43
The TH17 Cell, IL23, and IL17 in PsA, SpA, and Psoriasis: Pathobiology and Therapeutic Targeting Philip Mease MD Director, Rheumatology Research, Swedish Medical Center Clinical Professor, University of Washington School of Medicine Seattle, WA

The TH17 Cell, IL23, and IL17 in PsA, SpA, and Psoriasis: Pathobiology and Therapeutic ...nwrsmeeting.org/wp-content/uploads/2017/04/Mease-N… ·  · 2017-12-07PsA, SpA, and Psoriasis:

  • Upload
    voliem

  • View
    217

  • Download
    0

Embed Size (px)

Citation preview

The TH17 Cell, IL23, and IL17 in PsA, SpA, and Psoriasis:

Pathobiology and Therapeutic Targeting

Philip Mease MD

Director, Rheumatology Research, Swedish Medical Center

Clinical Professor, University of Washington School of Medicine

Seattle, WA

DisclosuresResearch grants, consultation fees, and/or speaker honoraria: Abbvie, Amgen, Boehringer Ingelheim,

Bristol Myers, Celgene, Crescendo, Genentech, Janssen, Lilly, Merck, Novartis, Pfizer, SUN, UCB

Executive board GRAPPA, member of ASAS, SPARTAN, Scientific Director of CORRONA PsA/SpA registry,

Co-chair OMERACT PsA working groupCo-chair National Psoriasis Foundation PsA Task Force

ICHOM Inflammatory Arthritis Steering Group

Research and Education Association Memberships

Acknowledgments: Pierre Miossec for use of TH17/IL17 pathophysiology slides

• 1995: Discovery of IL-17 in human

• 2005: Identification of Th17 cells in mouse 3

T Cell Differentiation Pathways4

Autocrine Signaling Through IL-21 Promotes

the Development of TH17 Cells

5

RAR, retinoic acid receptor; ROR, RAR-related orphan receptor.

Permission granted from Palmer and Weaver.

Palmer MT, Weaver CT. Nature. 2007;448(7152):416-418.

Dendritic

cell

Naive precursor

T cell

TH17 cell

TGF-β

IL-6

Pathogens

IL-21

IL-23

IL-21

IL-23R

ROR-ɣ ROR-ɣ

Th17 cells produce multiple cytokines

Qu N, et al. Clin Devel Immunol. 2013:968549.

Under investigation

• Th17 cells• CD8+ T cells• γδ T cells• NK cells• NK T cells• LTi cells• Neutrophils

• Keratinocytes• Lung epithelial

cells• Colon epithelial

cells

Multiple cell types produce IL-17

Martin DA, et al. J Invest Dermatol. 2013;133:17-26.

• T cells• Intra-epithelial

lymphocytes• Lung epithelial cells• Cells of GI tract and

uterus • Eosinophils• Basophils• Mast cells• Alveolar

macrophagesIL-17E

(IL-25)

IL-17B

There are multiple IL-17R subtypes

Member Target Receptor Main Functions

IL-17A1,2 IL-17RA and IL-17RC

Neutrophil recruitment, host defense against extracellular pathogens, autoimmune pathology

IL-17B1,2 IL-17RB Proinflammatory activity?

IL-17C1,2 IL-17RE Proinflammatory activity?

IL-17D1,2 Unknown Proinflammatory activity?

IL-17E1,2 IL-17RB and IL-17RA

Stimulates Th2 responsesSuppresses Th17 responses

IL-17F1,2 IL-17RA and IL-17RC

Neutrophil recruitment (?), host defense against extracellular pathogens (?), immunoinflammatorypathology (?)

IL-17A – IL-17F heterodimer2

N/A Autoimmune pathology (presumed), neutrophil recruitment, and immunity to extracellular pathogens

1. Girolomoni G, et al. Br J Dermatol. 2012;167:717-724.2. Gaffen SL. Nat Rev Immunol. 2009;9:556-567.

IL-17 receptor signaling • Binding of IL-17A and/or IL-17F to

the heterodimeric IL-17R leads to the recruitment of Act1 through homotypic interactions between SEFIR domains (red ovals in diagram)

• This in turn allows the incorporation of TRAF6 into the signaling complex and then 'downstream' activation of the NF-κB and mitogen-activated protein kinase pathways

Hunter CA. Nature Immunol. 2007;8:232-234.

Psoriasis Pathophysiology

Nestle FNEJM. 2009; 361:496-509

Role of IL-17 Signaling in Joint Inflammation and Matrix Destruction

MMPs, matrix metalloproteinases; RANKL, receptor activator of nuclear factor kappa-B ligand.Adapted with permission granted from Miossec and Kolls.

1. Miossec P, Kolls JK. Nat Rev Drug Discov. 2012;11(10):763-776. 2. Suzuki E et al. Autoimmun Rev. 2014;13(4-5):496-502.

TH17 cells

• Other T cells

• Mast cells

• Neutrophils

Endothelial cells

Fibroblasts

Macrophages or dendritic cells

Chondrocytes

RANKL Osteoclastogenesis

Nitricoxide

MMPs

IL-1, TNF-α, IL-6

IL-6, IL-8

IL-6, IL-8

Osteoblasts

Cartilage damage

Bone erosion

Inflammation

Thrombosis

IL-17

Cutaneous lesionsKeratinocytes

Synergy Between TNF and IL-17A and F on IL-6 Expression and Production by Synoviocytes1

IL-6 Protein

IL-6

Sec

reti

on

(n

g/m

L)

0

30

10

40

50

20

IL-17F+ TNFα

Control IL-17A IL-17F IL-17A+ TNFα

IL-6 mRNA

IL-6

Fo

ld I

nd

ucti

on

0

80

20

100

140

120

40

60

IL-17F+ TNFα

TNF α IL-17A IL-17F IL-17A+ TNFα

TNFα

TNFa = tumor necrosis factor alpha.

1. Zrioual S, et al. J Immunol. 2009;182:3112-3120. 12

IL-23 induces enthesitis and new

bone formation

• Sherlock J, et al. Nat Med 2012;18:1069-76.

CIA, collagen-induced arthritis

Passive transfer CIA IL-23 minicircle

IL-23 promotes enthesialinflammation and IL-22

osteoproliferationIL-23R+ CD3+CD4-CD8-ROR-gT+

IL-23 and resident T-cells promote

enthesitis and osteoproliferation

Lories R and McInnes I. Nat Med 2012;18:1018–9.

IL-17 in a mouse model of ankylosing spondylitis (AS)

Abe Y, et al. Mod Rheumatol. 2009;19:316-322.

Unaffected Affected Male (BXSB x NZB) F1

mice develop seronegative ankylosing enthesitis in ankle/tarsal joints only when caged in groups, with the incidence reaching 83% at 7 months of age

Elevated popliteal lymph node T cells producing IL-17 and IFN-γ are significantly associated with joint ankylosis

IL-17 expressing CD4+ cells are increased in peripheral blood of patients with (AS) or psoriatic arthritis (PsA)

• Peripheral blood was obtained from 10 PsA, 10 AS, 10 rheumatoid arthritis (RA), and 5 vitiligo (Vit) patients, as well as from 25 healthy donors (HD)

• Peripheral blood cells were analyzed by flow cytometry and immunohistochemistry

• Cytokine production was examined by enzyme-linked immunosorbent assay and intracellular cytokine staining using specific monoclonal antibodies

Jandus C, et al. Arthritis Rheum. 2008;58:2307-2317.

*** P<0.001 vs healthy donors

IL-17 is elevated in cells from facet joints in patients with AS

In situ analysis of IL-17+ cells in facet joints of AS patients and patients with osteoarthritis (OA) was carried out using immunofluorescence microscopy

Double-staining revealed that myeloperoxidase-positive (MPO+) and CD15+ cells are the major source of IL-17 expression

The frequency of these cells was significantly higher in AS than in OA (P<0.05 in both cases). The population of MPO+ cells included mononuclear cells and cells with polysegmental nuclei

Th17 cells and mast cells are also a source for IL-17+ expression, both of which were significantly higher in AS patients than in OA patients (P<0.05 in both cases)

Appel H, et al. Arthritis Res Ther. 2011;13:R95.

Psoriatic Arthritis SynoviocytesExpress High Levels of IL-17RA

18

• A higher proportion of synoviocytes express IL-17RA in psoriatic arthritis than in non-inflammatory joint conditions1

• Synoviocytes in PsA express high levels of IL-17RA protein1

OA = osteoarthritis.Raychaudhuri SP, et al. Mol Cell Biochem. 2012;359:419-429.

20

15

10

5

0

%IL

-17

RA

+ C

ells

Psoriatic

Arthritis

Meniscal

Tear (MT)

P = 0.01

P = 0.01

Osteo-

arthritis

Inflammatory Noninflammatory

PsA MT OA

IL-17RA

a-Tubulin

Protein Expression (Western Blot)

Inflammatory Non-inflammatory

18

Expression of IL-17 is correlated with disease severity in patients with psoriatic arthritis

• Mononuclear cells from paired samples of synovial fluid (SF) and peripheral blood from patients with PsA were stimulated ex vivo, and T cells were examined by flow cytometry for cytokine expression,

• Clinical measures of arthritis activity were assessed for correlations with immunologic markers

Correlation between the frequency of IL-17 expressing CD4- T cells in PsA SF and clinical parameters of disease

Menon B, et al. Arthritis Rheumatol. 2014;66:1272-1281.

Recently Approved and Emerging Therapies for PsA or AS

• IL12-23i– Ustekinumab

• IL-17i– Secukinumab (IL17Ai)– Ixekizumab (IL17Ai)– Bimekizumab (IL17Ai and IL17Fi)

• Phospodiesterase 4 (PDE4)i (Poly-cytokine inhibition)– Apremilast

• JAK (Poly-cytokine inhibition)– Tofacitinib

• IL-23i– Tildrakizumab– Guselkumab– Risankizumab

• Dual inhibitors of TNF and IL17– ABT-122

• Co-stimulatory blockade– Abatacept

• IL-6i– Clazakizumab

Mease P. Ann Rheum Dis. 2011; 70 (Suppl 1) 77-84.

Ustekinumab: p40 inhibitor

Ustekinumab*Treatment response at Week 24

*p<0.001 Placebo (n=206) Ustekinumab 90 mg (n=204)Ustekinumab 45 mg (n=205)

60

40

20

0

Pat

ien

ts(%

)

22.8

*42.4

*49.5

ACR2060

40

20

8.7

*24.9

*27.9

ACR5060

40

20

0

2.4

*12.2

*14.2

ACR70

PSUMMIT I

McInnes IB et al. Lancet 2013; 382(9844);780-9.

0

Ustekinumab - EnthesitisMedian percent change from baseline in enthesitis scores at Week 24 and

Week 52

0

Me

dia

nch

ange

(%)

−40

−80

−100

0

−20

−60 p<0.001p=0.002

−42.9−50.0

Enthesitis scoring based on Modified MASES Index.Includes only randomized patients with enthesitis at baseline.

(n=148)(n=140)(n=137)0

−33.3

−48.3

0

p=0.008

p=0.098

(n=76)(n=72)(n=73)0

−87.5−83.3

−74.2

(n=142)(n=133)(n=122)

PSUMMIT I

1. McInnes IB et al. Lancet 2013; 382(9844);780-9.2. Ritchlin CT, et al. ACR/AHRP 2012, November 10-14, Washington, DC, USA. Abs 2557.

Week 241

PSUMMIT II

Week 242Week 521

PSUMMIT I and PSUMMIT II

Placebo→ustekinumab 45 mg Ustekinumab 90 mgUstekinumab 45 mgPlacebo

Ustekinumab - DactylitisMedian percent change from baseline in dactylitis scores at Week 24 and

Week 52

0

Me

dia

nch

ange

(%)

−40

−80

−100

0

−20

−60

−75.0*

0

−100 −100 −100

(n=91)(n=97)(n=86)

−70.8*

(n=95)(n=99)(n=92)0

0

−64.6

0

(n=41)(n=48)(n=38)

PSUMMIT I

Week 241

PSUMMIT II

Week 242Week 521

PSUMMIT I and PSUMMIT II

Placebo→ustekinumab 45 mg Ustekinumab 90 mgUstekinumab 45 mgPlacebo

*p<0.001

1. McInnes IB et al. Lancet 2013; 382(9844);780-9.2. Ritchlin CT, et al. ACR/AHRP 2012, November 10-14, Washington, DC, USA. Abs 2557.

EE & CO(PBO 45 mg)

UST

45 mg

UST

90 mg

Patients treated, n 189 205 204

Avg. duration of f/u, wks 29.8 50.4 50.2

Patients with AEs 78 (41.3%) 137 (66.8%) 132 (64.7%)

Patients with Infections 39 (20.6%) 77 (37.6%) 84 (41.2%)

Patients with SAEs 10 (5.3%) 12 (5.9%) 7 (3.4%)

Patients with ≥1 malignancy 0 0 0

Ustekinumab - SafetySafety summary through Week 52

PSUMMIT I

Through Week 52:

No cases of TB, opportunistic infections, or malignancies were reported

There were 3 MACE events: 2 MIs in PBO→45 mg group, 1 CVA in the UST 45 mg group, and no events in the

UST 90 mg group

McInnes IB et al. Lancet 2013; 382(9844);780-9:supplementary appendix

Copyright 2013 TREG Consultants LLC

• Open label trial – 20 pts with modified NY criteria AS and BASDAI >4

despite NSAIDs

– 90 mg USK at BL, Wk 4, Wk 16 – 1 endpoint: ASAS40 at Wk 24

Ustekinumab (IL-12/23i) in AS

75%65%

30%

55%

35%

0%

20%

40%

60%

80%

100%

ASAS20 ASAS40 ASAS PartialResponse

BASDAI50 ASDASInactiveDisease

% R

esponders

• CRP reduction in responders vs. non-responders

• 1 SAE (AS worsening), no SIEs

• ASAS40 response better than comparable OL trials MTX, ABA, RTX,

anakinra

These data suggest RCTs are indicated

Podubbnyy D, et al. ACR 2013, San Diego, #1798

IL-17 Inhibitors in PsA

Figure adapted from Strzepa A, et al. Pharmacol Reports. 2011;63(1):30-44, courtesy of Frank Nestle

Secukinumab (AIN457)Ixekizumab (LY2439821)

Brodalumab (AMG827)

Psoriasis/Psoriatic ArthritisIL-6

IL-1

TNF

TGFβIL-23

TH17

IL-17A

IL-17R

IL-22

IFN-γ

IL-17F

27

IL-17i: Secukinumab in Psoriasis

*P < 0.0001; ̂ P < 0.001 secukinumab vs. placebo; ‡P < 0.0001 secukinumab vs. etanercept.#P < 0.0001; †P < 0.001; § P < 0.01 secukinumab 300 mg vs. 150 mgMissing data were imputed as nonresponses

Langley RG, et al. NEJM 2014; 371:326

0

20

40

60

80

100

0 4 8 12 16 20 24 28 32 36 40 44 48 52

* †

~

PA

SI 7

5 R

espo

nse

0

20

40

60

80

100

0 4 8 12 16 20 24 28 32 36 40 44 48 52

Re

sp

on

de

rs (

%)

SEC 300 mg (n = 245)

SEC 150 mg (n = 243)

PBO (n = 246) SEC 300 mg (n = 323)

SEC 150 mg (n = 327)

PBO (n = 324)

ETN (n = 323)

ERASURE FIXTURE

ERASURE & FIXTURE

Re

sp

on

de

rs (

%) ‡

Week Week

28

IGA

0/1

Res

pons

e

0

20

40

60

80

100

0 4 8 12 16 20 24 28 32 36 40 44 48 52

#

0

20

40

60

80

100

0 4 8 12 16 20 24 28 32 36 40 44 48 52

* #

^

Re

sp

on

de

rs (

%)

Re

sp

on

de

rs (

%)

Secukinumab: ACR20 Response at Week 24:FUTURE 1and 2

0 4 248 12 16 201 2

Weeks

51.0%

29.3%

15.3%

3

54.0%

**

*

*

*

* *

*

*

*

*

*

‡ ‡

0 4 248 12 16 201 2

Weeks

50.0%

50.5%

17.3%

3

*

*

*

*

*

*

*

*

*

*

*

*

*

*

*

*

Secukinumab 300 mg s.c. (n=100)

Secukinumab 150 mg s.c. (n=100)

Secukinumab 75 mg s.c. (n=99)

Placebo (n=98)

Secukinumab IV-150 mg (n=202)

Secukinumab IV-75 mg (n=202)

Placebo (n=202)

Res

po

nd

ers

(%)

FUTURE 1 FUTURE 2

*P<0.0001; †P<0.001; §P<0.01; ‡P<0.05 vs. placebo;

(P-values at Week 24 adjusted for multiplicity);

Missing values imputed as nonresponse (non-responder imputation)

80

60

40

20

0

80

60

40

20

0

Mease P, et al. N Engl J Med. 2015;373:1329-39

McInnes IB et al. Lancet 2015;386:1137-46

45.5

27.3

15.2

29.7

18.9

10.8

14.7

5.9 5.9

14.3

8.6

0

0

20

40

60

80

58.2

38.8

22.4

63.5

44.4

27.0

36.9

24.6

6.2

15.9

6.31.6

0

20

40

60

80

Secukinumab 300 mg s.c. Secukinumab 75 mg s.c. PlaceboSecukinumab 150 mg s.c.

*P<0.0001; †P<0.001; §P<0.01; ‡P<0.05 vs. placebo;

Missing values were imputed as nonresponse (non-responder

imputation); Statistical analysis of ACR70 response in

anti–TNF-inadequate responders (anti–TNF-IR) in FUTURE 1

not performed due to lack of responders in placebo arm McInnes IB et al. Lancet 2015;386:1137-46

Secukinumab Efficacy in Anti–TNF-naïve and Anti–TNF-IR Patients (FUTURE 2)

Anti‒TNF-IR

ACR50 ACR70ACR20

33 37 34 35 3733 34 35 3733 34 35

Anti‒TNF-naïve

Res

po

nd

ers

(%)

ACR20 ACR50 ACR70

6367 63 65 6763 63 65 63 67 63 65

§

*

§

*

*

*

*†

§

56.550.0

30.3

14.8

48.242.2

32.4

21.5

Secukinumab: Improvements in Dactylitisand Enthesitis at Week 24 (FUTURE 2)

§P < 0.01; ‡P < 0.05 vs. placebo. Resolution of dactylitis and enthesitis amongst those subjects with these symptoms at baseline (dactylitis: n = 46 [300 mg], 32 [150 mg], 33 [75 mg] and 27 [placebo]; enthesitis: n = 56 [300 mg], 64 [150 mg], 68 [75 mg] and 65 [placebo]). Missing values were imputed as nonresponse (nonresponder imputation) at Week 24.

100

Perc

enta

ge o

f Su

bje

cts 80

60

0

100

Perc

enta

ge o

f Su

bje

cts 80

60

0

Week 24 Week 24

40

20

40

20

Resolution of Dactylitis Resolution of Enthesitis

§

§ §‡

Secukinumab 300 mg s.c. Secukinumab 75 mg s.c. PlaceboSecukinumab 150 mg s.c.

McInnes IB et al. Lancet 2015;386:1137-46

0.13

0.02

0.08

0.57

0.5

0.6

FUTURE 1: Mean Change from Baseline in mTSS at Week 24

‡P < 0.05 vs. placeboP-values for pooled secukinumab group adjusted for multiplicity of testing.

Mease P, et al. New Engl J Med 2015. 373;1329-1339

Secukinumab 10/mg/kg i.v. 75 mg s.c. (n = 202)

Placebo (n = 202)

Secukinumab 10/mg/kg i.v. 150 mg s.c. (n = 202)

Secukinumab pooled doses (n = 404)

mTSS

0.4

0.3

0.2

0Me

an

Change F

rom

Ba

se

line

in

mT

SS

0.1

Infections

• No cases of active tuberculosis (new or reactivation of latent infection); 1 opportunistic infection (300 mg)

• 11 cases of candidiasis; 6 oral (300 mg: 2; 150 mg: 3; 75 mg: 1), 1 esophageal (300 mg), 1 candida infection (300 mg), 4 vulvovaginal (300 mg: 1; 150 mg: 3)

• All mild or moderate, resolved spontaneously or with oral antifungal therapy; no relapses and all continued in study

Inflammatory Bowel Disease

• No reports of Crohn’s disease; 1 case of hemorrhagic diarrhea (300mg; resolved); 2 cases of ulcerative colitis were reported with secukinumab (300mg [resolved]); 150mg [did not resolve]); all patients continued in study

MACE

• One patient had an MI (75 mg); had history of sinus tachycardia and ongoing HTN and HLD; continued in the study

Neutropenia*

• Transient CTCAE grade 3 abnormality in 1 subject; no cases of CTCAE grade 4 neutropenia; no patients discontinued

• There was no increased risk of infection in patients with neutropenia

Treatment-emergent anti-secukinumab antibodies

• Treatment-emergent anti-secukinumab antibodies detected in 1 (0.2%) subject; no loss of efficacy was observed

FUTURE 2: ResultsAEs of Special Interest Through Week 52

Variable

Any SEC300 mg s.c.

(n = 145)

Any SEC150 mg s.c.

(n = 143)

Any SEC75 mg s.c.

(n = 99)Placebo (n = 98)

Exposure to treatment, mean days (SD) 411.7 (106.9) 130.6 (29.0)

*Three cases of squamous cell carcinoma were reported with secukinumab (two in the 75 mg group and one in the 150 mg group). Both cases in the 75 mg group resulted in discontinuation of study treatment.. CTCAE, Common Terminology Criteria for Adverse Events; HLD, hyperlipidemia; HTN, hypertension; MACE, mMI, myocardial infarction; SEC, secukinumab.McInnes IB, et al. Lancet 2015;386:1137–46. 33

Copyright 2014 TREG Consultants LLC

46%

21%

59%

29%

18%

6%

010203040506070

ASAS 20 ASAS 40

Secukinumab in AS - MEASURE 1 and 2:

TNFi-naïve vs TNFi-IR patients

66

49%

60%

34%33%

16%

010203040506070

ASAS20 ASAS40

SEC 150 mg

SEC 75 mg

PBO

Re

sp

on

de

rs (%

)

Re

sp

on

de

rs (%

)

MEASURE 1: TNFi-naïve (73% of pts)1 MEASURE 1: TNFi-IR (27% of pts)1

1. Baeten D, et al. ACR 2014, Boston, #819;

2. Sieper J, et al. Ibid, #536

* † †

§

Slightly lower efficacy of SEC in TNFi-IR population

n=92 n=90 n=89 n=92 n=90 n=89

*P<0.0001, †P<0.001, ‡P<0.01, §P<0.05 vs PBO

50%

25%25%18%

24%

0%

010203040506070

ASAS 20 ASAS 40

MEASURE 2: TNFi-naïve (62% of pts)2 MEASURE 2: TNFi-IR (38% of pts)2

68%

43%51%

31%31%

18%

010203040506070

ASAS20 ASAS40

Re

sp

on

de

rs (%

)

Re

sp

on

de

rs (%

)

n=44 n=45 n=45 n=44 n=45 n=45

§

n=28 n=28 n=29 n=28 n=28 n=29

§

§‡

n=33 n=34 n=33 n=33 n=34 n=33

Copyright 2015 TREG Consultants LLC

Radiographic progression in MEASURE 1

• 371 patients received SKB 75 mg or 150 mg for 2 years (pooled)

• Cervical/lumbar X-rays read for modified Stoke AS Spinal Score (mSASSS)

• In patients treated with continuous SKB (no placebo period), mean baseline

mSASSS 10.22, mean change 0.30 at 104 weeks; 80% no progression

• Greater change in males, baseline syndesmophytes, elevated CRP

Baraliakos X, et al. ACR 2015, San Francisco, #L6

Little change in radiographs over 2 years.

No controls for comparison

Change f

rom

baselin

e 5

Cumulative probability

–5

10

0

0 20 40 60 80 100

SKB 150 mg iv (n=86)

SKB 75 mg iv (n=82)

mSASSS, Week 104

Copyright 2016 TREG Consultants LLC

IBD in secukinumab (SEC)-treated patients

• 14 Phase 2 and 3 trials in

Ps (N=3430), PsA (N=974),

AS (N=571); >5200 pt-y exposure1

– All included patients with

unconfirmed prior history of,

but not active, IBD

• Reported AEs of IBD across

indications:

– UC 9 (4 flares, 5 new onset)

– CD 12 (6 flares, 6 new onset)

• No dose effect

• No pattern to time of onset

• Expected 15% flare rate IBD

1. Schreiber S et al. EULAR 2016, London, #OP0113; 2. Egeberg

A et al. Br J Dermatol 2016 Mar 9. doi: 10.1111/bjd.14528. [Epub

ahead of print]; 3. Braun et al. Arthritis Rheum 2007;57:639–47

New IBD infrequent in SEC trials. Exacerbates pre-existing disease?

AS

0.29 (0.06–0.84)

0.7 (0–3.7)

0.77 (0.33–1.51)

0.7 (0–3.7)

Crohn’s

disease3

SEC

program

Ulcerative

colitis3

SEC

program

0.2 0.80.4 0.60 1

0.14 (0.02–0.50)

0.11 (0.09–0.15)

0.07 (0–0.39)

0.06 (0.04–0.08)

Crohn’s

disease2

SEC

program

Ulcerative

colitis2

SEC

program

PsA

Exposure-adjusted incidence rate

(95% CI) per 100 patient-years

0.2 0.80.4 0.60 1

Copyright 2015 TREG Consultants LLC

-4

-2

0

2

4

6

8

0 20 40 60 80 100

mT

SS

ch

an

ge

fro

m B

L

Cumulative probability

10 6 3

54

3724

7156

43

8068

53

0

20

40

60

80

100

PASI 75 PASI 90 PASI 100

Re

sp

on

de

rs (

%)

3015

6

57

3926

58

40

23

6247

34

0

20

40

60

80

100

ACR20 ACR50 ACR70

Re

sp

on

de

rs (

%)

PASI responses at Week 24

Ixekizumab (IXE) with active control (ADA) in a

biologic-naïve PsA population

*P≤0.05; †P≤0.01; ‡P≤0.001 vs PBO; IXE loading dose of 160 mg.

ACR responses at Week 241,2

ΔmTSS PBOADA

40 mg

IXE 80 mg

q4w

IXE 80 mg

q2w

≤0 73 92† 83 83

≤0.5 78 96‡ 89* 95‡

≤0.95 84 96† 94* 97†

mTSS Week 24

patients with no progression, %

PBO (n=106)

ADA 40 mg (n=101) IXE 80 mg q4w (n=107)

IXE 80 mg q2w (n=103)

PBO (n=67)

ADA 40 mg (n=68) IXE 80 mg q4w (n=73)

IXE 80 mg q2w (n=59)

PBO (n=106)

ADA 40 mg (n=101)

IXE 80 mg q4w (n=107)

IXE 80 mg q2w (n=103)

‡ ‡ ‡

‡ ‡‡

‡ ‡

‡‡

1. Mease PJ, et al. ACR 2015, San Francisco, #977;

2. Mease PJ, et al. Ann Rheum Dis 2017. 76;79-87

Copyright 2015 TREG Consultants LLC

29 2649 4949 49

64 58

0

50

100

2819

35 33284347 39

0

50

100

Co

mp

lete

resp

on

de

rs (

%)

Ixekizumab (IXE) with active control (ADA) in a

biologic-naïve PsA population (cont.)

• Improvement in SF-36 PCS

and work productivity for all

treatment arms vs PBO2

1. Mease PJ, et al. ACR 2015, San Francisco, #977;

2. Mease PJ, et al. Ann Rheum Dis 2017. 76;79-87

39 3152 52

61 6765 70

0

50

100

b

Leeds Enthesitis Index1 Leeds Dactylitis Index-Basic1

HAQ-DI1,2

MC

ID (

%)

Baseline 2.9 3.0 2.7 3.1 2.9 3.0 2.7 3.1

Change –0.8 –0.8 –0.9 –1.5* –0.8 –0.9 –1.3 –1.4

3.5 5.8 4.0 3.8 3.5 5.8 4.0 3.8

–1.5 –2.2 –3.0† –3.2† –2.1 –3.3* –3.4† –3.5†

Week 12 Week 24

Baseline 1.15 1.12 1.24 1.17 1.15 1.12 1.24 1.17

Change –0.13 –0.35‡–0.37‡–0.47‡ –0.18 –0.37†–0.44‡–0.50‡

PBO (n=57)

ADA 40 mg (n=54) IXE 80 mg q4w (n=68)

IXE 80 mg q2w (n=57)

PBO (n=92)

ADA 40 mg (n=89) IXE 80 mg q4w (n=100)

IXE 80 mg q2w (n=90)

PBO (n=36)

ADA 40 mg (n=21) IXE 80 mg q4w (n=46)

IXE 80 mg q2w (n=37)

* ‡ ‡

* ‡‡

Week 12 Week 24 Week 12 Week 24

† ‡‡

*P≤0.05; †P≤0.01; ‡P≤0.001 vs PBO; IXE loading

dose of 160 mg. Patient ns for mean change: LEI:

PBO, n=57; ADA, n=56; IXE q4w, n=70; IXE q2w,

n=59. LDI: PBO, n=39; ADA, n=23; IXE q4w, n=54;

IXE q2w, n=41. HAQ-DI scores: PBO, n=106; ADA,

n=101; IXE q4w n=107; IXE q2w, n=103.

2716 33

Week52

Week12

Week24

4715 33

384 20

Brodalumab (IL-17ARi) in PsA: ACR 20 and 50 Responses Through 52 Weeks: Observed Analysis

39

= indicates time point at which all subjects began receiving 280 mg Q2W brodalumab

SE = standard error.

Brodalumab 280 mg Q2W (n = 56)PBO (n = 55) Brodalumab 140 mg Q2W (n = 57)

Mease P, et al. New England J Med. 2014. 370: 2295-306

ACR 50

Per

cen

tag

e (9

5% C

I) o

f S

ub

ject

s A

chie

vin

g A

CR

50

Week

100

0

20

30

40

50

60

90

80

70

10

242 4 8 12 16 52

280 mg Q2W

PBO

140 mg Q2W

4550 55 53 51 50 45

4654 53 51 52 50 42

4956 56 53 53 51 45

Number of Subjects

ACR 20

Per

cen

tag

e (9

5% C

I) o

f S

ub

ject

s A

chie

vin

g A

CR

20

Week

5019 44

Week52

Week12

Week24

7140 51

5644 64

100

0

20

30

40

50

60

90

80

70

10

242 4 8 12 16 52

280 mg Q2W

PBO

140 mg Q2W

4550 55 53 50 49 45

4654 52 51 52 49 42

4756 56 51 53 51 45

Number of Subjects

Copyright 2016 TREG Consultants LLC

Bimekizumab (IL-17Ai and IL-17Fi) proof-of-

concept study in PsA

Baeten D et al. EULAR 2016, London, #OP0108

• Monoclonal antibody directed against both IL-17A and IL-17F– in vitro human assay showed additive effects of IL-17A and IL-17F inhibition

• 4 doses at 0, 3, 6 weeks vs placebo– 40, 80, 160, 320 mg iv, with double dose at baseline

• Primary endpoint ACR-N at Week 8

ACR20 and ACR50 response rates (95% CI) at Week 8

Bimekizumab demonstrated high efficacy for skin and joints. No

major safety signals in this early proof-of-concept study in PsA

• 100% PASI 75 at Week 8

• No significant safety signals. 3 SAEs in 1 patient (related to a fall)

PBO

(n=12)

Bimekizumab top 3 doses

(n=30)

ACR20, % 17 (5–45) 80 (63–91)

ACR50, % 8 (2–35) 40 (25–58)

Copyright 2016 TREG Consultants LLC

ABT-122 in PsA: Week 12 Phase II data

• ABT-122 is a dual-variable-domain Ig inhibitor of TNF and IL-17A

Mease P et al. ACR 2016, Washington DC, #958

*P<0.05 vs placebo, †P<0.05 vs ADA

2513

4

68

38

15

65

3723

75

53

32

0

50

100

ACR20 ACR50 ACR70

Pa

tie

nts

(%

)Placebo (n=24)

ADA 40 mg eow(n=72)

ACR responders at

Week 12

**

*†

** *†

27

5874 78

0

50

100

PASI 75

Pa

tie

nts

(%

)

PASI 75

responders at

Week 12

* *†

-1.2

-2.8-2.8-2.5-3

-2

-1

0

Mean change frombaseline SPARCC

enthesitis index at Week12

Me

an

Δ, B

L to

Wk 1

2

SPARCC enthesitis

index

**

AEs, n (%)Placebo

(n=24)

ADA 40 mg

eow

(n=72)

ABT-122 120 mg

ew

(n=71)

ABT-122 240

mg ew

(n=71)

Any infection 5 (21) 20 (28) 14 (20) 15 (21)

Serious infection 0 0 0 0

Oral candidiasis 0 0 1 (1) 1 (1)

Nasopharyngitis 0 6 (8) 4 (6) 3 (4)

ABT-122 was highly effective, potentially superior to ADA for high-level

response in joints and skin. No difference from ADA in SIE

Copyright 2016 TREG Consultants LLC

Guselkumab (IL23i) in PsA: Week 24 Phase II data

• Human monoclonal Ab against P19 subunit of IL-

23

• DBRPCT: 149 pts active PsA despite SOC,

randomized

2:1 to GUS 100 mg sc vs PBO at 0, 4, then q8

wks

– 9% received prior TNFi

– 44% concomitant MTX

Deodhar A et al. ACR 2016, Washington DC, #4L

Guselkumab effective for all aspects of disease

Response at Week

24

• Significant improvements in

HAQ, MDA, other composite

endpoints

• No deaths, malignancies,

SIEs

p19

IL-2

3R

p40

IL-1

2R

β1

STAT3

P

STAT4

P

IL-23

p35

IL-1

2R

β2

p40

IL-1

2R

β1

STAT4

P

STAT4

P

IL-12

GUS

Teng MW et al. Nat Med 2015;21:719–29

1810

213

6 6

29

17

58

34

14

79

66

40

57 55

0

20

40

60

80

100

Re

sp

on

de

rs (

%) PBO GUS

USTUST

1° endpoint *P≤0.05, †P<0.01, ‡P<0.001 vs

PBO

*

*

‡‡

T Cell Differentiation Pathways43