All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a healthcare professional.

The PsOPsA Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your PsOPsA Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The PsOPsA Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the PsOPsA Hub cannot guarantee the accuracy of translated content. The PsOPsA Hub and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2022-07-11T15:13:22.000Z

Upadacitinib vs adalimumab or placebo in patients with an inadequate response to non-biological therapy: Week 56 results from the SELECT-PsA 1 study

Jul 11, 2022
Share:
Learning objective: After reading this article, learners will be able to recall key efficacy and safety data from the SELECT-PsA 1 trial of upadacitinib for the treatment of psoriatic arthritis.

Bookmark this article

Psoriatic arthritis (PsA) is an inflammatory disease that can present with multiple clinical manifestations, including psoriasis, peripheral arthritis, dactylitis, enthesitis, spondylitis, quality of life, physical function, pain, and fatigue.1 Treatment strategies focus on maximizing patient outcomes by controlling inflammation and preventing irreversible joint damage and disability. Currently, treatment options for PsA include several disease-modifying antirheumatic drugs (DMARDs), yet <1/3 of patients with PsA achieve minimal disease activity (MDA) in most placebo-controlled trials so new treatment options are needed.1

Upadacitinib, an oral, reversible, Janus associated kinase (JAK) inhibitor, is approved in Europe for the treatment of rheumatoid arthritis, PsA, and ankylosing spondylitis.2 The SELECT-PsA 1 trial (NCT03104400) is investigating the effect of upadacitinib compared with adalimumab (standard of care treatment) or placebo in adult patients with active PsA and an inadequate response or intolerance to ≥1 non-biological DMARD. Results of the 56-week efficacy and safety analysis were published by Iain McInnes et al.1 in the journal Rheumatic & Musculoskeletal Diseases Open. A summary of these results is provided below.

Study design and patient characteristics

This was a phase III, randomized, double-blind, placebo-controlled trial to assess the efficacy and safety of upadacitinib (15 mg or 30 mg once daily) compared with adalimumab (40 mg every other week) or placebo in 1,705 adult patients (≥18 years of age) with active PsA and inadequate response or intolerance to ≥1 non-biological DMARD. The study design can be seen in Figure 1.

Figure 1. Study design*

ADA, adalimumab; D, day; DMARD, disease-modifying antirheumatic drug; EOW, every other week; PsA, psoriatic arthritis; QD, once daily; SJC, swollen joint count; TCJ, tender joint count; UPA, upadacitinib; W, week.
*Data from McInnes et al.1
Data from ClinicalTrials.gov.3
One patient in the UPA 15 mg cohort did not receive study drug.
§Compared with baseline at two consecutive visits.

Baseline characteristics were well balanced across all cohorts (Table 1).

Table 1. Baseline characteristics*

Characteristic, % (unless otherwise stated)

Placebo
(n = 423)

UPA
15 mg QD
(n = 429)

UPA
30 mg QD
(n = 423)

ADA
40 mg EOW
(n = 429)

Mean age ± SD, years

50.4 ± 12.2

51.6 ± 12.2

49.9 ± 12.4

51.4 ± 12.0

Female

49.9

55.5

55.8

51.7

Ethnicity, white

89.1

90.0

89.1

87.4

Mean time since PsA diagnosis ± SD, years

6.2 ± 7.0

6.2 ± 7.4

5.9 ± 6.4

5.9 ± 7.1

Monotherapy

18.0

17.7

18.2

19.1

Any NB DMARD at baseline

82.0

82.3

81.8

80.9

              MTX alone

63.1

65

63.4

62.9

              MTX + another NB DMARD

6.1

4.7

6.4

3.7

              NB DMARD other than MTX

12.8

12.6

12.1

14.2

Steroid use at baseline

16.5

17.0

16.8

16.8

Mean TJC68 ± SD

20.0 ± 14.3

20.4 ± 14.7

19.4 ± 13.3

20.1 ± 13.8

Mean SJC66 ± SD

11.0 ± 8.2

11.6 ± 9.3

10.6 ± 7.1

11.6 ± 8.8

hs-CRP >ULN§

76.6

75.5

76.6

71.8

Mean HAQ-DI ± SD

1.1 ± 0.6

1.2 ± 0.7

1.1 ± 0.6

1.1 ± 0.6

Mean patient’s assessment of pain (NRS 0–10) ± SD

6.1 ± 2.1

6.2 ± 2.1

5.9 ± 2.1

6.0 ± 2.1

BSA-psoriasis ≥3%

49.9

49.9

49.6

49.2

Mean PASI for baseline BSA-psoriasis ≥3% ± SD

11.2 ± 11.4

9. 8 ± 10.0

9.5 ± 8.8

9.4 ± 8.5

BSA-psoriasis ≥0%,

94.6

91.8

93.9

92.5

sIGA of psoriasis score

 

 

 

 

              0

5.7

7.9

5.0

7.9

              1

20.3

17.0

18.4

15.2

              2

39.5

39.6

40.9

42.2

              3

28.1

31.0

30.3

30.8

              4

6.4

4.4

5.4

4.0

Presence of enthesitis
(defined as LEI >0)

57.0

62.9

63.1

61.8

Presence of dactylitis
(defined as LDI >0)

29.8

31.7

30.0

29.6

ADA, adalimumab; BSA, body surface area; DMARD, disease-modifying anti-rheumatic drug; EOW, every other week; HAQ-DI, Health Assessment Questionnaire - Disability Index; hs-CRP, high-sensitivity C-reactive protein; LDI, Leeds Dactylitis Index; LEI, Leeds Enthesitis Index; MTX, methotrexate; NB, non-biologic; NRS, numeric rating scale; NSAID, nonsteroidal anti-inflammatory drug; PASI, psoriasis area severity index; PsA, psoriatic arthritis; QD, once daily; SD, standard deviation; sIGA, Static Investigator Global Assessment of Psoriasis; SJC66, swollen joint count of 66 joints; TJC68, tender joint count of 68 joints; ULN, upper limit normal; UPA, upadacitinib.
*Adapted from McInnes, et al.1
Participants were categorized according to self-reported ethnicity.4
Permitted concomitant non-biologic DMARDs included methotrexate, sulfasalazine, leflunomide, apremilast, hydroxychloroquine, bucillamine, and iguratimod.
§
ULN >2.87 mg/L.

Results

In total, 83.2% of the patients enrolled in the trial completed 56 weeks of treatment. The most common reasons for discontinuation were withdrawal by the patient and lack of efficacy.

Efficacy

The proportions of patients achieving ≥20%/50%/70% improvement in American College of Rheumatology response criteria (ACR20/50/70) response were maintained from Week 24 to Week 56 across all treatment groups. A greater proportion of patients originally randomized to upadacitinib achieved ACR20/50/70 compared with those randomized to adalimumab at Week 56 (non-responder imputation analysis: nominal p ≤ 0.05 for 15 mg upadacitinib vs adalimumab for ACR50/70; nominal p ≤ 0.05 for 30 mg upadacitinib vs adalimumab for ACR20/50/70). ACR70 responses can be seen in Figure 2.

Figure 2. Percentage of patients achieving ACR70*

ACR70, 70% improvement in American College of Rheumatology response criteria; ADA, adalimumab; UPA, upadacitinib; PBO, placebo.

*Adapted from McInnes, et al.1

All ACR components were improved from baseline across all cohorts. Patients who switched from placebo to upadacitinib had comparable results to those originally randomized to upadacitinib (Table 2).

Table 2. ACR components and patient-reported outcomes at 56 weeks*

Parameter

Placebo-UPA 15 mg QD
(n = 211)

Placebo-UPA 30 mg QD
(n = 212)

UPA 15 mg QD
(n = 429)

UPA 30 mg QD
(n = 423)

ADA 40 mg EOW
(n = 429)

PhGA

−4.7

(−4.9 to −4.5)

−4.8

(−5.1 to −4.6)

−4.9

(−5.0 to −4.7)

−4.9

(−5.0 to −4.7)

−4.7

(−4.8 to −4.5)

SJC66

−9.2

(−9.7 to −8.7)

−9.6

(−10.1 to −9.2)

−9.7

(−10.1 to −9.4)

−9.8

(−10.1 to −9.4)

−9.6

(−9.9 to −9.3)

TJC68

−15.4

(−16.4 to −14.3)

−15.6

(−16.7 to −14.6)

−15.7

(−16.4 to −14.9)

−15.8

(−16.5 to −15.0)

−15.3

(−16.1 to −14.6)

C-reactive

protein

−7.5

(−8.6 to −6.5)

−8.7

(−9.8 to −7.7)

−7.8

(−8.6 to −7.1)

−8.6

(−9.3 to −7.9)

−7.5

(−8.2 to −6.7)

HAQ-DI

−0.40

(−0.48 to −0.32)

−0.45

(−0.53 to −0.38)

−0.54

(−0.59 to −0.48)§

−0.56

(−0.61 to −0.50)

−0.43

(−0.49 to −0.38)

FACIT-F

7.2

(5.9 to 8.6)

8.8

(7.4 to 10.1)

8.9

(8.0 to 9.9)

8.4

(7.4 to 9.4)

7.6

(6.7 to 8.6)

SF-36 PCS

9.3

(8.0 to 10.5)

9.8

(8.6 to 11.1)

10.8

(10.0 to 11.7)§

10.5

(9.6 to 11.4)

8.9

(8.0 to 9.8)

SF-36 MCS

3.6

(2.3 to 4.9)

4.0

(2.7 to 5.3)

5.2

(4.2 to 6.1)

4.4

(3.4 to 5.3)

4.3

(3.4 to 5.2)

Self-assessment

of psoriasis symptoms

−28.1

(−30.5 to −25.6)

−30.3

(−32.8 to −27.9)

−29.6

(−31.4 to −27.9)§

−30.4

(−32.2 to −28.7)

−25.8

(−27.6 to −24.1)

WPAI overall work

impairment

 

−21.9

(−27.1 to −16.7)

−24.7

(−30.0 to −19.5)

−24.5

(−28.1 to −20.9)

−22.9

(−26.5 to −19.3)

−20.8

(−24.5 to −17.1)

PtGA (NRS)

−3.4

(−3.7 to −3.0)

−3.7

(−4.0 to −3.3)

−3.7

(−3.9 to −3.4)§

−3.6

(−3.9 to −3.4)

−3.2

(−3.4 to −2.9)

Patients’

assessment of pain (NRS)

−3.1

(−3.5 to −2.8)

−3.3

(−3.7 to −3.0)

−3.3

(−3.6 to −3.1)§

 

−3.3

(−3.6 to −3.1)

−2.9

(−3.1 to −2.7)

DAPSA

−38.9

(−41.1 to −36.7)

−41.6

(−43.8 to −39.3)

−40.0

(−41.5 to −38.4)

−41.0

(−42.6 to −39.4)

−38.5

(−40.1 to −36.9)

ACR, American College of Rheumatology; ADA, adalimumab; DAPSA, Disease Activity in Psoriatic Arthritis; EOW, every other week; FACIT-F, Functional Assessment of Chronic Illness Therapy-Fatigue; HAQ-DI, Health Assessment Questionnaire-Disability Index; MCS, Mental Component Summary; MMRM, mixed-effects model repeated measures; NRS, numeric rating scale; PCS, Physical Component Summary; PhGA, Physicians’ Global Assessment of Disease Activity; PtGA, Patients' Global Assessment of Disease Activity; QD, once daily; SF-36, Short Form Health Survey questionnaire; SJC66 swollen joint count of 66 joints; TJC68, tender joint count of 68 joints; UPA, upadacitinib; WPAI, Work Productivity and Activity Impairment.
*Data adapted from McInnes, et al.1
All values are least squares mean change from baseline (95% confidence Interval), and analyses were conducted using an MMRM model based on as observed data.
p ≤ 0.05; for upadacitinib 30 mg QD versus adalimumab.
§
p ≤ 0.05; for upadacitinib 15 mg QD versus adalimumab.
Assessed in patients employed at baseline.

  • In all treatment groups, the proportion of patients achieving Psoriatic Arthritis Response Criteria (PsARC) was also maintained from Week 24 to Week 56.
    • At Week 56, more patients achieved PsARC response with upadacitinib 30 mg compared with adalimumab (nominal p ≤ 0.05).
  • Based on linear extrapolation at Week 56, mean changes from baseline in radiographic endpoints were comparable between both upadacitinib cohorts and the adalimumab cohort.
  • MDA responses can be seen in Figure 3.
    • For those originally randomized to upadacitinib or adalimumab, the proportion of patients achieving MDA were maintained from Week 24 to Week 56.
    • Overall, more patients in the upadacitinib 30 mg cohort achieved MDA compared with the adalimumab cohort (nominal p ≤ 0.05).
    • For the placebo cohorts, the percentage of patients achieving MDA increased when they were switched to upadacitinib.
  • Improvements in the skin outcomes Psoriasis Area and Severity Index (PASI75/90/100) and Static Investigator Global Assessment of Psoriasis of 0 or 1 (sIGA 0/1) response rates were maintained over time.
    • After patients switched to upadacitinib from placebo, the proportion of patients achieving PASI75/90/100 and sIGA 0/1 increased and were similar to the upadacitinib cohorts by Week 36.

Figure 3. Percentage of patients achieving MDA (NRI)*

ADA, adalimumab; MDA, minimal disease activity; NRI, non-responder imputation; PBO, placebo; UPA, upadacitinib.
*Adapted from McInnes, et al.1
NRI with additional rescue handling was used, where patients rescued at Week 16 are imputed as non-responders.

Patient-reported outcomes

Improvements in Health Assessment Questionnaire-Disability Index (HAQ-DI), Functional Assessment of Chronic Illness Therapy-Fatigue, Short Form Health Survey questionnaire (SF-36), Physical Component Summary (PCS) and Mental Component Summary (MCS), patients’ assessment of pain, Patients’ Global Assessment of Disease Activity (PtGA), and Work Productivity and Activity Impairment (WPA) were maintained from Week 24 to Week 56.

Patient reported outcomes at Week 56 are detailed in Table 2.

Safety

  • At Week 56, treatment-emergent adverse event (TEAE) rates were highest in the 30 mg upadacitinib cohort (333.9 events/100 patient years vs 281.1 [UPA 15 mg] and 265.9 [ADA]).
  • The most frequent AEs were upper respiratory tract infection and blood creatine phosphokinase elevations.
  • Exposure-adjusted event rates of TEAEs at Week 56 can be seen in Table 3.
    • Of note, patients in the UPA 30 mg cohort had higher rates of infection.
  • Event rates of malignancy were similar across the upadacitinib and adalimumab treatment groups; no notable malignancy patterns or types were observed.

Table 3. Exposure-adjusted event rates of treatment-emergent AEs at Week 56.

AE, events/100 PY

UPA 15 mg
(n = 617)
PY = 839.1

UPA 30 mg
(n = 613)
PY = 842.8

ADA 40 mg
(n = 429)
PY = 631.4

Infection

94.9

114.3

71.1

              Serious infection

2.9

4.7

1.3

              Opportunistic infection

0.4

0.8

0.0

              Herpes zoster

3.9

6.4

0.5

GI perforation

0.2

0.0

0.0

Hepatic disorder

19.1

22.2

24.9

Anemia

3.0

6.2

1.6

Neutropenia

2.4

6.4

4.3

Lymphopenia

3.2

4.0

0.2

CPK elevation

11.9

17.3

7.3

Renal dysfunction

0.2

0.2

0.0

ADA, adalimumab; AE, adverse event; CPK, creatine phosphokinase; GI, gastrointestinal; PY, patient years, UPA, upadacitinib.
*Adapted from McInnes, et al.1

  • Deaths were reported in
    • two patients in the upadacitinib 15 mg cohort (due to metastatic lung cancer and lower respiratory tract infection);
    • two patients in the upadacitinib 30 mg cohort (due to coronavirus infection and interstitial lung disease);
    • one patient in the adalimumab cohort (due to a traffic accident); and
    • one patient in the placebo group (due to an unspecified emergency while driving).

Conclusion

The 56-week efficacy data across various domains of PsA support the benefit of continued upadacitinib therapy in patients with active PsA and inadequate response or intolerance to ≥1 non-biological DMARD. Moreover, the 56-week safety data were comparable to findings from Week 24. However, a major limitation of this trial includes the lack of axial imaging to assess for psoriatic spondylitis. As the diagnosis was made on presumptive criteria, patients without true spondylitis may have been included. Furthermore, the study was not powered or designed to include a prespecified statistical comparison for efficacy between the upadacitinib arms and adalimumab arm through to Week 56.

Despite its limitations, the results of this trial along with those of the SELECT-PSA 2 trial lead the National Institute for Health and Care Excellence (NICE) to recommend upadacitinib alone or in combination with methotrexate as a treatment option for adult patients with active PsA whose disease has not responded well enough to DMARDs or those that cannot tolerate them. This recommendation was published earlier this year and is only for patients who have peripheral arthritis with ≥3 tender joints and ≥3 swollen joints and

  • have had two conventional DMARDs and ≥1 biological DMARD; or
  • where TNF-alpha inhibitors are contraindicated.2

  1. McInnes IB, Kato K, Magrey M, et al. Upadacitinib in patients with psoriatic arthritis and an inadequate response to non-biological therapy: 56-week data from the phase 3 SELECT-PsA 1 study. RMD Open. 2021;7(3):e001838. DOI: 1136/rmdopen-2021-001838.
  2.  Upadacitinib for treating active psoriatic arthritis after inadequate response to DMARDs. https://www.nice.org.uk/guidance/ta768/chapter/1-Recommendations. Published Feb 2, 2022. Accessed Apr 22, 2022.
  3. ClinicalTrials.gov. A study comparing upadacitinib (ABT-494) to placebo and to adalimumab in participants with psoriatic arthritis who have an inadequate response to at least one non-biologic disease modifying anti-rheumatic drug (DMARD) (SELECT - PsA 1). https://www.clinicaltrials.gov/ct2/show/NCT03104400. Updated Jan 25, 2022. Accessed May 30, 2022.
  4. McInnes IB, Anderson JK, Magrey M, et al. Trial of upadacitinib and adalimumab for psoriatic arthritis. N Engl J Med. 2021;384(13):1227-1239. DOI: 1056/NEJMoa2022516

Newsletter

Subscribe to get the best content related to Psoriasis and Psoriatic Arthritis delivered to your inbox