You are on page 1of 17

Received: 19 November 2020 | Accepted: 24 November 2020

DOI: 10.1111/1346-8138.15727

REVIEW ARTICLE

Psoriasis: Recent progress in molecular-targeted therapies

Masaru Honma1,2 | Kei Hayashi2

Abstract
1
Department of Dermatology, Asahikawa
Medical University Hospital, Asahikawa,
Japan Psoriasis is a multifactorial recalcitrant inflammatory skin disease characterized by
2
International Medical Support Center, bothersome scaly reddish plaques especially on frequently chafed body parts, such as
Asahikawa Medical University Hospital,
the extensor sites of the extremities and scalp. Nonetheless, through recent advance in
Asahikawa, Japan
molecular-targeted therapies including biologics and small-molecule inhibitors, even the
Correspondence
severest symptoms of psoriasis and its comorbidities, such as psoriatic arthritis, can be
Masaru Honma, Department of
Dermatology, Asahikawa Medical excellently treated. The superb clinical effects lead to not only remarkable alleviation of
University Hospital, 2-1-1-1 Midorigaoka-
symptoms but also a deep understanding of patients’ impaired “quality of life” caused
Higashi, Asahikawa 078-8550, Japan.
Email: wanwan@asahikawa-med.ac.jp by this disease. Along with the development of novel treatment options targeting vari-
ous specific molecules, such as proinflammatory cytokines and signal transduction-as-
sociated molecules, clinicians have thoroughly understood the molecular mechanism of
psoriasis, and discovered that the IL-23/IL-17 axis mainly depending on Th17 cell function
is a crucial pathogenesis of this disease. Accumulation of knowledge about the working
mechanism and clinical effect of molecular-targeted therapies is indispensable for clini-
cians to establish a more refined therapeutic strategy for treating psoriasis.

KEYWORDS
biologics, cytokines, IL-17, IL-23, IL-36, JAK, molecular mechanism, PDE4, psoriasis, psoriatic
arthritis, RORγT, small-molecule inhibitors, TNF

1 | I NTRO D U C TI O N The molecular-targeted therapies can be classified into two


representative groups: biologics targeting cytokines and the
Psoriasis is a recurrent, persistent inflammatory skin disorder receptors involved in psoriasis pathomechanism, and small-molecule
­accompanied with various extracutaneous comorbidities, such as inhibitors targeting intracellular signaling molecules. Through the
psoriatic arthritis (PsA), uveitis, metabolic disorders, and cardio- development of novel therapeutic options focusing on specific mol-
vascular diseases.1-5 While the conventional therapies have been ecules, the detailed molecular mechanism of psoriasis can be deeply
insufficiently effective in controlling severe psoriasis symptoms, understood. Knowledge about the working mechanism and clinical
novel molecular-targeted therapies can generally treat such condi- effect of various molecular-targeted therapies is indispensable for
tion successfully. The successful clinical effects provide not only establishing a more adequate refined strategy for treating psoriasis.
remarkable mitigation of skin lesions and the related symptoms Hence, this review describes these systemic molecular-targeted
including various comorbidities but also a deep understanding of therapies including those agents under development highlighting
patients’ impaired “quality of life” and “unmet needs” on the treat- the working mechanisms and clinical effects.
ment for psoriasis.6-9 Psoriasis can induce and enhance systemic
inflammatory responses causing metabolic abnormality/cardiovas-
cular diseases called as “psoriatic march”10-13 and irreversible joint 2 | M O LEC U L A R M EC H A N I S M O F
destruction caused by PsA. Thus, these effective systemic thera- PSORIASIS
pies should be considered to minimize the risk of systemic visceral
involvements and permanent functional loss at the appropriate Given the excellent efficacy of the molecular-targeting therapies,
timing.14-17 the interleukin (IL)-23/IL-17 axis that mainly depends on Th17 cell

J Dermatol. 2021;48:761–777. wileyonlinelibrary.com/journal/jde


© 2021 Japanese Dermatological Association | 761
762 | HONMA and HAYASHI

function is crucial for psoriasis pathogenesis (Figure 1).1,4,18,19 recruits CCR6-expresssing lymphocytes, such as Th17 cells, into
In the initiation phase, plasmacytoid dendritic cells (DCs) that psoriatic skin lesions. 25,26 The CCL20/CCR6 axis is also indispen-
are activated by a complex of antimicrobial peptide (AMP) and sable for psoriasis pathomechanism. 27,28
self-nucleotide derived from injured keratinocytes via Toll-like
receptors (TLRs) release a large amount of interferon (IFN)-α,
which stimulates myeloid (conventional) DCs. 20,21 These acti- 3 | C Y TO K I N E-TA RG E TE D TH E R A PI E S
vated DCs produce tumor necrosis factor (TNF) and IL-23, which
both help maintain an immune response. TNF potentiates DCs Regarding the cytokine-targeted therapy, approximately 10 mon-
by themselves and accelerates the inflammatory response by oclonal antibodies are available for treating psoriasis, and these
1,22
various immune cells, whereas IL-23 significantly fortifies biologics target Th17-related cytokines such as TNF, IL-23, and
the pathogenic potential of Th17 cells, which are differentiated IL-17. TNF inhibitors can effectively treat plaque-type psoriasis,
from naive CD4 + T cells in the presence of TGF-β, IL-21, and IL- PsA, other inflammatory disorders including rheumatoid arthritis
6. 23,24 Furthermore, IL-17 and IL-22 released by Th17 and other (RA), inflammatory bowel disease (IBD), and Behcet’s disease. IL-23
cells that possess more innate characteristics, such as innate and IL-17 inhibitors demonstrate excellent clinical effects rela-
lymphoid cell (ILC)-3 and γδ-T cells, induce epidermal keratino- tively specific for psoriasis and its comorbidities (Table 1). In ad-
cyte hyperproliferation and enhance the production of inflam- dition to full-size immunoglobulin, several next-generation agents,
matory cytokines and chemokines, such as IL-17C, IL-8 (CXCL-8), such as affibodies and nanobodies, are currently being developed
and vascular endothelial growth factor (VEGF) from the epider- (Table 1). 29-31 These novel therapeutics are composed of consider-
1,4,18,19
mis. These factors contribute to the formation of psoriatic ably smaller proteins, such as a single domain of antibody and non-
epidermis phenotype and vicious inflammatory loop in the psori- immunoglobulin-based protein, with high affinity and specificity
atic lesion. Th17 cytokines and TNF independently and synergis- to the targeted molecules, and enabling a considerably higher
tically induce the expression of CCL20, which is a chemokine that bioavailability. 29-32

F I G U R E 1 Molecular mechanism of psoriasis. Plasmacytoid dendritic cells (DCs) activated by a complex of antimicrobial peptide (AMP)
and self-nucleotide release a large amount of interferon (IFN)-α, which stimulates myeloid DCs. Tumor necrosis factor (TNF) potentiates
DCs by themselves and accelerates the inflammatory response of various immune cells. Interleukin (IL)-23 is a key factor fortifying the
pathogenic potential of Th17 cells differentiated depending on transforming growth factor (TGF)-β, IL-21, and IL-6. IL-17 and IL-22 released
by Th17 and the other cells possessing more innate characteristics (e.g. innate lymphoid cell [ILC]-3 and γδ-T cells) induce epidermal
keratinocyte hyperproliferation. IL-17 also enhances the production of inflammatory cytokines and chemokines from the epidermis
synergistically with TNF. The accelerated inflammatory response hardens the vicious loop in the molecular mechanism of psoriasis [Color
figure can be viewed at wileyonlinelibrary.com]
TA B L E 1 Molecular-targeted therapies for the treatment of psoriasis and other inflammatory disorders (approved agent in the US and in Japan)

FDA approval (approval in Japan)

Target Behcet
HONMA and HAYASHI

Classification molecule Agent Structure Psoriasis PsA AS RA CD UC uveitis D HS PG PPP

TNF-inhibitor TNF Infliximab chimeric IgG1 + (+) + (+) + (+) + (+) + (+) + (+) + (+) + (+) - - -
Adalimumab human IgG1 + (+) + (+) + (+) + (+) + (+) + (+) + (+) - (+) + (+) applied -
certolizumab PEGylated, humanized + (+) + (+) + (-) + (+) + (-) - - - - - -
pegol Fab fragment
Etanercept fusion protein of + (-) + (-) + (-) + (+) - - - - - - -
TNFR and IgG1
IL-17 inhibitor IL-17A Secukinumab human IgG1κ + (+) + (+) + (+) - - - - - Phase 3 - -
Ixekizumab humanized IgG4 + (+) + (+) + (+) - - - - - - - -
ABY-035 affibody Phase 2 - - - - - - - - - -
netakimab humanized IgG1 Phase 3 Phase 3 Phase - - - - - - - -
(BCD-085) 3
vunakizumab humanized IgG1k Phase 2 - - - - - - - - - -
(SHR-1314)
IL-17A/F bimekizumab humanized IgG1 Phase 3 Phase 3 Phase - - - - - Phase 3 - -
(UCB4940) 3
sonelokinab bispecific nanobody Phase 2b - - - - - - - - - -
(M1095)
IL-17RA Brodalumab human IgG2 + (+) - (+) - (+) - - - - - - - Phase 3
IL-12/23 inhibitor IL-12/23 p40 Ustekinumab human IgG1 + (+) + (+) - - + (+) + (+) - - - - -
subunit
IL-23 p19 inhibitor IL-23 p19 Guselkumab human IgG1 + (+) + (+) - - Phase 2/3 Phase - - - - - (+)
subunit 2/3
Risankizumab humanized IgG1 + (+) - (+) - - Phase 3 Phase 3 - - Phase 2 - Phase 3
Tildrakizumab humanized IgG1 + (+) - - - - - - - - - -
mirikizumab humanized IgG4 Phase 3 - - - Phase 3 Phase 3 - - - - -
(LY3074828)
IL-36 inhibitor IL-36 spesolimab (BI humanized IgG1 - - - - - Phase - - - - Phase 2
receptor 655130)b 2/3
imsidolimab humanized Ig - - - - - - - - - - Phase 2
(ANB019)b
|
763

(Continues)
764
|

TA B L E 1 (Continued)

FDA approval (approval in Japan)

Target Behcet
Classification molecule Agent Structure Psoriasis PsA AS RA CD UC uveitis D HS PG PPP
a
PDE4 inhibitor PDE4 Apremilast small molecule + (+) + (+) - - - - - + (+) - - -
Roflumilast small molecule Phase 2 - - - - - - - - - -
Hemay005 small molecule Phase 2 - - - - - - - - - -
JAK inhibitor JAK1/3 Tofacitinib small molecule - + (-) - + (+) - + (+) - - - -
JAK1/2 Baricitinib small molecule - - - + (+) - - - - - - -
JAK1 upadacitinib small molecule - applied Phase + (+) Phase 3 Phase 3 - - Phase 2 - -
(ABT-494) 3
JAK1 Filgotinib small molecule - Phase 3 Phase applied Phase 3 Phase 3 - - - - -
3
TYK2 BMS-986165 small molecule Phase 3 - - - Phase 3 Phase 3 - - - - -
TYK2 PF-06826647 small molecule Phase 2 - - - - - - - - - -
TYK2/JAK1 brepocitinib small molecule Phase 2 - - - - - - - - - -
(PF-
06700841-
00)
Other inhibitors RORgT AUR101 small molecule Phase 2 - - - - - - - - - -
RORgT BI 730357 small molecule Phase 2 - - - - - - - - - -
IRAK-4 CF101 small molecule Phase 3 - Phase Phase - - Phase - - - -
3 3 2

Notes: Indication of each agent is described according to the label information in the US (https://daily​med.nlm.nih.gov/daily​med/) and Japan (https://www.info.pmda.go.jp/psear​ch/html/menu_tenpu_base.
html). Information of clinical trials (phase 2 or 3) is described according to the data at the site of database (https://clini​c altr​ials.gov/).
Green: agents approved in both the US and Japan. Light blue: agents approved in the US but not in Japan. Yellow: agents approved in Japan but not in the US. Orange: agents in clinical trials.
Abbreviations: PsA, psoriatic arthritis; AS, ankylosing spondylitis; RA, rheumatoid arthritis; CD, Crohn’s disease; UC, ulcerative colitis; Behcet D, Behcet’s disease; HS, hidradenitis suppurativa; PG,
pyoderma gangrenosum; PPP, palmoplantar pustulosis; applied, applied for approval.
a
Indication for recalcitrant oral ulcers in Behcet D.
b
Phase 2 studies evaluating efficacy and safety for GPP treatment are ongoing.
HONMA and HAYASHI
HONMA and HAYASHI | 765

3.1 | TNF and the inhibitors and PASI90 were 91% (placebo 23%), 80% (placebo 7%), and 57%
(placebo 1%) at week 10, respectively.46 In the IMPACT2 study, in-
While TNF is mainly produced by DCs, it is also manufactured by fliximab was also highly effective for treating PsA; at week 24, the
other cells, such as Th1, Th17, macrophages, mast cells, neutrophils, ACR20, ACR50, and ACR70 were 54% (placebo 16%), 41% (placebo
keratinocytes, and endothelial cells. 2,21,33,34 TNF acts as a proin- 4%), and 27% (placebo 2%), respectively.47 After infliximab therapy,
flammatory cytokine via NF-κB signaling activation through TNF the modified Sharp/van der Heijde score (modified total Sharp score
receptors, which are broadly expressed on various cell types. 22 [mTSS]) including the evaluation of radiographic progression is sig-
The TNF-mediated response is shared by inflammatory diseases nificantly reduced.48 Given the chimeric structure, antidrug antibody
and several conditions, such as RA, spondyloarthritis, IBD, uveitis, (ADA) against infliximab can be more frequently detected and affect
22
Behcet’s disease, and severe drug eruption (Table 1). TNF is also the clinical efficacy than that against other biologics.49 Nevertheless,
indispensable for granuloma-formation and the maintenance against MTX coadministration can diminish ADA frequency, and contribute
Mycobacterium tuberculosis infection.35-37 The reactivation risk of a higher drug survival rate of infliximab.50,51 Considering that the
tuberculosis is possibly lower during IL-17 or IL-23 inhibitor therapy trough level of infliximab with the clinical effect against RA52 and
than during TNF inhibitor therapy.38 psoriasis,53 the dose increment of infliximab of up to 6 mg/kg every
4 weeks or 10 mg/kg every 8 weeks (approved only in Japan) can
be used for psoriasis cases with the loss of treatment efficacy.54
3.1.1 | Etanercept Monitoring of the trough level of infliximab and ADA may contrib-
ute to making proper decisions for further therapeutic options.55
Etanercept, which is a fusion protein of TNF-receptor and the Fc Unfortunately, the mechanism of the infusion reaction, which refers
region of IgG1, can bind to soluble and membrane-bound TNF and to the relatively infliximab-specific adverse reaction among various
neutralize the function.39 Although the Fc region has a complement- biologics used for psoriasis treatment, remains insufficiently under-
binding domain, etanercept does not display antibody-dependent stood; however, it may possibly involve an immunological reaction
cellular cytotoxicity.39 The United States (US) and European Union via the ADA and cytokine-release syndrome.56,57
have approved the use of etanercept for treating RA, juvenile RA,
ankylosing spondylitis (AS), psoriasis, and PsA; however, Japan has
not yet approved this drug for treating psoriasis and PsA (Table 1). 3.1.3 | Adalimumab
Etanercept alone (50 mg/week) has shown preferable efficacy
against plaque-type psoriasis, and the improvement ratios in the pso- Adalimumab is a human IgG1-type monoclonal antibody neutralizing
riasis-area and severity index (PASI) by 50%, 70%, and 90% (PASI50, human TNF.58 Similar to infliximab, adalimumab specifically binds
PASI70, and PASI90) were 58% (placebo 14%), 34% (placebo 4%), to both soluble and membrane-bound TNF-α, inducing apoptosis of
and 12% (placebo 1%), respectively.40 Etanercept monotherapy is activated T cells and macrophages.44,45 It is broadly used for treat-
comparative with the combination therapy of etanercept and meth- ing RA, plaque-type psoriasis, PsA, AS, IBD, uveitis, and Behcet’s
otrexate (MTX), but it is more effective in treating PsA than MTX disease. In a randomized, double-blinded, placebo-controlled, and
monotherapy. The American College of Rheumatology improvement multicenter phase 3 study, adalimumab monotherapy (80, 40, and
ratio by 20%, 50%, and 70% (ACR20, ACR50, and ACR70) is 60.9%, 40 mg at week 0, 1, and then every other week [eow], respectively)
44.4%, and 29.2%, respectively.41 Regardless of MTX coadministra- was highly effective against moderate-to-severe plaque-type pso-
tion, the reproducible effect of etanercept reflects considerably riasis; the ratios of PASI75, PASI90, and PASI100 at week 12 were
42
lower immunogenicity than that of other TNF inhibitors. 68% (placebo 5%), 37% (placebo 2%), and 14% (placebo < 1%), re-
spectively.58 In a double-blind and placebo-controlled phase 2/3
trial focusing on the treatment of adult Japanese psoriatic cases,
3.1.2 | Infliximab the PASI50, PASI75, and PASI90 ratios at week 16 were 81.4% (pla-
cebo 19.6%), 62.8% (placebo 4.3%), and 39.5% (placebo 0%) in the
Infliximab is a chimeric anti-TNF-α monoclonal antibody composed group treated with 40 mg eow of adalimumab subcutaneously, and
of a constant region of human IgGlκ and the murine variable anti- 90.5%, 81.0%, and 61.9% in the group treated with 80 mg eow of
gen-binding region of high-affinity anti-human TNF-α antibody.43 adalimumab, respectively.59 Adalimumab effectively treats PsA,
It specifically binds to both soluble and membrane-bound TNF, and the ratios achieving ACR20, ACR50, and ACR70 at week 24
and a complex of membrane-associated TNF can induce apoptosis are 57% (placebo 15%), 39% (placebo 6%), and 23% (placebo 1%),
of activated T cells and macrophages.44,45 Intravenous infliximab is respectively.60 In addition, adalimumab could inhibit radiographic
broadly approved for treating RA, psoriasis, PsA, AS, IBD, uveitis, progression evaluated by mTSS.60 During adalimumab therapy, ADA
and Behcet’s disease. In a multicenter, double-blind, and placebo- frequency is relatively increased, affecting the clinical efficacy of
controlled phase 3 trial, infliximab administration (5 mg/kg at weeks this drug61 similar to infliximab. Therefore, for cases with secondary
0, 2, and 6, then every 8 weeks) was excellently effective against loss of treatment efficacy, increasing dose to 80 mg eow or applying
moderate-to-severe plaque-type psoriasis, and the PASI50, PASI75, MTX coadministration can be the therapeutic options.50,62
766 | HONMA and HAYASHI

3.1.4 | Certolizumab pegol homodimers of each subtype or a heterodimer of IL-17A and IL-
17F; the IL-17-signaling is transmitted via ligand-specific IL-17
Certolizumab pegol (CZP), which is a PEGylated, humanized Fab fragment receptors (IL-17R) (Figure 2a), whereas IL-17A signaling can be
neutralizing human TNF,63 lacks the Fc portion of immunoglobulin, which transmitted via IL-17RA, IL-17RC, and IL-17RD. 68-71
IL-17 recep-
64
plays an essential role in transplacental translocation. In contrast to inf- tors share a similar expression of fibroblast growth factor and
liximab and adalimumab, it does not induce apoptosis of activated T cells IL-17R (SEFIR) domain, which is an intracellular domain recruiting
and macrophages.44 According to CIMPASI-1 and CIMPASI-2 studies fo- Act-1; Act-1 activates NF-κB and mitogen-activated protein kinase
cusing on CZP efficacy in treating plaque-type psoriasis, the PASI75 and (MAPK) pathways (Figure 2b). 68-70 IL-17 is important for prevent-
PASI90 ratios at week 16 were 76.7% (placebo 9.9%) and 45.9% (placebo ing the cutaneous and mucosal infection of Candida albicans and
4.3%) in the group treated with 200 mg eow of CZP subcutaneously, while Staphylococcus aureus, 68-70,72 and maintaining the epithelial bar-
82.0% and 52.2% in the group treated by 400 mg eow of CZP, respec- rier formation of the intestine.73,74 Among the subtypes, IL-17A
63
tively. The clinical response of the CZP-treated group against PsA was is the most essentially implicated in healthy and disease condi-
significantly higher than that of the placebo group at week 24 (RAPID- tions including psoriasis69 and is produced by Th17, Tc17, tissue-
PsA study).65 Additionally, the response ratios of groups treated with 200 resident memory T (T RM), ILC3, invariant natural killer T (iNKT),
and 400 mg eow of CZP are were 63.8% and 56.3% (placebo 23.5%) in γδ-T, and mucosal associated invariant T (MAIT) cells.75 Fatty acid-
ACR20, 44.2% and 40.0% (placebo 12.5%) in ACR50, and 28.3 and 23.7% dependent, long-lived CD8-positive T RM is present even in healed
(placebo 4.4%) in ACR70, respectively.65 As observed in other TNF inhibi- epidermis, and IL-17-production from TRM is closely related to skin
tors, ADA can frequently be detected in CZP-treated cases, but the exist- lesion recurrence.76,77 In the synovial fluid of spondyloarthritis in-
66,67
ence of ADA is usually not associated with the trough level of CZP. cluding PsA, adaptive immunocytes, such as Th17 and Tc17 cells
can IL-23-dependently produce IL-17.75 Conversely, MAIT, iNKT,
and γδ-T cells can possibly produce IL-17 IL-23-independently.78,79
3.2 | IL-17 and the inhibitors IL-17A is involved in the progression of atherosclerosis, 80 liver cir-
rhosis, 81-83 and the mechanism of insulin resistance. 84 Generally,
The IL-17 family consists of six subunits (IL-17A, IL-17B, IL-17C, IL-17 inhibitors have lower immunogenicity than that of TNF in-
IL-17D, IL-17E, and IL-17F). Functional IL-17 is composed of hibitors, and the re-initiation of IL-17 inhibitor therapy shows a

F I G U R E 2 (a) Interleukin (IL)-17 family and the receptors. Signaling of IL-17 families are transmitted via specific receptor pairs. IL-17A-
signaling is transmitted via IL-17RA, IL-17RC, and IL-17RD. IL-17RA is shared by IL-17A, IL-17F, IL-17C, and IL-17E-signaling. (b) Molecular
structure of an IL-17 receptor (IL-17R). Two fibronectin III (FN)-like domains constitute the extracellular position, which is necessary for the
specific binding between ligands and receptors. A similar expression of fibroblast growth factor and IL-17R (SEFIR) domain is shared as an
intracytoplasmic region of the IL-17R family, recruiting Act1 which is an adaptor protein required to activate the IL-17-dependent signaling
pathways. Act1 also possesses the SEFIR-domain, which is required for the homophilic interaction with that of IL-17R. The cytoplasmic tail is
composed of the C/EBP-β activation domain (CBAD) [Color figure can be viewed at wileyonlinelibrary.com]
HONMA and HAYASHI | 767

reproducible effect in cases that had once achieved treatment inhibiting IL-17RA (Figure 2).68-71 In randomized phase 3 studies, the
success. 85-88 PASI75, PASI90, and PASI100 ratios at week 12 in the brodalumab-
treated group (210 mg eow) were 86% (placebo 8%), 70% (placebo
3%), and 44% (placebo 1%) in AMAGINE-2 study and 85% (placebo
3.2.1 | Secukinumab 6%), 52% (placebo 2%), and 37% (placebo 0.3%) in AMAGINE-3 study,
respectively.101 In a randomized phase 3 studies evaluating drug effi-
Secukinumab, which is a human IgG1κ-type monoclonal antibody against cacy against PsA (AMVISION-1 and AMVISION-2 trials), brodalumab
human IL-17A, has excellent clinical efficacy and the lowest immuno- showed comparable clinical effects with other IL-17A inhibitors.102
genicity among the biologics used for treating psoriasis, and other inflam- The ACR20, ACR50, and ACR70 response rates in the brodalumab-
89,90
matory disorders. While secukinumab shows an excellent effect on treated group at week 24 were 53.6% (placebo 23.8%), 36.4% (pla-
psoriasis, PsA and AS, it can possibly worsen IBD including Crohn’s dis- cebo 10.4%), and 20.9% (placebo 4.7%), respectively.102
91
ease. In randomized phase 3 studies, the ratios of PASI75, PASI90, and
PASI100 at week 12 were 81.6% (placebo 4.5%), 59.2% (placebo 1.2%),
and 28.6% (placebo 0.8%) in ERASURE study and 77.1% (placebo 4.9%), 3.2.4 | Bimekizumab (under development)
54.2% (placebo 1.5%), and 24.1% (placebo 0%) in FIXTURE study, respec-
tively.92 Head-to-head trials versus adalimumab in PsA revealed that the Bimekizumab is a monoclonal humanized IgG1 antibody that is still
clinical responses of secukimumab and adalimumab are comparable at currently being developed; this drug specifically neutralizes both
week 52 (response rates in the secukinumab- and adalimumab-treated IL-17A and IL-17F.103 While IL-17A and IL-17F present similar bioac-
93
groups: ACR20, 67% and 62%; ACR50, 49% and 42%, respectively). tivity, the affinity of IL-17A to IL-17RA is considerably higher than
In the combined evaluation of PASI90 and ACR50, the secukinumab- that of IL-17F.104 IL-17F is produced by lymphocytes as well as by
treated group showed a superior response rate to the adalimumab- the colonic epithelium.105 In a randomized, double-blinded and pla-
treated group.93 Secukinumab presents an excellent clinical effect in AS cebo-controlled phase 2b study (BE ABLE 1), the PASI75, PASI90,
treatment; the effect is evident even by administering a lower dose of and PASI100 ratios at week 12 in the bimekizumab-treated (320 mg
secukinumab compared with that in PsA treatment.94 every 4 weeks) group reached 93.0% (placebo 4.8%), 79.1% (placebo
0%), and 55.8% (placebo 0%), respectively.106 Bimekizumab was also
highly effective for active PsA in a phase 2b study (BE ACTIVE), and
3.2.2 | Ixekizumab the response rates of ACR20, ACR50, and ACR70 at week 24 in the
bimekizumab-treated (160 mg every 4 week with 320 mg loading
Ixekizumab is a humanized IgG4 antibody that lacks binding capacity dose at baseline) group were 78%, 62%, and 41%, respectively.107
to complement, and the amino acid in the hinge region is replaced to
prevent forming semi-antibodies and inadequate heterodimerized an-
tibodies.95 Its binding affinity to IL-17A is considerably stronger than 3.3 | IL-12/23 p40, IL-23 p19 and the inhibitors
that of secukinumab.95 In randomized phase 3 studies, the PASI75,
PASI90, and PASI100 ratios at week 12 in the ixekizumab-treated The IL-12 cytokine family consists of IL-12, IL-23, IL-27, and IL-35
108
group (160 and 80 mg at week 0 and then eow, respectively) were . These cytokines share subunits and the specific receptors
89.1% (placebo 3.9%), 70.9% (placebo 0.5%), and 35.3% (placebo 0.0%) (Figure 3).108,109 For instance, p40 subunit is shared by IL-12 and IL-23,
in UNCOVER-1 study and 87%, 68%, and 38% in UNCOVER-3 study.96 and p19 subunit is specifically used by IL-23 (Figure 3). In the psoriatic
Skin lesions improved earlier in the ixekizumab-treated group than in skin, the expression of both p40 and p19 subunits is increased but
the guselkumab-treated group in a head-to-head study (IXORA-R not that of p35 subunit, which is another component of IL-12, sug-
study).97 In a head-to-head trial comparing the clinical effects between gesting that IL-23 has a more specific effect in psoriatic skin.110 IL-12
ixekizumab and adalimumab against PsA, these drugs demonstrated and IL-23 a show distinct function in the T cell diversity. IL-12 mainly
similar efficacy at week 24 (response rates: ACR20 68.9% and 72.1%; works by inducing Th1 cells, whereas IL-23 mainly induces the patho-
ACR50 50.5% and 46.6%, respectively).98 In the combined evalua- genic potential of Th17 cells (Figure 3).108,109 Under IL-23 stimulation,
tion of PASI100 and ACR50, the ixekizumab-treated group showed FoxP3-positive regulatory T cells (Treg) are potentiated to produce
a superior response rate to the adalimumab-treated group.98 Similar IL-17.111,112 Similar to IL-17 inhibitors, IL-23 inhibitors have lower im-
to secukinumab, ixekizumab has excellent clinical efficacy against AS munogenicity than TNF-inhibitors, and the reproducible effect can be
symptoms, and its effect is comparable with that of adalimumab.99 observed on cases that had once achieved treatment success. 88,113

3.2.3 | Brodalumab 3.3.1 | Ustekinumab

Brodalumab, which is a monoclonal human IgG2 antibody against IL-17 Ustekinumab is a human IgG1 antibody neutralizing human IL-12/23
receptor A (IL-17RA),100 interrupts the signal of IL-17A, F, C, and E by p40 subunit.114,115 Thus, it inhibits both IL-12 and IL-23-dependent
768 | HONMA and HAYASHI

F I G U R E 3 Interleukin (IL)-12 family cytokines and the signaling via specific receptors. IL-12 family cytokines, such as IL-12, IL-23, and IL-
35, share the subcomponents with each other. While a p40 subunit is shared by IL-12 and IL-23, a p19 subunit is specific for IL-23. The signal
is intracellularly transmitted via the receptor-specific activation of the JAK/STAT pathways. IL-12 and IL-23 utilize the receptor pairs of IL-12
receptor β1 (IL-12Rβ1)/IL-12Rβ2 and IL-23R/ IL-12Rβ1, respectively. IL-35 can bind to the homodimer or heterodimer of IL-12Rβ2 and gp130.
IL-12, IL-23, and IL-35 mainly activate STAT4, STAT3, and STAT1/4, respectively. In terms of T cell differentiation, IL-12, IL-23, and IL-35-
signaling are closely related to the induction of Th1, Th17, and Treg, respectively [Color figure can be viewed at wileyonlinelibrary.com]

signaling. Ustekinumab presents a high clinical effect in treating 3.3.2 | Guselkumab


not only psoriasis and PsA but also IBD. Hence, IL-23-independent
IL-17A production can contribute to maintaining an intestinal epi- Guselukumab, which is a monoclonal human IgG1 antibody
73
thelial barrier. In randomized phase 3 studies evaluating the effi- specifically neutralizing IL-23 p19 subunit, specifically in-
cacy of ustekinumab in treating plaque-type psoriasis, the PASI75, hibits IL-23 signaling.113,120 It can effectively treat psoriasis,
PASI90, and PASI100 ratios at week 12 in the ustekinumab-treated PsA, and palmoplantar pustulosis.121 In addition, clinical tri-
(45 mg at week 0 and 4) group were 67.1% (placebo 3.1%), 41.6% als evaluating its efficacy in treating Crohn’s disease are still
(placebo 2.0%), and 12.5% (placebo 0.0%) in PHOENIX-1 study ongoing (Table 1). In randomized phase 3 studies evaluating
and 66.7% (placebo 3.7%), 42.3% (placebo 0.7%), and 18.1% (pla- the efficacy of guselkumab in treating moderate-to-severe
cebo 0.0%) in PHOENIX-2 study, respectively.114,115 In a real-world plaque-type psoriasis, the PASI75, PASI90, and PASI100 ratios
setting, ustekinumab tends to present a longer survival rate than at week 16 in the guselkumab-treated (100 mg at weeks 0 and
116-118
TNF inhibitors. Ustekinumab also shows favorable efficacy 4) group were 91.2% (placebo 5.7%), 73.3% (placebo 2.9%), and
in treating PsA, and the response ratios of ACR20, ACR50, and 37.4% (placebo 0.6%) in VOYAGE-1 study 120 and 86.3% (pla-
ACR70 at week 24 in the ustekinumab-treated (45 mg at weeks cebo 8.1%), 70.0% (placebo 2.4%), and 34.1% (placebo 0.8%)
0 and 4, then 45 mg every 12 weeks) group were 42.4% (pla- in VOYAGE-2 study, respectively.113 Guselkumab is also effec-
cebo 22.8%), 24.9% (placebo 8.7%), and 12.2% (placebo 2.2%) in tive in treating PsA, and the response ratios of ACR20, ACR50,
PHOENIX-1 study115 and 43.7% (placebo 20.2%), 17.5% (placebo and ACR70 at week 24 in the guselkumab-treated (100 mg at
6.7%), and 6.8% (placebo 2.9%) in PHOENIX-2 study, respec- weeks 0 and 4, then 100 mg every 8 weeks) group were 52%
tively.119 While increasing the dose up to 90 mg every 12 weeks (placebo 22.8%), 30% (placebo 9%), and 12% (placebo 6%) in
can be an effective therapeutic option for cases with insufficient DISCOVER-1 study.122 The clinical effect of guselkumab can
clinical effect or high body weight (≥100 kg), the approved condi- be reproduced independent of the previous treatment using
tion is different by country. biologics.122,123
HONMA and HAYASHI | 769

3.3.3 | Risankizumab weeks 0 and 8) group were 78% (placebo 4%), 59% (placebo 0%),
and 31% (placebo 0%) in the 100-mg mirikizumab-treated group
Risankizumab is a monoclonal humanized IgG1 antibody and 75%, 67%, and 31% in the 300-mg mirikizumab-treated group,
neutralizing IL-23 p19 subunit.124 In randomized, double-blind, respectively.130 According to a phase 2 study, mirikizumab can also
and placebo-controlled phase 3 studies evaluating the efficacy effectively treat ulcerative colitis.131
of risankizumab in treating plaque-type psoriasis, the response
rates of PASI90 and PAI100 in the risankizumab-treated (150 mg
at weeks 0 and week 4) group at week 16 were 75.3% (placebo 3.4 | IL-36 and the inhibitors
4.9%) and 35.9% (placebo 0%) in UltIMMA-1 study and 74.8%
(placebo 2.0%) and 50.9% (placebo 2.0%) in UltIMMA-2 study, Interleukin-36, which is a proinflammatory cytokine in the IL-1 fam-
respectively.124 Furthermore, risankizumab has a significantly ily, consists of three subtypes, namely, IL-36α, IL-36β, and IL-36 γ.
higher response rate than ustekinumab and adalimumab.124,125 In The IL-36 signal is transmitted through a heterodimer of IL-36 re-
another phase 3 study, risankizumab-treated cases maintained a ceptor (IL-36R)/IL-1RAcP, and activates MAPK and NF-κB pathways
sufficient effect on skin lesions even after withdrawing risanki- by recruiting MyD88 to the intracellular Toll/interleukine-1 resist-
zumab administration.126 In this study, the risankizumab-treated ance (TIR) domain, causing the inflammatory response of various
group (subcutaneous injection of 150 mg of risankizumab at tissues (Figure 4).132-135 In addition to the agonistic cytokines, IL-36
weeks 0, 4, and 16), which achieved static physician global as-
sessment score (sPGA) of 0/1 at week 28, was randomized into
a group regularly treated with risankizumab (150 mg every
12 weeks) or a group receiving placebo. The median time to loss
of PASI90 in the risankizumab-withdrawal group was 210 days
(interquartile range 113–296 days).126 Despite its excellent ef-
ficacy in treating psoriatic skin lesions, risankizumab failed to
show significant improvement of symptoms in AS cases.127 Thus,
IL-23-independent IL-17A production has a crucial role in axial
spondylitis pathomechanism.78,79

3.3.4 | Tildrakizumab

Tildrakizumab is a monoclonal humanized IgG1 antibody against


128
IL-23 p19 subunit. It is the most recently approved agent for
psoriasis treatment with relatively abundant data about its efficacy
and safety on long-term use.128 In randomized, double-blind, and
placebo-controlled phase 3 studies evaluating the efficacy of tild-
rakizumab in treating plaque-type psoriasis, the response rates of
PASI75, PASI90, and PASI100 in the tildrakizumab-treated (100 mg
at weeks 0 and 4) group at week 12 were 64% (placebo 6%), 35%
(placebo 3%), and 14% (placebo 1%) in reSURFACE-1 study and 61%
(placebo 6%), 39% (placebo 1%), and 12% (placebo 0%) in resurface-2
study, respectively.129 In most of cases, tildrakizumab efficacy was
maintained for 142 weeks.128

3.3.5 | Mirikizumab (under development) F I G U R E 4 Interleukin (IL)-36 and the receptors. IL-36 signaling
is transmitted by the receptor pair of IL-36R and IL-1Acp. The
Mirikizumab is a monoclonal humanized IgG4 antibody against extracellular region of the receptor consists of three Ig-like domains
human IL-23 p19 subunit; however, the clinical trials evaluating its necessary for the ligand-specific binding. The Toll/interleukine-1
efficacy and safety in treating psoriasis and IBD are still ongoing resistance (TIR) domain recruits the adaptor protein MyD88, which
interacts with the signaling molecules associated with downward
(Table 1).130 In a randomized, double-blind, and placebo-controlled
pathways, such as NF-κB and MAP kinase via the activation of
phase 2 study evaluating the efficacy of mirikizumab in treating the interleukin-1 receptor associated kinases (IRAKs).138,194 IL-36a
plaque-type psoriasis, the response rates of PASI75, PASI90, and works as a physiological competitive inhibitor against IL-36-
PAI100 at week 16 in the mirikizumab-treated (100 or 300 mg at signaling [Color figure can be viewed at wileyonlinelibrary.com]
770 | HONMA and HAYASHI

receptor antagonist (IL-36 Ra) is an intrinsic antagonistic cytokine

IFN-αR2
receptor

IFN-αR1/
that can bind to IL-36R. IL-36 family cytokines are produced not only

TYK-2
IFN-α

JAK-1
IFN-α
by immune cells such as macrophages, DCs, and T lymphocytes but
also epithelial tissues including the epidermis.132,136-138 Functional
deletion of IL-36Ra is one of the essential causes in generalized

receptor

IFN-γR2
IFN-γR1/
pustular psoriasis (GPP) (deficiency of interleukin thirty six receptor

IFN- γ

JAK-2
JAK-1
IFN-γ
antagonist, DITRA), and the uncontrolled IL-36 signaling causes an
139-141
accelerated inflammatory reaction in GPP. IL-17A can induce

homodimeric
IL-36-expression, and these cytokines synergistically amplify a vi-
cious circle of the inflammatory response. 136,137,142

CR

EPOR

JAK-2
EPO
3.4.1 | Spesolimab (BI 655130) (under development)

IL-6Rα/gp130
CR sharing
gp130

TYK-2
JAK-2
JAK-1
Spesolimab is a humanized monoclonal antibody against IL-36 recep-

IL-6
tor.143 In a phase 1 proof-of-concept study on seven GPP cases, a
single intravenous administration of spesolimab (10 mg/kg) signifi-

IL-4Rα/γ-
chain
cantly reduced the GPP severity score and maintained the status up
to week 20.144

IL-4

CR sharing γ-chain

β/γ-chain
IL-2Rα/IL-2R
4 | S M A LL M O LEC U LE S

JAK-3
JAK-1
IL-2
4.1 | Phosphodiesterase-4 (PDE-4) and the
inhibitors

IL-12Rβ1
TA B L E 2 Receptors and required JAKs in the signaling of representative cytokines and interferons

IL-23R/
Phosphodiesterases, which are hydrolytic enzymes of the cyclic CR sharing IL-12R β1
IL-23

nucleotides, cAMP and cGMP, consists of at least 11 subfamilies


based on the biochemical functions and homologies.145-147 In par-

IL-12Rβ2
ticular, the PDE-4 family works specifically on cAMP degradation
IL-12Rβ1/

TYK-2
and is expressed in the brain, cardiac muscle, smooth muscles, ke-

JAK-2
IL-12

ratinocytes, and immunocytes such as T lymphocytes, monocytes,


macrophages, neutrophils, DCs, and eosinophils.147 Protein kinase
IL22R1

A (PKA) activated by intracellular cAMP accumulation inhibits the


IL-10R2/

MAPK pathway associated with proinflammatory cytokine pro-


IL-22

duction and cell proliferation via Raf-1 inhibition.145-147 PKA also


suppresses transcriptional activity of NF-κB by regulating the in-
20R1 or
IL-22R1/IL-

teraction between NF-κB and CREB-binding protein (CBP)/p300


related to proinflammatory response (Figure 5).147 Conversely, PKA
R2
IL-20

accelerates the transcriptional activity of CREB/ATF-1 working on


anti-inflammatory-cytokine production by increasing CREB phos-
IL-20R2

phorylation (Figure 5).147 A competitive mechanism between CREB


IL-20R1/

IL, interleukin; JAK, Janus-activated kinase.

and NF-κB for CBP can be involved in transcriptional regulation.148


IL-19

IL-10 family receptor

EPACs, which are guanine nucleotide exchange factors specific for


Rap1/2, are activated by cAMP and can reduce the IL-6-dependent
inflammatory response via SOCS3 induction.149 EPAC1 is also indis-
IL-10R2
IL-10R1/

pensable for the adequate response of effector T cells against inhibi-


TYK-2
JAK-2
JAK-1
IL-10

tory cytokines derived from Treg.150 In addition to Treg-mediated


inhibitory mechanism via CD25-dependent IL-2 deprivation, inhibi-
tory cytokine production, and immune-checkpoint molecules,151,152
receptors

receptor pair
Cytokines

cAMP transfer can be an alternative suppression-mechanism of ef-


cytokine

(CR)

fector T cells by Tregs. As FoxP3, which is the master transcriptional


JAKs

factor of Treg, inhibits the expression of PDE3b to degrade cAMP,


HONMA and HAYASHI | 771

a high intracellular concentration of cAMP is maintained in Treg 33.1% (placebo 5.3%) and 9.8% (placebo 0.4%) in ESTEEM-1 study161
compared with other effector T cells.153,154 Treg transfers the ac- and 28.8% (placebo 5.8%) and 8.8% (1.5%) in ESTEEM-2 study, re-
cumulated cAMP into the target cells through gap junctions, and the spectively.162 In these clinical studies, apremilast displayed a favora-
153,154
elevated cAMP can inhibit activity of the target cells. PDE-4 ble clinical effect on nail and scalp psoriasis.163 In a retrospective
inhibition reduces proinflammatory-cytokine production from vari- observational study, apremilast was more effective in cases with
ous cell types including DCs, monocytes, neutrophiles, T-cells, and small-plaque psoriasis than in those with large-plaque psoriasis.164
147,155
synovial macrophages. In contrast, PDE-4 inhibition induces Apremilast is also effective in treating PsA; the response ratios of
the production of the anti-inflammatory cytokine IL-10 in mono- ACR20, ACR50, and ACR70 at week 16 in the apremilast-treated
cytes and increases the function of Tregs.147,155,156 During apremi- (30 mg twice daily) group were 32.1% (placebo 18.9%), 10.5% (pla-
last administration for the treatment of plaque-type psoriasis, the cebo 5%), and 1.2% (placebo 0.6%) in PALACE-2 study165 and 41%
157,158
plasma levels of IL-17A, IL-17F, and IL-22 significantly declined. (placebo 18%), 15% (placebo 8%), and 4% (placebo 2%) in PALACE-3
Moreover, clinical trials evaluating the efficacy of the topical or sys- study, respectively.166 Further, apremilast is considerably safe.
temic PDE-4 inhibitors, crisaborole and roflumilast, are currently However, it is contraindicated for pregnant women. Nevertheless,
159,160
ongoing. this drug does not always require regular screening for infectious
disorders, such as tuberculosis and hepatitis B virus infection.147
Headache and secretory diarrhea are the most common adverse
4.1.1 | Apremilast reactions of apremilast leading to discontinuation.167 Diarrhea is
induced by the pharmacological effect of PDE-4 inhibition, activat-
Apremilast is an oral, small-molecule, selective PDE-4 inhibitor.161 In ing chloride channel of mucosal epithelia depending on cAMP accu-
randomized, double-blind, and placebo-controlled phase 3 studies mulation.168 Apremilast also presents a favorable effect on the oral
evaluating the efficacy of apremilast in treating moderate-to-severe ulcer of Behcet’s disease,169 and clinical trials on several inflamma-
plaque-type psoriasis, the response rates of PASI75 and PASI90 in tory skin diseases including atopic dermatitis and hidradenitis sup-
the apremilast-treated (30 mg twice daily) group at week 16 were purativa, are ongoing (Table 1).

F I G U R E 5 Function of cAMP in an inflammatory response. Adenylate cyclase (AC) activated in the downstream of G-protein-coupled
receptors (GPCRs), such as β-adrenergic receptor and prostaglandin E2 receptors, increases intracellular cAMP specifically hydrolyzed
by phosphodiesterase 4 (PDE4). Accumulation of cAMP activates protein kinase A (PKA), resulting in anti-inflammatory-cytokine
production by increasing the transcriptional activity through CREB/ATF-1. Activated PKA inhibits the MAP-kinase pathway associated with
proinflammatory-cytokine production via Raf-1 inhibition, and it also suppresses the transcriptional activity of neuclear factor-kappa B (NF-
κB) by regulating the interaction between NF-κB and CBP/p300 related to the proinflammatory response in a competitive mechanism with
phosphorylated CREB by activated PKA. EPACs can reduce interleukin (IL)-6-dependent inflammatory response, and they are indispensable
for the adequate response of effector T cells against inhibitory cytokines derived from regulatory T cells [Color figure can be viewed at
wileyonlinelibrary.com]
772 | HONMA and HAYASHI

4.2 | Janus-activated kinase REFERENCES


(JAK) and the inhibitors 1. Armstrong AW, Read C. Pathophysiology, clinical presentation,
and treatment of psoriasis: a review. JAMA. 2020;323:1945–60.
2. Ogawa E, Sato Y, Minagawa A, Okuyama R. Pathogenesis of psori-
Most of cytokines and interferons transmit their signal via type I
asis and development of treatment. J Dermatol. 2018;45:264–72.
and II cytokine receptors lacking an enzymatic activity and JAKs, 3. Harden JL, Krueger JG, Bowcock AM. The immunogenetics of pso-
which are nonreceptor tyrosine kinases.170-179. JAK subsequently riasis: a comprehensive review. J Autoimmun. 2015;64:66–73.
phosphorylates and activates signal transducer and activators of 4. Boehncke WH, Schön MP. Psoriasis. Lancet. 2015;386:983–94.
5. Egeberg A, Gisondi P, Carrascosa JM, Warren RB, Mrowietz U.
transcription (STATs). The JAK family consists of JAK1, JAK2, JAK3,
The role of the interleukin-23/Th17 pathway in cardiometabolic
and TYK2; inhibition of each subtype can interrupt specific STAT- comorbidity associated with psoriasis. J Eur Acad Dermatology
dependent signaling pathways (Table 2).180-182 Tofacitinib, which Venereol. 2020;34:1695–706.
is an oral JAK1/3-selective inhibitor, has been approved for treat- 6. Conrad C, Gilliet M. Psoriasis: from Pathogenesis to Targeted
Therapies. Clin Rev Allergy Immunol. 2018;54:102–13.
ing plaque-type psoriasis and psoriatic arthritis in the US but not
7. Boehncke WH, Prinz J, Gottlieb AB. Biologic therapies for psoria-
in Japan. Although tofacitinib shows a favorable clinical effect on sis. A systematic review. J Rheumatol. 2006;33:1447–51.
plaque-type psoriasis and PsA symptoms,183-185 herpes zoster can 8. Salame N, Perez-Chada LM, Singh S, Callis Duffin K, Garg A,
occur during tafacitinib treatment, especially in Asian populations Gottlieb AB, et al. Are your patients satisfied a systematic re-
including the Japanese.186,187 Baricitinib, an oral JAK1/2-selective view of treatment satisfaction measures in psoriasis. Dermatology.
2018;234:157–65.
inhibitor, is effective for treating moderate-to-severe psoriasis,
9. Torii H, Sato N, Yoshinari T, Nakagawa H. Dramatic impact of a
and the response ratio of PASI75 in a phase 2b trial was 42.9% and Psoriasis Area and Severity Index 90 response on the quality of life
54.1% (placebo 16.7%) in 8 and 10 mg of baricitinib-treated groups, in patients with psoriasis: An analysis of Japanese clinical trials of
respectively.188 infliximab. J Dermatol. 2012;39:253–9.
10. Boehncke W, Boehncke S, Tobin A, Kirby B. The ‘psoriatic march’:
a concept of how severe psoriasis may drive cardiovascular comor-
bidity. Exp Dermatol. 2011;20:303–7.
4.3 | Others 11. Takeshita J, Grewal S. Psoriasis and comorbid diseases. J Am Acad
Dermatol. 2017;76:393–403.
12. Takahashi H, Iizuka H. Psoriasis and metabolic syndrome. J
Oral small molecule inhibitors targeting Tyk2,189 which is one of
Dermatol. 2012;39:212–8.
JAK families related to IL-23-signaling, and ROR γT,190 which is 13. Gelfand JM, Neimann AL, Shin DB, Wang X, Margolis DJ, Troxel
a master transcriptional factor of Th17 cells, is currently under AB. Risk of myocardial infarction in patients with psoriasis. JAMA.
development, especially for plaque-type psoriasis treatment. 2006;296:1735–41.
14. Haroon M, Gallagher P, FitzGerald O. Diagnostic delay of more
In addition, nuclear receptors, such as NURR1 (NR4a), which is
than 6 months contributes to poor radiographic and functional
a transcriptional factor associated with Treg differentiation,191 outcome in psoriatic arthritis. Ann Rheum Dis. 2015;74:1045–50.
and interleukin-1 receptor associated kinase 4 (IRAK-4), which 15. Gisondi P, Altomare G, Ayala F, Conti A, Dapavo P, De Simone C,
is a kinase mediating cytokine signaling via MyD88 associa- et al. Consensus on the management of patients with psoriatic ar-
thritis in a dermatology setting. J Eur Acad Dermatology Venereol.
tion, are focused as therapeutic targets of plaque-type psoriasis
2018;32:515–28.
(Table 1).192,193 16. Gossec L, Baraliakos X, Kerschbaumer A, de Wit M, McInnes I,
Dougados M, et al. EULAR recommendations for the management
of psoriatic arthritis with pharmacological therapies: 2019 update.
5 | CO N C LU S I O N Ann Rheum Dis. 2020;79:S700–12.
17. Coates LC, Kavanaugh A, Mease PJ, Soriano ER, Laura Acosta-
Felquer M, Armstrong AW, et al. Group for research and as-
The recent progress of highly potent therapeutic options for pso- sessment of psoriasis and psoriatic arthritis 2015 treatment
riasis treatment brings the era of “introducing and maintaining re- recommendations for psoriatic arthritis. Arthritis Rheumatol.
mission” in psoriasis cases. Simultaneously, the excellent effect of 2016;68:1060–71.
18. Nestle FO, Kaplan DH, Barker J. Psoriasis. N Engl J Med.
these molecular-targeted therapies explains the detailed molecular
2009;361:496–509.
mechanism of psoriatic diseases. However, further basic research 19. Brembilla NC, Senra L, Boehncke WH. The IL-17 family of cyto-
and refinement of the therapeutic strategy are required to clarify kines in psoriasis: IL-17A and beyond. Front Immunol. 2018;9:1682.
unmet needs on psoriasis treatment. 20. Takahashi T, Yamasaki K. Psoriasis and antimicrobial peptides. Int J
Mol Sci. 2020;21:6791.
21. Wang A, Bai YP. Dendritic cells: The driver of psoriasis. J Dermatol.
C O N FL I C T O F I N T E R E S T 2020;47:104–13.
None declared. 22. Kalliolias GD, Ivashkiv LB, Program TD. TNF biology, patho-
genic mechanisms and emerging therapeutic strategies. Nat Rev
Rheumatol. 2016;12:49–62.
ORCID
23. Yasuda K, Takeuchi Y, Hirota K. The pathogenicity of Th17 cells in
Masaru Honma https://orcid.org/0000-0003-1566-5331 autoimmune diseases. Semin Immunopathol. 2019;41:283–97.
HONMA and HAYASHI | 773

24. Schmitt N, Ueno H. Regulation of human helper T cell subset dif- interaction with anti-TNF agents. Rheumatol (United Kingdom).
ferentiation by cytokines. Curr Opin Immunol. 2015;34:130–6. 2010;49:1215–28.
25. Harper EG, Guo C, Rizzo H, Lillis JV, Kurtz SE, Skorcheva I, et al. 46. Reich K, Nestle FO, Papp K, Ortonne J-P, Evans R, Guzzo C, et al.
Th17 cytokines stimulate CCL20 expression in keratinocytes in Infliximab induction and maintenance therapy for moderate-to-se-
vitro and in vivo: Implications for psoriasis pathogenesis. J Invest vere psoriasis: A phase III, multicentre, double-blind trial. Lancet.
Dermatol. 2009;129:2175–83. 2005;366:1367–74.
26. Chiricozzi A, Guttman-Yassky E, Suárez-Farĩas M, Nograles KE, 47. Antoni C, Krueger GG, De Vlam K, Birbara C, Beutler A, Guzzo
Tian S, Cardinale I, et al. Integrative responses to IL-17 and TNF-α C, et al. Infliximab improves signs and symptoms of psori-
in human keratinocytes account for key inflammatory pathogenic atic arthritis: Results of the IMPACT 2 trial. Ann Rheum Dis.
circuits in psoriasis. J Invest Dermatol. 2011;131:677–87. 2005;64:1150–7.
27. Hedrick MN, Lonsdorf AS, Shirakawa AK, Lee C-CR, Liao F, Singh 48. Van Der Heijde D, Kavanaugh A, Gladman DD, Antoni C, Krueger
SP, et al. CCR6 is required for IL-23-induced psoriasis-like inflam- GG, Guzzo C, et al. Infliximab inhibits progression of radiographic
mation in mice. J Clin Invest. 2009;119:2317–29. damage in patients with active psoriatic arthritis through one year
28. Furue K, Ito T, Tsuji G, Nakahara T, Furue M. The CCL20 and CCR6 of treatment: Results from the induction and maintenance psori-
axis in psoriasis. Scand J Immunol. 2020;91:e12846. atic arthritis clinical trial 2. Arthritis Rheum. 2007;56:2698–707.
29. Svecova D, Lubell MW, Casset-Semanaz F, Mackenzie H, 49. Carrascosa JM, van Doorn MBA, Lahfa M, Nestle FO, Jullien D,
Grenningloh R, Krueger JG. A randomized, double-blind, pla- Prinz JC. Clinical relevance of immunogenicity of biologics in pso-
cebo-controlled phase 1 study of multiple ascending doses riasis: implications for treatment strategies. J Eur Acad Dermatology
of subcutaneous M1095, an anti–interleukin 17A/F nano- Venereol. 2014;28:1424–30.
body, in moderate-to-severe psoriasis. J Am Acad Dermatol. 50. Garcês S, Demengeot J, Benito-Garcia E. The immunogenicity of
2019;81:196–203. anti-TNF therapy in immune-mediated inflammatory diseases:
30. Löfblom J, Feldwisch J, Tolmachev V, , Carlsson J, Ståhl S, Frejd A systematic review of the literature with a meta-analysis. Ann
FY. Affibody molecules: Engineered proteins for therapeu- Rheum Dis. 2013;72:1947–55.
tic, diagnostic and biotechnological applications. FEBS Lett. 51. Magis Q, Jullien D, Gaudy-Marqueste C, Baumstark K, Viguier M,
2010;584:2670–80. Bachelez H, et al. Predictors of long-term drug survival for inflix-
31. Dumet C, Pottier J, Gouilleux V, Watier H. New structural for- imab in psoriasis. J Eur Acad Dermatology Venereol. 2017;31:96–101.
mats of therapeutic antibodies for rheumatology. Jt Bone Spine. 52. St. Clair EW, Wagner CL, Fasanmade AA, Wang B, Schaible T,
2018;85:47–52. Kavanaugh A, et al. The relationship of serum infliximab concen-
32. Bannas P, Hambach J, Koch-Nolte F. Nanobodies and Nanobody- trations to clinical improvement in rheumatoid arthritis: Results
Based Human Heavy Chain Antibodies As Antitumor Therapeutics. from ATTRACT, a multicenter, randomized, double-blind, place-
Front Immunol. 2017;8:1603. bo-controlled trial. Arthritis Rheum. 2002;46:1451–9.
33. Mylonas A, Conrad C. Psoriasis: classical vs. paradoxical. the yin- 53. Takahashi H, Tsuji H, Ishida-Yamamoto A, Iizuka H. Plasma trough
yang of TNF and Type i interferon. Front Immunol. 2018;9(2746). levels of adalimumab and infliximab in terms of clinical efficacy
34. Krueger G, Callis K. Potential of tumor necrosis factor inhibitors in during the treatment of psoriasis. J Dermatol. 2013;40:39–42.
psoriasis and psoriatic arthritis. Arch Dermatol. 2004;140:218–25. 54. Torii H, Nakano M, Yano T, Kondo K, Nakagawa H. Efficacy and
35. Lin PL, Plessner HL, Voitenok NN, Flynn JAL. Tumor necrosis factor safety of dose escalation of infliximab therapy in Japanese pa-
and tuberculosis. J Investig Dermatology Symp Proc. 2007;12:22–5. tients with psoriasis: Results of the SPREAD study. J Dermatol.
36. Ray JCJ, Flynn JL, Kirschner DE. Synergy between Individual 2017;44:552–9.
TNF-Dependent Functions Determines Granuloma Performance 55. Laine J, Sakari Jokiranta T, Eklund KK, Väkeväinen M, Puolakka K.
for Controlling Mycobacterium tuberculosis Infection. J Immunol. Cost-effectiveness of routine measuring of serum drug concentra-
2009;182:3706–17. tions and anti-drug antibodies in treatment of rheumatoid arthritis
37. Harris J, Keane J. How tumour necrosis factor blockers interfere patients with TNF-α blockers. Biol Targets Ther. 2016;10:67–73.
with tuberculosis immunity. Clin Exp Immunol. 2010;161:1–9. 56. Vultaggio A, Matucci A, Nencini F, Pratesi S, Parronchi P, Rossi
38. Nogueira M, Warren RB, Torres T. Risk of tuberculosis reactivation O, et al. Anti-infliximab IgE and non-IgE antibodies and induc-
with interleukin (IL)-17 and IL-23 inhibitors in psoriasis – time for a tion of infusion-related severe anaphylactic reactions. Allergy.
paradigm change. J Eur Acad Dermatology Venereol. 2020. https:// 2010;65:657–61.
doi.org/10.1111/jdv.16866 57. Lichtenstein L, Ron Y, Kivity S, Ben-Horin S, Israeli E, Fraser GM,
39. Spencer-Green G. Etanercept (Enbrel): Update on therapeutic use. et al. Infliximab-related infusion reactions: systematic review. J
Ann Rheum Dis. 2000;59:46–9. Crohns Colitis. 2015;9:806–15.
40. Leonardi CL, Powers JL, Matheson RT, Goffe BS, Zitnik R, Wang A, 58. Menter A, Tyring SK, Gordon K, Kimball AB, Leonardi CL, Langley
et al. Etanercept as monotherapy in patients with psoriasis. N Engl RG, et al. Adalimumab therapy for moderate to severe psoria-
J Med. 2003;349:2014–22. sis: a randomized, controlled phase III trial. J Am Acad Dermatol.
41. Mease PJ, Gladman DD, Collier DH, Ritchlin CT, Helliwell PS, Liu 2008;58:106–15.
L, et al. Etanercept and methotrexate as monotherapy or in com- 59. Asahina A, Nakagawa H, Etoh T, Ohtsuki M. Adalimumab in
bination for psoriatic arthritis: primary results from a randomized. Japanese patients with moderate to severe chronic plaque pso-
Controlled Phase III Trial. Arthritis Rheumatol. 2019;71:1112–24. riasis: Efficacy and safety results from a Phase II/III randomized
42. Emi Aikawa N, De Carvalho JF, Artur Almeida Silva C, Bonfá E. controlled study. J Dermatol. 2010;37:299–310.
Immunogenicity of anti-TNF-α agents in autoimmune diseases. 60. Mease PJ, Gladman DD, Ritchlin CT, Ruderman EM, Steinfeld SD,
Clin Rev Allergy Immunol. 2010;38:82–9. Choy EHS, et al. Adalimumab for the treatment of patients with
43. Winterfield LS, Menter A. Infliximab. Dermatol Ther. 2004;17: moderately to severely active psoriatic arthritis: Results of a dou-
409–26. ble-blind, randomized, placebo-controlled trial. Arthritis Rheum.
44. Chan AC, Carter PJ. Therapeutic antibodies for autoimmunity and 2005;52:3279–89.
inflammation. Nat Rev Immunol. 2010;10:301–16. 61. Mostafa NM, Nader AM, Noertersheuser P, Okun M, Awni
45. Horiuchi T, Mitoma H, Harashima S, Tsukamoto H, Shimoda WM. Impact of immunogenicity on pharmacokinetics, efficacy
T. Transmembrane TNF-alpha : structure, function and and safety of adalimumab in adult patients with moderate to
774 | HONMA and HAYASHI

severe chronic plaque psoriasis. J Eur Acad Dermatology Venereol. cells, which contribute to the Th17-axis in ankylosing spondylitis.
2017;31:490–7. Ann Rheum Dis. 2016;75:2124–32.
62. Asahina A, Ohtsuki M, Etoh T, Gu Y, Okun MM, Teixeira HD, 80. Gelfand JM, Shin DB, Duffin KC, Armstrong AW, Blauvelt A, Tyring
et al. Adalimumab treatment optimization for psoriasis: re- SK, et al. A randomized placebo-controlled trial of secukinumab on
sults of a long-term phase 2/3 Japanese study. J Dermatol. aortic vascular inflammation in moderate-to-severe plaque psoria-
2015;42:1042–52. sis (VIP-S). J Invest Dermatol. 2020;140:1784–93.e2.
63. Gottlieb AB, Blauvelt A, Thaçi D, Leonardi CL, Poulin Y, Drew J, 81. Gomes AL, Teijeiro A, Burén S, Tummala KS, Yilmaz M, Waisman
et al. Certolizumab pegol for the treatment of chronic plaque pso- A, et al. Metabolic inflammation-associated IL-17A causes non-al-
riasis: Results through 48 weeks from 2 phase 3, multicenter, ran- coholic steatohepatitis and hepatocellular carcinoma. Cancer Cell.
domized, double-blinded, placebo-controlled studies (CIMPASI-1 2016;30:161–75.
and CIMPASI-2). J Am Acad Dermatol. 2018;79:302–14.e6. 82. Giles DA, Moreno-Fernandez ME, Stankiewicz TE, Graspeuntner
64. Mariette X, Förger F, Abraham B, Flynn AD, Moltó A, Flipo R-M, S, Cappelletti M, Wu D, et al. Thermoneutral housing exacerbates
et al. Lack of placental transfer of certolizumab pegol during preg- nonalcoholic fatty liver disease in mice and allows for sex-inde-
nancy: Results from CRIB, a prospective, postmarketing, pharma- pendent disease modeling. Nat Med. 2017;23:829–38.
cokinetic study. Ann Rheum Dis. 2018;77:228–33. 83. Xu J, Ma HY, Liu X, Rosenthal S, Baglieri J, McCubbin R, et al.
65. Mease PJ, Fleischmann R, Deodhar AA, Wollenhaupt J, Khraishi M, Blockade of IL-17 signaling reverses alcohol-induced liver injury
Kielar D, et al. Effect of certolizumab pegol on signs and symptoms and excessive alcohol drinking in mice. JCI Insight. 2020;5:e131277.
in patients with psoriatic arthritis: 24-week results of a Phase 3 84. Ikumi K, Odanaka M, Shime H, Imai M, Osaga S, Taguchi O, et al.
double-blind randomised placebo-controlled study (RAPID-PsA). Hyperglycemia Is Associated with Psoriatic Inflammation in Both
Ann Rheum Dis. 2014;73:48–55. Humans and Mice. J Invest Dermatol. 2019;139:1329–1338.e7.
66. Berkhout LC, Vogelzang EH, Hart MM, Loeff FC, Dijk L, Derksen 85. Blauvelt A, Reich K, Warren RB, Szepietowski JC, Sigurgeirsson B,
NIL, et al. The effect of certolizumab drug concentration and Tyring SK, et al. Secukinumab re-initiation achieves regain of high
anti-drug antibodies on TNF neutralisation. Clin Exp Rheumatol. response levels in patients who interrupt treatment for moderate
2020;38:306–13. to severe plaque psoriasis. Br J Dermatol. 2017;177:879–81.
67. Gehin JE, Goll GL, Warren DJ, Syversen SW, Sexton J, Strand EK, 86. Blauvelt A, Papp KA, Sofen H, Augustin M, Yosipovitch G, Katoh
et al. Associations between certolizumab pegol serum levels, an- N, et al. Continuous dosing versus interrupted therapy with ixeki-
ti-drug antibodies and treatment response in patients with inflam- zumab: an integrated analysis of two phase 3 trials in psoriasis. J
matory joint diseases: Data from the NOR-DMARD study. Arthritis Eur Acad Dermatology Venereol. 2017;31:1004–13.
Res Ther. 2019;21:256. 87. Bagel J, Lebwohl M, Israel RJ, Jacobson A. Immunogenicity and
68. Amatya N, Garg AV, Gaffen SL. IL-17 signaling: the yin and the skin clearance recapture in clinical studies of brodalumab. J Am
yang. Trends Immunol. 2017;38:310–22. Acad Dermatol. 2020;82:344–51.
69. McGeachy MJ, Cua DJ, Gaffen SL. The IL-17 family of cytokines in 88. Al-Hammadi A, Ruszczak Z, Magariños G, Chu CY, El Dershaby Y,
health and disease. Immunity. 2019;50:892–906. Tarcha N. Intermittent use of biologic agents for the treatment of
70. Monin L, Gaffen SL. Interleukin 17 family cytokines: signal- psoriasis in adults. J Eur Acad Dermatology Venereol. 2020. https://
ing and therapeutic implications. Cold Spring Harb Perspect Biol. doi.org/10.1111/jdv.16803
2018;10:a028522. 89. Strand V, Balsa A, Al-Saleh J, Barile-Fabris L, Horiuchi T, Takeuchi
71. Su Y, Huang J, Zhao X, Lu H, Wang W, Yang XO, et al. Interleukin-17 T, et al. Immunogenicity of biologics in chronic inflammatory dis-
receptor D constitutes an alternative receptor for interleu- eases: a systematic review. BioDrugs. 2017;31:299–316.
kin-17A important in psoriasis-like skin inflammation. Sci Immunol. 90. Spindeldreher S, Maille B, Correia E, Tenon M, Karle A, Jarvis
2019;4:eaau9657. P, et al. Secukinumab demonstrates significantly lower immu-
72. Matsuzaki G, Umemura M. Interleukin-17 family cytokines in nogenicity potential compared to ixekizumab. Dermatol Ther.
protective immunity against infections: role of hematopoietic 2018;8:57–68.
cell-derived and non-hematopoietic cell-derived interleukin-17s. 91. Hueber W, Sands BE, Lewitzky S, Vandemeulebroecke M, Reinisch
Microbiol Immunol. 2018;62:1–13. W, Higgins PD, et al. Secukinumab, a human anti-IL-17A monoclo-
73. Lee JS, Tato CM, Joyce-Shaikh B, Gulen MF, Cayatte C, Chen Y, nal antibody, for moderate to severe Crohn’s disease: Unexpected
et al. Interleukin-23-independent IL-17 production regulates intes- results of a randomised, double-blindplacebo- controlled trial. Gut.
tinal epithelial permeability. Immunity. 2015;43:727–38. 2012;61:1693–700.
74. Maxwell JR, Zhang Y, Brown WA, Smith CL, Byrne FR, Fiorino M, 92. Langley RG, Elewski BE, Lebwohl M, Reich K, Griffiths CE, Papp
et al. Differential roles for interleukin-23 and interleukin-17 in in- K, et al. Secukinumab in plaque psoriasis — results of two phase 3
testinal immunoregulation. Immunity. 2015;43:739–50. trials. N Engl J Med. 2014;371:326–38.
75. McGonagle DG, McInnes IB, Kirkham BW, Sherlock J, Moots 93. McInnes IB, Behrens F, Mease PJ, Kavanaugh A, Ritchlin C,
R. The role of IL-17A in axial spondyloarthritis and psoriatic ar- Nash P, et al. Secukinumab versus adalimumab for treatment
thritis: recent advances and controversies. Ann Rheum Dis. of active psoriatic arthritis (EXCEED): a double-blind, paral-
2019;78:1167–78. lel-group, randomised, active-controlled, phase 3b trial. Lancet.
76. Cheuk S, Wiken M, Blomqvist L, Nylén S, Talme T, Ståhle M, et al. 2020;395:1496–505.
Epidermal Th22 and Tc17 cells form a localized disease memory in 94. Baeten D, Sieper J, Braun J, Baraliakos X, Dougados M, Emery
clinically healed psoriasis. J Immunol. 2014;192:3111–20. P, et al. Secukinumab, an interleukin-17A inhibitor, in ankylosing
77. Pan Y, Tian T, Park CO, Lofftus SY, Mei S, Liu X, et al. Survival of tis- spondylitis. N Engl J Med. 2015;373:2534–48.
sue-resident memory T cells requires exogenous lipid uptake and 95. Liu L, Lu J, Allan BW, Tang Y, Tetreault J, Chow CK, et al. Generation
metabolism. Nature. 2017;543:252–6. and characterization of ixekizumab, a humanized monoclonal anti-
78. Venken K, Jacques P, Mortier C, Labadia ME, Decruy T, Coudenys body that neutralizes interleukin-17A. J Inflamm Res. 2016;9:39–50.
J, et al. RORγt inhibition selectively targets IL-17 producing iNKT 96. Gordon KB, Blauvelt A, Papp KA, Langley RG, Luger T, Ohtsuki M,
and γδ-T cells enriched in Spondyloarthritis patients. Nat Commun. et al. Phase 3 trials of ixekizumab in moderate-to-severe plaque
2019;10:9. psoriasis. N Engl J Med. 2016;375:345–56.
79. Gracey E, Qaiyum Z, Almaghlouth I, Lawson D, Karki S, Avvaru 97. Blauvelt A, Papp K, Gottlieb A, Jarell A, Reich K, Maari C,
N, et al. IL-7 primes IL-17 in mucosal-associated invariant T (MAIT) et al. A head-to-head comparison of ixekizumab vs. guselkumab
HONMA and HAYASHI | 775

in patients with moderate-to-severe plaque psoriasis: 12-week patients with moderate to severe psoriasis with randomized with-
efficacy, safety and speed of response from a randomized, dou- drawal and retreatment: Results from the phase III, double-blind,
ble-blinded trial. Br J Dermatol. 2020;182:1348–58. p. J Am Acad Dermatol. 2017;76:418–31.
98. Mease PJ, Smolen JS, Behrens F, Nash P, Liu Leage S, Li L, et al. A 114. Leonardi CL, Kimball AB, Papp KA, Yeilding N, Guzzo C, Wang Y.
head-to-head comparison of the efficacy and safety of ixekizumab Efficacy and safety of ustekinumab, a human interleukin-12/23
and adalimumab in biological-naïve patients with active psoriatic monoclonal antibody, in patients with psoriasis: 76-Week re-
arthritis: 24-week results of a randomised, open-label, blinded-as- sults from a randomised, double-blind, placebo-controlled trial
sessor trial. Ann Rheum Dis. 2020;79:123–31. (PHOENIX 1). Lancet. 2008;371:1665–74.
99. van der Heijde D, Cheng-Chung Wei J, Dougados M, Mease P, 115. Papp KA, Langley RG, Lebwohl M, Krueger GG, Szapary P, Yeilding
Deodhar A, Maksymowych WP, et al. Ixekizumab, an interleu- N, et al. Efficacy and safety of ustekinumab, a human interleu-
kin-17A antagonist in the treatment of ankylosing spondylitis kin-12/23 monoclonal antibody, in patients with psoriasis: 52-
or radiographic axial spondyloarthritis in patients previously week results from a randomised, double-blind, placebo-controlled
untreated with biological disease-modifying anti-rheumatic trial (PHOENIX 2). Lancet. 2008;371:1675–84.
drugs (COAST-V): 16 week results of a phase 3 ra. Lancet. 116. Yiu ZZN, Mason KJ, Hampton PJ, Reynolds NJ, Smith CH, Lunt M,
2018;392:2441–51. et al. Drug survival of adalimumab, ustekinumab and secukinumab in
100. Papp KA, Reich K, Paul C, Blauvelt A, Baran W, Bolduc C, et al. patients with psoriasis: a prospective cohort study from the British
A prospective phase III, randomized, double-blind, placebo-con- Association of Dermatologists Biologics and Immunomodulators
trolled study of brodalumab in patients with moderate-to-severe Register (BADBIR). Br J Dermatol. 2020;183:294–302.
plaque psoriasis. Br J Dermatol. 2016;175:273–86. 117. Menter A, Papp KA, Gooderham M, Pariser DM, Augustin M,
101. Lebwohl M, Strober B, Menter A, Gordon K, Weglowska J, Puig L, Kerdel FA, et al. Drug survival of biologic therapy in a large, dis-
et al. Phase 3 studies comparing brodalumab with ustekinumab in ease-based registry of patients with psoriasis: results from the
psoriasis. N Engl J Med. 2015;373:1318–28. Psoriasis Longitudinal Assessment and Registry (PSOLAR). J Eur
102. Mease PJ, Helliwell PS, Hjuler KF, Raymond K, McInnes I. Acad Dermatology Venereol. 2016;30:1148–58.
Brodalumab in psoriatic arthritis: results from the randomised 118. Lin PT, Wang SH, Chi CC. Drug survival of biologics in treat-
phase III AMVISION-1 and AMVISION-2 trials. Ann Rheum Dis. ing psoriasis: a meta-analysis of real-world evidence. Sci Rep.
2020. https://doi.org/10.1136/annrh​eumdi​s-2019-216835 2018;8:16068.
103. Adams R, Maroof A, Baker T, Lawson ADG, Oliver R, Paveley R, 119. Ritchlin C, Rahman P, Kavanaugh A, McInnes IB, Puig L, Li S, et al.
et al. Bimekizumab, a novel humanized IgG1 antibody that neutral- Efficacy and safety of the anti-IL-12/23 p40 monoclonal antibody,
izes both IL-17A and IL-17F. Front Immunol. 2020;11:1894. ustekinumab, in patients with active psoriatic arthritis despite
104. Goepfert A, Lehmann S, Wirth E, Rondeau JM. The human IL- conventional non-biological and biological anti-tumour necrosis
17A/F heterodimer: a two-faced cytokine with unique receptor factor therapy: 6-month and 1-year results of the phase 3, multi-
recognition properties. Sci Rep. 2017;7:8906. centre, double-blind, placebo-controlled, randomised PSUMMIT 2
105. Ishigame H, Kakuta S, Nagai T, Kadoki M, Nambu A, Komiyama Y, trial. Ann Rheum Dis. 2014;73:990–9.
et al. Differential roles of interleukin-17A and -17F in host defense 120. Blauvelt A, Papp KA, Griffiths CEM, Randazzo B, Wasfi Y, Shen
against mucoepithelial bacterial infection and allergic responses. YK, et al. Efficacy and safety of guselkumab, an anti-interleukin-23
Immunity. 2009;30:108–19. monoclonal antibody, compared with adalimumab for the con-
106. Papp KA, Merola JF, Gottlieb AB, Griffiths CEM, Cross N, Peterson tinuous treatment of patients with moderate to severe psoriasis:
L, et al. Dual neutralization of both interleukin 17A and interleu- Results from the phase III, double-blinded, placebo- and active
kin 17F with bimekizumab in patients with psoriasis: Results from comparator. J Am Acad Dermatol. 2017;76:405–17.
BE ABLE 1, a 12-week randomized, double-blinded, placebo-con- 121. Terui T, Kobayashi S, Okubo Y, Murakami M, Zheng R, Morishima
trolled phase 2b trial. J Am Acad Dermatol. 2018;79:277–86.e10. H, et al. Efficacy and safety of guselkumab in Japanese patients
107. Ritchlin CT, Kavanaugh A, Merola JF, Schett G, Scher JU, Warren with palmoplantar pustulosis: a phase 3 randomized clinical trial.
RB, et al. Bimekizumab in patients with active psoriatic arthritis: JAMA Dermatol. 2019;155:1153–61.
results from a 48-week, randomised, double-blind, placebo-con- 122. Deodhar A, Helliwell PS, Boehncke WH, Kollmeier AP, Hsia EC,
trolled, dose-ranging phase 2b trial. Lancet. 2020;395:427–40. Subramanian RA, et al. Guselkumab in patients with active psoriatic
108. Sakkas LI, Mavropoulos A, Perricone C, Bogdanos DP. IL-35: a new arthritis who were biologic-naive or had previously received TNFα
immunomodulator in autoimmune rheumatic diseases Treg Breg. inhibitor treatment (DISCOVER-1): a double-blind, randomised,
Immunol Res. 2018;66:305–12. placebo-controlled phase 3 trial. Lancet. 2020;395:1115–25.
109. Teng MWL, Bowman EP, McElwee JJ, Smyth MJ, Casanova JL, 123. Mease PJ, Rahman P, Gottlieb AB, Kollmeier AP, Hsia EC, Xu XL,
Cooper AM, et al. IL-12 and IL-23 cytokines : from discovery to et al. Guselkumab in biologic-naive patients with active psoriatic
targeted therapies for immune-mediated inflammatory diseases. arthritis (DISCOVER-2): a double-blind, randomised, placebo-con-
Nat Med. 2015;21:719–29. trolled phase 3 trial. Lancet. 2020;395:1126–36.
110. Lee E, Trepicchio WL, Oestreicher JL, Pittman D, Wang F, Chamian 124. Gordon KB, Strober B, Lebwohl M, Augustin M, Blauvelt A, Poulin
F, et al. Increased Expression of Interleukin 23 p19 and p40 Y, et al. Efficacy and safety of risankizumab in moderate-to-severe
in Lesional Skin of Patients with Psoriasis Vulgaris. J Exp Med. plaque psoriasis (UltIMMa-1 and UltIMMa-2): results from two
2004;199:125–30. double-blind, randomised, placebo-controlled and ustekinum-
111. Koenen HJPM, Smeets RL, Vink PM, van Rijssen E, Boots AM, ab-controlled phase 3 trials. Lancet. 2018;392:650–61.
Joosten I. Human CD25 high Foxp3 pos regulatory T cells differ- 125. Reich K, Gooderham M, Thaçi D, Crowley JJ, Ryan C, Krueger JG, et al.
entiate into IL-17 – producing cells. Blood. 2008;112:2340–53. Risankizumab compared with adalimumab in patients with moder-
112. Bovenschen HJ, Van De Kerkhof PC, Van Erp PE, Woestenenk R, ate-to-severe plaque psoriasis (IMMvent): a randomised, double-blind,
Joosten I, Koenen HJ. Foxp3 regulatory T cells of psoriasis patients active-comparator-controlled phase 3 trial. Lancet. 2019;394:576–86.
easily differentiate into IL-17A-producing cells and are found in le- 126. Blauvelt A, Leonardi CL, Gooderham M, Papp KA, Philipp S, Wu
sional skin. J Invest Dermatol. 2011;131:1853–60. JJ, et al. Efficacy and safety of continuous risankizumab therapy vs
113. Reich K, Armstrong AW, Foley P, Song M, Wasfi Y, Randazzo B, et al. treatment withdrawal in patients with moderate to severe plaque
Efficacy and safety of guselkumab, an anti-interleukin-23 mono- psoriasis: a phase 3 Randomized Clinical Trial. JAMA Dermatol.
clonal antibody, compared with adalimumab for the treatment of 2020;156:649–58.
776 | HONMA and HAYASHI

127. Baeten D, Østergaard M, Wei JC, Sieper J, Järvinen P, Tam LS, et al. 148. Gerlo S, Kooijman R, Beck IM, Kolmus K, Spooren A, Haegeman G.
Risankizumab, an IL-23 inhibitor, for ankylosing spondylitis: Results Cyclic AMP: A selective modulator of NF-κB action. Cell Mol Life
of a randomised, double-blind, placebo-controlled, proof-of-concept, Sci. 2011;68:3823–41.
dose-finding phase 2 study. Ann Rheum Dis. 2018;77:1295–302. 149. Barker G, Parnell E, van Basten B, Buist H, Adams DR, Yarwood
128. Reich K, Warren RB, Iversen L, Puig L, Pau-Charles I, Igarashi A, SJ. The Potential of a novel class of EPAC-selective agonists
et al. Long-term efficacy and safety of tildrakizumab for moder- to combat cardiovascular inflammation. J Cardiovasc Dev Dis.
ate-to-severe psoriasis: pooled analyses of two randomized phase 2017;4:22.
III clinical trials (reSURFACE 1 and reSURFACE 2) through 148 150. Arumugham VB, Baldari CT. cAMP: a multifaceted modulator
weeks. Br J Dermatol. 2020;182:605–17. of immune synapse assembly and T cell activation. J Leukoc Biol.
129. Reich K, Papp KA, Blauvelt A, Tyring SK, Sinclair R, Thaçi D, et al. 2017;101:1301–16.
Tildrakizumab versus placebo or etanercept for chronic plaque 151. Noval Rivas M, Chatila TA. Regulatory T cells in allergic diseases. J
psoriasis (reSURFACE 1 and reSURFACE 2): results from two ran- Allergy Clin Immunol. 2016;138:639–52.
domised controlled, phase 3 trials. Lancet. 2017;390:276–88. 152. Wing JB, Tanaka A, Sakaguchi S. Human FOXP3 + regulatory T cell
130. Reich K, Rich P, Maari C, Bissonnette R, Leonardi C, Menter A, heterogeneity and function in autoimmunity and cancer. Immunity.
et al. Efficacy and safety of mirikizumab (LY3074828) in the treat- 2019;50:302–16.
ment of moderate-to-severe plaque psoriasis: results from a ran- 153. Rueda CM, Jackson CM, Chougnet CA, August A. Regulatory
domized phase II study. Br J Dermatol. 2019;181:88–95. T-cell-mediated suppression of conventional T-Cells and dendritic
131. Sandborn WJ, Ferrante M, Bhandari BR, Berliba E, Feagan BG, Hibi cells by different cAMP intracellular pathways. Front Immunol.
T, et al. Efficacy and Safety of Mirikizumab in a Randomized Phase 2016;7:216.
2 Study of Patients With Ulcerative Colitis. Gastroenterology. 154. Klein M, Bopp T, Bopp T. Cyclic AMP represents a crucial com-
2020;158:537–49. ponent of treg cell-mediated immune regulation. Front Immunol.
132. Walsh PT, Fallon PG. The emergence of the IL-36 cytokine fam- 2016;7:315.
ily as novel targets for inflammatory diseases. Ann N Y Acad Sci. 155. Samrao A, Berry TM, Goreshi R, Simpson EL. A pilot study of an
2016;1417:23–34. oral phosphodiesterase inhibitor (apremilast) for atopic dermatitis
133. Yi G, Ybe JA, Saha SS, Caviness G, Raymond E, Ganesan R, et al. in adults. Arch Dermatol. 2012;148:890–7.
Structural and functional attributes of the interleukin-36 receptor. 156. Bopp T, Dehzad N, Reuter S, Klein M, Ullrich N, Stassen M, et al.
J Biol Chem. 2016;291:16597–609. Inhibition of cAMP degradation improves regulatory T cell-medi-
134. Fields JK, Günther S, Sundberg EJ. Structural basis of IL-1 family ated suppression. J Immunol. 2009;182:4017–24.
cytokine signaling. Front Immunol. 2019;10:1412. 157. Garcet S, Nograles K, Correa da Rosa J, Schafer PH, Krueger
135. Han Y, Huard A, Mora J, da Silva P, Brüne B, Weigert A. IL-36 fam- JG. Synergistic cytokine effects as apremilast response pre-
ily cytokines in protective versus destructive inflammation. Cell dictors in patients with psoriasis. J Allergy Clin Immunol.
Signal. 2020;75:109773. 2018;142(1010–1013):e6.
136. Furue K, Yamamura K, Tsuji G, Mitoma C, Uchi H, Nakahara T, et al. 158. Strober B, Alikhan A, Lockshin B, Shi R, Cirulli J, Schafer P.
Highlighting interleukin-36 signalling in plaque psoriasis and pus- Apremilast mechanism of efficacy in systemic-naive patients with
tular psoriasis. Acta Derm Venereol. 2018;98:5–13. moderate plaque psoriasis: Pharmacodynamic results from the
137. Mercurio L, Failla CM, Capriotti L, Scarponi C, Facchiano F, Morelli UNVEIL study. J Dermatol Sci. 2019;96:126–33.
M, et al. Interleukin ( IL ) -17 / IL-36 axis participates to the cross- 159. Hashim PW, Chima M, Kim HJ, Bares J, Yao CJ, Singer G, et al.
talk between endothelial cells and keratinocytes during inflamma- Crisaborole 2% ointment for the treatment of intertriginous, ano-
tory skin responses. PLoS One. 2020;15:e0222969. genital, and facial psoriasis: A double-blind, randomized, vehi-
138. Buhl AL, Wenzel J. Interleukin-36 in infectious and inflammatory cle-controlled trial. J Am Acad Dermatol. 2020;82:360–5.
skin diseases. Front Immunol. 2019;10:1162. 160. Lebwohl MG, Papp KA, Stein Gold LS, Gooderham MJ, Kircik LH,
139. Akiyama M, Takeichi T, McGrath JA, Sugiura K. Autoinflammatory Draelos ZD, et al. Trial of roflumilast cream for chronic plaque pso-
keratinization diseases: An emerging concept encompassing vari- riasis. N Engl J Med. 2020;383:229–39.
ous inflammatory keratinization disorders of the skin. J Dermatol 161. Papp K, Reich K, Leonardi CL, Kircik L, Chimenti S, Langley RG,
Sci. 2018;90:105–11. et al. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor,
140. Akiyama M, Takeichi T, McGrath JA, Sugiura K. Autoinflammatory in patients with moderate to severe plaque psoriasis: Results of
keratinization diseases. J Allergy Clin Immunol. 2017;140:1545–7. a phase III, randomized, controlled trial (Efficacy and Safety Trial
141. Marrakchi S, Guigue P, Renshaw BR, Puel A, Pei XY, Fraitag S, et al. Evaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1). J Am
Interleukin-36–receptor antagonist deficiency and generalized Acad Dermatol. 2015;73:37–49.
pustular psoriasis. N Engl J Med. 2011;365:620–8. 162. Paul C, Cather J, Gooderham M, Poulin Y, Mrowietz U, Ferrandiz C,
142. Pfaff CM, Marquardt Y, Fietkau K, Baron JM, Lüscher B. The pso- et al. Efficacy and safety of apremilast, an oral phosphodiesterase
riasis-associated IL-17A induces and cooperates with IL-36 cyto- 4 inhibitor, in patients with moderate-to-severe plaque psoriasis
kines to control keratinocyte differentiation and function. Sci Rep. over 52 weeks: A phase III, randomized controlled trial (ESTEEM
2017;7:15631. 2). Br J Dermatol. 2015;173:1387–99.
143. Wang WM, Jin HZ. Biologics in the treatment of pustular psoriasis. 163. Rich P, Gooderham M, Bachelez H, Goncalves J, Day RM, Chen R,
Expert Opin Drug Saf. 2020;19:969–80. et al. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients
144. Bachelez H, Choon S, Marrakchi S, Burden AD, Tsai TF, Morita A, with difficult-to-treat nail and scalp psoriasis: Results of 2 phase
et al. Inhibition of the interleukin-36 pathway for the treatment of III randomized, controlled trials (ESTEEM 1 and ESTEEM 2). J Am
generalized pustular psoriasis. N Engl J Med. 2019;380:981–983. Acad Dermatol. 2016;74:134–42.
145. Sassone-Corsi P. The cyclic AMP pathway. Cold Spring Harb 164. Kishimoto M, Komine M, Hioki T, Kamiya K, Sugai J, Ohtsuki M.
Perspect Biol. 2012;4:10–3. Real-world use of apremilast for patients with psoriasis in Japan. J
146. Kumar N, Goldminz AM, Kim N, Gottlieb AB. Phosphodiesterase Dermatol. 2018;45:1345–8.
4-targeted treatments for autoimmune diseases. BMC Med. 165. Cutolo M, Myerson GE, Fleischmann RM, Lioté F, Díaz-González
2013;11:96. F, Van den Bosch F, et al. A phase III, randomized, controlled trial
147. Li H, Zuo J, Tang W. Phosphodiesterase-4 inhibitors for the treat- of apremilast in patients with psoriatic arthritis: Results of the
ment of inflammatory diseases. Front Pharmacol. 2018;9:1048. PALACE 2 trial. J Rheumatol. 2016;43:1724–34.
HONMA and HAYASHI | 777

166. Edwards CJ, Blanco FJ, Crowley J, Birbara CA, Jaworski J, Aelion J, et al. 183. Papp KA, Menter MA, Abe M, Elewski B, Feldman SR, Gottlieb
Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with pso- AB, et al. Tofacitinib, an oral Janus kinase inhibitor, for the treat-
riatic arthritis and current skin involvement: A phase III, randomised, ment of chronic plaque psoriasis : results from two randomized,
controlled trial (PALACE 3). Ann Rheum Dis. 2016;75:1065–73. placebo-controlled, phase III trials. Br J Dermatol. 2015;173:
167. Lee EB, Amin M, Egeberg A, Wu JJ. Adverse events associated 949–61.
with apremilast use and withdrawal for psoriasis in a real-world 184. Mease P, Hall S, Fitzgerald O, van der Heijde D, Merola JF, Avila-
setting. J Eur Acad Dermatology Venereol. 2018;32:e393–4. Zapata F, et al. Tofacitinib or Adalimumab versus Placebo for
168. Moon C, Zhang W, Sundaram N, Yarlagadda S, Reddy VS, Arora K, Psoriatic Arthritis. N Engl J Med. 2017;377:1537–50.
et al. Drug-induced secretory diarrhea: A role for CFTR. Pharmacol 185. Rigby W, Azevedo VF, Behrens F, Blanco R, Kaszuba A. , Gladman
Res. 2015;102:107–12. D, et al. Tofacitinib for psoriatic arthritis in patients with an in-
169. Paris M, Chen M, Yazici Y. Trial of Apremilast for Oral Ulcers in adequate response to TNF inhibitors. N Engl J Med. 2017;377:
Behçet’s Syndrome. N Engl J Med. 2019;381:1918–28. 1525–36.
170. Leonard WJ, Lin J. Cytokine receptor signaling pathways. J Allergy 186. Winthrop KL, Lebwohl M, Cohen AD, Weinberg JM. Herpes
Clin Immunol. 2000;105:877–88. zoster in psoriasis patients treated with tofacitinib. J Am Acad
171. Gadina M, Gazaniga N, Vian L, Furumoto Y. Small molecules to the Dermatology. 2017;77:302–9.
rescue : Inhibition of cytokine signaling in immune-mediated dis- 187. Abe M, Nishigori C, Torii H, Ihn H, Ito K, Nagaoka M, et al. Tofacitinib
eases. J Autoimmun. 2017;85:20–31. for the treatment of moderate to severe chronic plaque psoriasis in
172. Liao W, Lin J, Leonard WJ. IL-2 family cytokines : new insights into Japanese patients : Subgroup analyses from a randomized, place-
the complex roles of IL-2 as a broad regulator of T helper cell dif- bo-controlled phase 3 trial. J Dermatol. 2017;44:1228–37.
ferentiation. Curr Opin Immunol. 2011;23:598–604. 188. Papp KA, Menter MA, Raman M, Disch D, Schlichting DE, Gaich
173. Floss DM, Schröder J, Franke M, Scheller J. Insights into IL-23 C, et al. A randomized phase 2b trial of baricitinib, an oral Janus ki-
biology: From structure to function. Cytokine Growth Factor Rev. nase ( JAK ) 1 / JAK2 inhibitor, in patients with moderate-to-severe
2015;26:569–78. psoriasis. Br J Dermatol. 2016;174:1266–76.
174. Zdanov A. Structural analysis of cytokines comprising the IL-10 189. Papp K, Gordon K, Thaçi D, Morita A, Gooderham M, Foley P, et al.
family. Cytokine Growth Factor Rev. 2010;21:325–30. Phase 2 trial of selective tyrosine kinase 2 inhibition in psoriasis. N
175. Bhoopalan SV, Huang LJ, Weiss MJ. Erythropoietin regulation Engl J Med. 2018;379:1313–21.
of red blood cell production : from bench to bedside and back. 190. Capone A, Volpe E. Transcriptional regulators of T helper 17 cell
F1000Research. 2020;9(1153). differentiation in health and autoimmune diseases. Front Immunol.
176. Murakami M, Kamimura D, Hirano T. Pleiotropy and Specificity: 2020;11:348.
Insights from the Interleukin 6 Family of Cytokines. Immunity. 191. Won HY, Hwang ES. Transcriptional modulation of regulatory
2019;50:812–31. T cell development by novel regulators NR4As. Arch Pharm Res.
177. Schreiber G. The molecular basis for differential type I interferon 2016;39:1530–6.
signaling. J Biol Chem. 2017;292:7285–94. 192. Zaro BW, Vinogradova EV, Lazar DC, Blewett MM, Suciu RM,
178. Ng CT, Mendoza JL, Garcia KC, Oldstone MBA. Alpha and beta Takaya J, et al. Dimethyl fumarate disrupts human innate immune
type 1 interferon signaling: passage for diverse biologic outcomes. signaling by targeting the IRAK4–MyD88 complex. J Immunol.
Cell. 2016;164:349–52. 2019;202:2737–46.
179. Green DS, Young HA, Valencia JC. Current prospects of type 193. Zarrin AA, Bao K, Lupardus P, Vucic D. Kinase inhibition in autoim-
II interferon-gamma signaling and autoimmunity. J Biol Chem. munity and inflammation. Nat Rev Drug Discov. 2020;11:348.
2017;292:13925–33. 194. Melton E, Qiu H. Interleukin-36 cytokine/receptor signaling: a
180. Solimani F, Meier K, Ghoreschi K. Emerging Topical and Systemic new target for tissue fibrosis. Int J Mol Sci. 2020;21:4–9.
JAK Inhibitors in Dermatology. Front Immunol. 2019;10:2847.
181. Schwartz DM, Bonelli M, Gadina M, O’Shea JJ. Type I/II cytokines,
JAKs, and new strategies for treating autoimmune diseases. Nat
How to cite this article: Honma M, Hayashi K. Psoriasis:
Rev Rheumatol. 2016;12:25–36.
182. Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, O'Shea JJ.
Recent progress in molecular-targeted therapies. J Dermatol.
JAK inhibition as a therapeutic strategy for immune and inflamma- 2021;48:761–777. https://doi.org/10.1111/1346-8138.15727
tory diseases. Nat Rev Drug Discov. 2017;17:78.

You might also like