis. 2016;75(6):1065–73.CrossRefPubMed Edwards CJ, Blanco FJ, Crowley J, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with psoriatic arthritis and current skin involvement: a phase III, randomised, controlled trial (PALACE 3). Ann Rheum Dis. 2016;75(6):1065–73.CrossRefPubMed
27.
Zurück zum Zitat Wells AF, Edwards CJ, Kivitz AJ, et al. Apremilast monotherapy in DMARD-naive psoriatic arthritis patients: results of the randomized, placebo-controlled PALACE 4 trial. Rheumatology (Oxford). 2018;57(7):1253–63.CrossRefPubMed Wells AF, Edwards CJ, Kivitz AJ, et al. Apremilast monotherapy in DMARD-naive psoriatic arthritis patients: results of the randomized, placebo-controlled PALACE 4 trial. Rheumatology (Oxford). 2018;57(7):1253–63.CrossRefPubMed
28.
Zurück zum Zitat Kavanaugh A, Gladman DD, Edwards CJ, et al. Long-term experience with apremilast in patients with psoriatic arthritis: 5-year results from a PALACE 1–3 pooled analysis. Arthritis Res Ther. 2019;21(1):118.CrossRefPubMedPubMedCentral Kavanaugh A, Gladman DD, Edwards CJ, et al. Long-term experience with apremilast in patients with psoriatic arthritis: 5-year results from a PALACE 1–3 pooled analysis. Arthritis Res Ther. 2019;21(1):118.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Papadavid E, Rompoti N, Theodoropoulos K, Kokkalis G, Rigopoulos D. Real-world data on the efficacy and safety of apremilast in patients with moderate-to-severe plaque psoriasis. J Eur Acad Dermatol Venereol. 2018;23(7):1173–9.CrossRef Papadavid E, Rompoti N, Theodoropoulos K, Kokkalis G, Rigopoulos D. Real-world data on the efficacy and safety of apremilast in patients with moderate-to-severe plaque psoriasis. J Eur Acad Dermatol Venereol. 2018;23(7):1173–9.CrossRef
30.
Zurück zum Zitat Armstrong A, Levi E. Real-world clinical experience with apremilast in a large US retrospective cohort study of patients with moderate to severe plaque psoriasis. J Drugs Dermatol. 2017;16(12):1240–5.PubMed Armstrong A, Levi E. Real-world clinical experience with apremilast in a large US retrospective cohort study of patients with moderate to severe plaque psoriasis. J Drugs Dermatol. 2017;16(12):1240–5.PubMed
31.
Zurück zum Zitat Gottlieb AB, Merola JF, Cirulli J, et al. Characteristics of patients with psoriasis treated with apremilast in the Corrona Psoriasis Registry. Dermatol Ther (Heidelb). 2021;11(1):253–63. Gottlieb AB, Merola JF, Cirulli J, et al. Characteristics of patients with psoriasis treated with apremilast in the Corrona Psoriasis Registry. Dermatol Ther (Heidelb). 2021;11(1):253–63.
32.
Zurück zum Zitat Poulin Y, Beauchemin C, Royer C, et al. Real-world experience with apremilast in the treatment of adults with moderate to severe plaque psoriasis in Québec: a claims-based analysis of drug utilization and healthcare resource utilization. J Cutan Med Surg. 2020;24(6):573–87.CrossRefPubMed Poulin Y, Beauchemin C, Royer C, et al. Real-world experience with apremilast in the treatment of adults with moderate to severe plaque psoriasis in Québec: a claims-based analysis of drug utilization and healthcare resource utilization. J Cutan Med Surg. 2020;24(6):573–87.CrossRefPubMed
Metadaten
Titel
Use of Apremilast to Achieve Psoriatic Arthritis Treatment Goals and Satisfaction at 1 Year in the Canadian Real-World APPRAISE Study
verfasst von
Vinod Chandran
Louis Bessette
Carter Thorne
Maqbool Sheriff
Proton Rahman
Dafna D. Gladman
Sabeen Anwar
Jennifer Jelley
Anne-Julie Gaudreau
Manprit Chohan
John S. Sampalis
Publikationsdatum
28.02.2024
Verlag
Springer Healthcare
Erschienen in
Rheumatology and Therapy / Ausgabe 2/2024
Print ISSN: 2198-6576
Elektronische ISSN: 2198-6584
DOI
https://doi.org/10.1007/s40744-024-00641-w

Kompaktes Leitlinien-Wissen Innere Medizin (Link öffnet in neuem Fenster)

Mit medbee Pocketcards schnell und sicher entscheiden.
Leitlinien-Wissen kostenlos und immer griffbereit auf ihrem Desktop, Handy oder Tablet.

Neu im Fachgebiet Innere Medizin

Fortgeschrittenes Melanom: Wann den Checkpoint-Inhibitor absetzen?

Eine ICI-Therapie sollte bei Betroffenen mit fortgeschrittenem Melanom mindestens ein Jahr fortgesetzt werden. Bei anhaltendem Ansprechen kann danach offenbar ohne hohes Risiko ein Therapieabbruch erwogen werden.

Therapiestopp bei älteren MS-Kranken kann sich lohnen

Eine Analyse aus Kanada bestätigt: Setzen ältere MS-Kranke die Behandlung mit Basistherapeutika ab, müssen sie kaum mit neuen Schüben und MRT-Auffälligkeiten rechnen.

Dank Nasenspray seltener in die Notaufnahme

Durch die intranasale Applikation von Etripamil lassen sich paroxysmale supraventrikuläre Tachykardien (PSVT) oft in Eigenregie beenden. Das erspart den Betroffenen das Aufsuchen von Notfallambulanzen.

Vorhofflimmern: So häufig kommt es bei Katheterablation zu Embolien

Arterielle Embolien – insbesondere Hirnembolien - sind eine mögliche periprozedurale Komplikation bei Katheterablation von Vorhofflimmern. Wie hoch ist das Risiko? Eine Analyse von weltweit mehr als 300.000 Ablationsprozeduren gibt darüber Auskunft.

EKG Essentials: EKG befunden mit System (Link öffnet in neuem Fenster)

In diesem CME-Kurs können Sie Ihr Wissen zur EKG-Befundung anhand von zwölf Video-Tutorials auffrischen und 10 CME-Punkte sammeln.
Praxisnah, relevant und mit vielen Tipps & Tricks vom Profi.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.