ArticlePDF AvailableLiterature Review

JAK3 inhibitors for the treatment of inflammatory and autoimmune diseases: a patent review (2016-present)

Authors:

Abstract and Figures

Introduction Up to now, a total of eight Janus kinase (JAK) inhibitors have been approved for the treatment of autoimmune and myeloproliferative disease. The JAK family belongs to the non-receptor tyrosine kinase family, consisting of JAK1, JAK2, JAK3, and tyrosine kinase 2. Among these four subtypes, only JAK3 is mainly expressed in hematopoietic tissue cells and is exclusively associated with the cytokines shared in the common gamma chain receptor subunit. Due to its specific tissue distribution and functional characteristics that distinguish it from the other JAKs family subtypes, JAK3 is a promising target for the treatment of autoimmune disease. Areas covered This study aimed to provide a comprehensive review of the available patent literature on JAK-family inhibitors published from 2016 to the present. In addition, an overview of the clinical activities of selective JAK3 inhibitors in recent years was provided. Expert opinion To date, no selective JAK3 inhibitors have been approved for use in clinics. Over the last five years, an increasing number of studies on JAK3 inhibitors, particularly ritlecitinib by Pfizer, have demonstrated their promising therapeutic potential. In this review, recent studies reported that selective JAK3 inhibitors may offer valid, interesting, and promising therapeutic potential in inflammatory and autoimmune diseases.
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=ietp20
Expert Opinion on Therapeutic Patents
ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ietp20
JAK3 inhibitors for the treatment of inflammatory
and autoimmune diseases: a patent review
(2016–present)
Chengjuan Chen, Dianxiang Lu, Tao Sun & Tiantai Zhang
To cite this article: Chengjuan Chen, Dianxiang Lu, Tao Sun & Tiantai Zhang (2022):
JAK3 inhibitors for the treatment of inflammatory and autoimmune diseases: a patent review
(2016–present), Expert Opinion on Therapeutic Patents, DOI: 10.1080/13543776.2022.2023129
To link to this article: https://doi.org/10.1080/13543776.2022.2023129
View supplementary material
Published online: 06 Jan 2022.
Submit your article to this journal
Article views: 142
View related articles
View Crossmark data
REVIEW
JAK3 inhibitors for the treatment of inflammatory and autoimmune diseases:
a patent review (2016–present)
Chengjuan Chen
a
*, Dianxiang Lu
b
*, Tao Sun
c
and Tiantai Zhang
a
a
State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences & Peking Union Medical College, Beijing, China;
b
Research Center for High Altitude Medicine, Key Laboratory of Ministry of Education for
High Altitude Medicine, Qinghai University, Xining, Qinghai, China;
c
Department of Cardiology, Huashan Hospital, Fudan University, Shanghai,
China
ABSTRACT
Introduction: Up to now, a total of eight Janus kinase (JAK) inhibitors have been approved for the
treatment of autoimmune and myeloproliferative disease. The JAK family belongs to the non-receptor
tyrosine kinase family, consisting of JAK1, JAK2, JAK3, and Tyk2. Among these four subtypes, only JAK3
is mainly expressed in hematopoietic tissue cells and is exclusively associated with the cytokines shared
in the common gamma-chain receptor subunit. Due to its specific tissue distribution and functional
characteristics that distinguish it from the other JAKs family subtypes, JAK3 is a promising target for the
treatment of autoimmune disease.
Areas covered: This study aimed to provide a comprehensive review of the available patent literature
on JAK-family inhibitors published from 2016 to the present. In addition, an overview of the clinical
activities of selective JAK3 inhibitors in recent years was provided.
Expert opinion: To date, no selective JAK3 inhibitors have been approved for use in clinics. Over the
last 5 years, an increasing number of studies on JAK3 inhibitors, particularly ritlecitinib by Pfizer, have
demonstrated their promising therapeutic potential. In this review, recent studies reported that selec-
tive JAK3 inhibitors may offer valid, interesting, and promising therapeutic potential in inflammatory
and autoimmune diseases.
ARTICLE HISTORY
Received 10 August 2021
Accepted 22 December 2021
KEYWORDS
Janus kinase; JAK3 subtype;
JAK3 inhibitor; autoimmune
disease; patent review
1. Introduction
The Janus kinases (JAKs) family, which includes JAK1,
JAK2, JAK3, and tyrosine kinase 2 (Tyk2), are intracellular
non-receptor tyrosine kinases that play a key role in deli-
vering cytokine signals from membrane receptors to the
nucleus for transcriptional regulation by signal transducers
and activators of transcription (STAT) [1]. There are over
50 cytokines in mammals, including interleukins, interfer-
ons, and colony-stimulating factors, that exert their effects
through the JAK-STAT signaling pathway. Therefore, JAKs
are critical regulators of cytokine pathways and an attrac-
tive target for the development of anti-inflammatory
drugs in both autoimmune and inflammatory diseases, as
well as myeloproliferative diseases [2].
JAKs have seven distinct Janus homology domain 1–7
(JH1-7) regions and contain approximately 1150 amino
acid residues with about 120–130 kDa molecular weights
[3]. JAK3 has a cysteine residue at position 909 (Cys909) in
its amino acid sequence, which is replaced by a serine
residue at the same position in the other three JAK iso-
forms [4]. In addition to JAK3, only 10 other kinases
possess a Cys909 residue in the ATP-binding site, and
most covalent inhibition strategies target Cys909 to
design selective inhibitor [5]. In contrast to the ubiquitous
expression of the other three JAK family members, JAK3 is
predominantly expressed in hematopoietic tissue cells,
such as NK cells, bone marrow cells, activated
B lymphocytes, and T lymphocytes. The leukocyte-specific
JAK3 was uniquely associated with a shared receptor sub-
unit of the common gamma chain (γc) for IL-2, IL-4, IL-7,
IL-9, IL-15, and IL-21, which regulate the growth and
maturation of NK cells, B cells, and T cells [6]. The loss-of-
function mutations of JAK3 caused severe combined
immunodeficiency syndrome (SCID) [7], which further sup-
ported the importance of JAK3 in the immune system.
Based on the structural and functional characteristics of
the four JAK family subtypes, as well as specific tissue
distribution, JAK3 has emerged as an ideal target for the
treatment of inflammatory or autoimmune diseases [8].
Given the potential of JAK3 as a target, this study aimed
to review the patents on small-molecule JAK3 inhibitors
from 2016 to the present, as well as providing
CONTACT Tao Sun 071105190@fudan.edu.cn Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200040; Tiantai Zhang
ttzhang@imm.ac.cn State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences & Peking Union Medical College, Beijing 100050, China: ao
*
Those authors contributed equally.
Supplemental data for this article can be accessed here.
EXPERT OPINION ON THERAPEUTIC PATENTS
https://doi.org/10.1080/13543776.2022.2023129
© 2022 Informa UK Limited, trading as Taylor & Francis Group
a structured feature analysis of scaffolds in those patents
and the related examples. The clinical activities of selective
JAK3 inhibitors are also briefly described in this review.
2. Recent clinical advances of JAK3 inhibitors
The updated information for approved JAKs inhibitors
(Table 1) and clinical trials of JAK3 inhibitors are described in
this section. The chemical structures of JAK inhibitors that
have been approved and those still in clinical development
are displayed in Figure 1. The status of JAK3 inhibitors in
clinical trials is shown in Table 2.
2.1. Approved JAK inhibitors
Previous studies have proven that the acquired expression or
mutation of JAK kinases is related to many autoimmune dis-
eases and cancers [9–11]. In theory, inhibiting JAK activity to
alleviate or even cure related diseases has become a popular
therapeutic option [12]. JAKs have been shown to be effective
therapeutic targets and there are currently eight JAK inhibitors
that have been approved for the market in the past 10
years [13].
Ruxolitinib (Jakafi®) is a JAK1 and JAK2 dual inhibitor devel-
oped by Incyte/Novartis for the treatment of myelofibrosis
(MF), polycythemia vera (PV), essential thrombocythemia (ET),
and graft-versus-host diseases (GVHD) [14,15]. Tofacitinib
(Xeljanz®) is a pan-JAK inhibitor developed by Pfizer for the
treatment of patients with moderate-to-severe rheumatoid
arthritis (RA), psoriatic arthritis (PsA), ulcerative colitis (UC),
juvenile idiopathic arthritis (JIA), and systemic juvenile idio-
pathic arthritis (SJIA) [16,17]. Baricitinib (Olumiant®) is a JAK1
and JAK2 dual inhibitor developed by Eli Lilly/Incyte for the
treatment of RA, atopic dermatitis (AD), and COVID-19 [18].
Peficitinib (Smyraf®) is a pan-JAK inhibitor developed by
Astellas Pharma for the treatment of RA [19]. Fedratinib
(Inrebic®) is a JAK2 inhibitor developed by Celgene for the
treatment of MF [20]. Upadacitinib (Rinvoq®) is a JAK1 inhibi-
tor developed by AbbVie for the treatment of RA, psoriatic
arthritis (PsA) and ankylosing spondylitis (AS) [21,22].
Delgocitinib (Corectim®) is a pan-JAK inhibitor developed by
Japan Tobacco and Torii Pharmaceutical for the treatment
of AD [23,24]. Filgotinib (Jyseleca®) is a JAK1 and JAK2 inhi-
bitor developed by Gilead for the treatment of RA [24].
2.2. JAK3 inhibitor in clinical development
JAK3 signaling is important for regulating the development,
maintenance, and function of lymphocytes due to its exclusive
association with the γc-containing cytokines receptor unit. As
a result, JAK3 has become a popular target for the treatment
of inflammatory and autoimmune diseases [25,26]. Currently,
several JAK3 inhibitors are being evaluated in clinical trials for
various therapeutic indications.
2.2.1. Ritlecitinib
Ritlecitinib, also known as PF-06651600 developed by Pfizer, is
an orally bioavailable and potent small molecule JAK3-
selective inhibitor with an IC
50
value of 33.1 nM at 1 mM
ATP concentration for JAK3 in biochemical assay and more
than 300-fold selectivity for JAK3 subtype than other subtypes
[27]. It irreversibly inhibits the common γc-related cytokine
signaling dependent on JAK3 by covalently acting on Cys909
residue in the catalytic domain of JAK3 [28]. Ritlecitinib now
was confirmed a dual JAK3/TEC family kinase inhibitor that
displays obviously therapeutic effects on autoimmune disease
such as rheumatoid arthritis (RA), inflammatory bowel disease,
alopecia areata (AA), and vitiligo, by inhibiting Th1 and Th17
cell differentiation and function via JAK3 kinase, and cytotoxic
Table 1. Approved JAK inhibitors and their current indications (Organized alphabetically by compound name, update to 2021.06).
No. Compound Target R&D company Indication
1 Baricitinib JAK1/JAK2 Incyte/Eli Lilly RA, AD, COVID-19 (EUA)
2 Delgocitinib pan-JAK Japan Tobacco AD (approved in Japan)
3 Fedratinib JAK2/FLT3/RET Impact Biomedicines MF
4 Filgotinib JAK1 Gilead, Galapagos NV RA (approved in EU, Japan)
5 Peficitinib pan-JAK Astellas Pharma RA (approved in Japan, South Korea)
6 Ruxolitinib JAK1/JAK2 Incyte/Novartis MF, PV, ET, GVHD
7 Tofacitinib JAK3/JAK1/JAK2 Pfizer RA, PsA, JIA, SJIA, UC
8 Upadacitinib JAK1 AbbVie RA, PsA (approved in EU), AS (approved in EU)
Abbreviations: AD, atopic dermatitis; AS, ankylosing spondylitis; EUA, Emergency Use Authorization; FLT3, fms related tyrosine kinase 3; GVHD, graft-versus-host
disease; JIA, juvenile idiopathic arthritis; MF, myelofibrosis; PV, polycythemia vera; ET, essential thrombocythemia; PsA, psoriatic arthritis; RA, rheumatoid arthritis;
RET, ret proto-oncogene; SJIA, systemic juvenile idiopathic arthritis
Article Highlights
JAK3 differs from other JAK family subtypes in its cytokine signaling
specificity. In contrast to the ubiquitous expression of other JAKs,
JAK3 is predominantly expressed in the hematopoietic system. In
addition, JAK3 uniquely binds to only one cytokine receptor, the
common gamma chain.
In principle, restricted expression and function within the hemato-
poietic compartment of JAK3 should result in very limited side effects
on other organs. Thus, selective inhibition of JAK3 should have
a better risk-benefit ratio and higher efficacy in autoimmune disorders
where lymphocyte differentiation is the driving factor.
The design of compounds based on difference of the active site
Cys909 residues between JAK3 and other JAK subtypes should theo-
retically lead to JAK3 inhibitors that are more selective than other JAK
subtypes.
Therapeutic opportunities for targeting selective inhibition of JAK3
include, but are not limited to rheumatoid arthritis, inflammatory
bowel disease, cancer, diabetes as well as skin-related diseases such
as alopecia areata, psoriasis, and systemic lupus erythematosus.
Given that the JAK3 subtype is only responsible for the function of
γc-associated cytokines, selective JAK3 inhibitor has the potential to
overcome clinically occurring side effects by pan-JAKs inhibitors, such
as severe infection, thrombosis and anemia.
2C. CHEN ET AL.
function of NK cells and CD8
+
T cells via TEC kinase [29].
Ritlecitinib has been evaluated in several human phase 1–3
clinical studies due to its favorable efficacy and safety profile.
In 2018, the indication of ritlecitinib for the treatment of AA
was recognized as breakthrough therapy by FDA, and a phase
3 clinical trial is currently evaluating the safety and efficacy of
ritlecitinib in adults and adolescents (12 years and older) with
AA or scalp hair loss of 50% or more (clinicaltrial.gov registry
number: NCT04006457 and NCT03732807).
In 2020, a phase 2a clinical study of patients with mod-
erate-to-severe RA (NCT02969044) showed that oral ritleciti-
nib treatment reduced RA disease activity and generally
well-tolerated over 8 weeks [30]. In this clinical trial, the
incidence of treatment-emergent adverse events (TEAEs)
was higher in the ritlecitinib group (47.6%) than in the
placebo group (17.9), but no serious TEAEs, severe TEAEs,
or deaths were reported. Meanwhile, no bleeding events,
reductions in platelet counts < 100 × 10
3
/mm
3
, or clinically
significant symptoms of anemia or hemodynamic compro-
mise were reported [30]. Another completed phase 2a clin-
ical trial in patients with AA (NCT02974868) showed that
treatment with ritlecitinib for 24 weeks resulted in clinically
significant hair regrowth in patients who have AA with ≥
50% scalp hair loss [31]. There was a significant difference of
Figure 1. (A) The currently approved JAK inhibitors (organized based on approved time). (B) JAK3 inhibitors that are currently in clinical development.
EXPERT OPINION ON THERAPEUTIC PATENTS 3
25% patients between ritlecitinib and placebo groups. The
incidence of adverse events reported in this trial was 74%
and 67% of patients in the placebo and ritlecitinib groups,
respectively. In ritlecitinib treatment group, there was no
clinically relevant change from baseline in hematology
tests, electrocardiogram findings, or vital signs [31]. Based
on above two clinical trials, the most frequently reported
side-effects of ritlecitinib were influenza, pruritus, nasophar-
yngitis, headache, acne, and nausea, but only influenza
(7.1% in the ritlecitinib group and 0% in the placebo
group) and pruritus (4.8% in the ritlecitinib group and 3.6%
in the placebo group) showed the difference compared with
the placebo group.
Now, several human clinical studies of ritlecitinib including
the treatment of Crohn’s Disease (NCT03395184), Ulcerative
colitis (NCT02958865), and vitiligo (NCT03715829) are cur-
rently underway.
2.2.2. ATI-1777
ATI-1777, developed by Aclaris Therapeutics, is an investiga-
tional topical ‘soft’ JAK1/3 inhibitor that is designed to
provide JAK inhibition at the site of application while limit-
ing systemic exposure. A phase 2a, multicenter, randomized,
double-blind, vehicle-controlled, parallel-group clinical trial
to determine the efficacy, safety, tolerability and pharmaco-
kinetics of ATI-1777 in subjects with moderate-to-severe AD
(NCT04598269). Subjects will apply ATI-1777 topical solution
2.0% (w/w) or vehicle twice daily for 4 weeks to explore
whether a topical JAK inhibitor can successfully treat mod-
erate-to-severe AD rather than mild disease [32]. In
Jun 2021, Aclaris Therapeutics announced that the trial
achieved positive results of ATI-1777 in subjects with mod-
erate-to-severe AD with minimal systemic exposure to drug.
The full analysis showed that the trial received its primary
endpoint with a high degree of statistical significance
(p < 0.001) (one-sided p-value), which corresponded to
a 74.4% reduction in modified Eczema Area and Severity
Index (mEASI) score from baseline at week 4 in subjects
applying ATI-1777 compared to a 41.4% reduction in sub-
jects applying vehicle [33]. The topical application for
patients with moderate-to-severe atopic dermatitis is parti-
cularly relevant in light of some of the potential safety
concerns with oral therapies.
2.2.3. TD-5202
TD-5202 is an investigational, gut-selective JAK3 inhibitor devel-
oped by Theravance Biopharma for the treatment of inflammatory
bowel diseases. TD-5202 targets Cys909 residue of JAK3 to conduct
irreversible inhibition. A phase 1, randomized, double-blinded,
placebo-controlled, trial of TD-5202 in healthy volunteers is com-
pleted on 14 January 2021 (NCT04044339) [34].
2.2.4. Decernotinib
Decernotinib, also known as VX-509, is an oral selective JAK3
inhibitor developed by Vertex Pharmaceuticals. It potently
inhibits the JAK3 kinase domain in enzyme assay with Ki
value of 2.5 nM, the potency is about fourfold for JAK3 versus
other three JAK isoforms (JAK1, JAK2, Tyk2). In cell-based
assays, the selectivity of decernotinib is more than 20-fold
for JAK3 over other JAKs subtype [35]. A 12-week phase 2a
clinical trial (NCT01052194) showed that decernotinib was
efficacious in improving clinical signs and symptoms of RA at
dosages of 50–150 mg twice a day as monotherapy in patients
with inadequate response to methotrexate [36]. Another
phase 2b clinical trial (NCT2011-004419-22) reported that
decernotinib significantly improved the signs and symptoms
of RA at weeks 12 and 24 compared with the placebo group
when it was administered in combination with methotrexate
[37]. The current development of decernotinib is presumed to
have been discontinued.
3. Patent assessment of JAK3 inhibitors
3.1. Organization of the review
Patent applications involving JAK inhibitors that were pub-
lished from 2010 to 2015 [38,39] and selective TYK2 inhibitors
that were published in 2019 have been included in a previous
review [40]. Apart from these clinical candidates, additional
structures have been reported in recent scientific publications
and patents. This study focused on publicly available structural
information for patented JAK3 inhibitors, from 2016 to 2021
(1 June 2021). Data from peer-reviewed literature and public
information disclosed by companies were included, where
relevant. Only patents that have been published as WIPO
applications were selected in this study, which was organized
alphabetically by assignee name. Furthermore, a summary and
Table 2. The development status of JAK3 inhibitors in clinic.
No. Compound Target Status R&D company Clinical focus
1 Izencitinib pan-JAK Phase 3 Theravance Biopharma Ulcerative colitis
2 Ritlecitinib JAK3/TEC Phase 3 Pfizer Atopic dermatitis, Vitiligo
3 ATI-1777 JAK1/ JAK3 Phase 2 Aclaris Therapeutics Atopic dermatitis
4 Nezulcitinib JAK1/ JAK2/JAK3 Phase 2 Theravance Biopharma Acute lung injury, COVID-19 complications, Cytokine release
syndrome
5 Ost-122 JAK3/ Tyk2/ NUAK family
kinase 1
Phase 2 Oncostellae Ulcerative colitis
6 Td-8236 pan-JAK Phase 2 Theravance Biopharma Asthma
7 CS-12192 JAK1/ JAK3/ TAK1 Phase 1 Shenzhen Chipscreen
Biosciences
Rheumatoid arthritis
8 R-256 JAK1/ JAK3 Phase 1 Rigel Asthma
9 TD-5202 JAK 3 Phase 1 Theravance Biopharma inflammatory bowel disease
10 Decernotinib JAK3 Discontinued Vertex Pharmaceuticals Rheumatoid arthritis
4C. CHEN ET AL.
analysis including relevant SAR information for the most
potent inhibitors were included.
3.2. Selective JAK3 inhibitors
The selectivity of selective JAK3 inhibitors against other JAK
family subtypes (such as JAK2) is crucial for minimizing potential
side effects and maximizing the desired pharmacological effects.
3.2.1. Aclaris therapeutics
Aclaris Therapeutics disclosed four N-(3-(quinoxalin-2-yl) phenyl)
acrylamide compounds and pharmaceutically acceptable salts
thereof as selective JAK3 inhibitors to be used in the treatment
of JAK3-associated diseases in patent WO2017091681A1 [41].
Compounds14 (Figure 2) all potently inhibit the JAK3 kinase activ-
ity with IC
50
values of 13–18 nM, and their JAK3 selectivity is greater
than 5000-fold than JAK1, JAK2, or TYK2 in biochemical assays.
Compound 2, in particular, is also highly and consistently soluble at
pH 7.4. The compounds also display favorable safety profiles based
on AMES, hERG, and cytotoxicity testing. The in vivo activity of the
compounds 1–4 were investigated in anagen induction hair
growth mode and AA prevention model. The anagen induction
model was used in telogen mouse skin to test the ability of
compounds 1–4 to induce the hair cycle. The results indicated
that Darkening of the skin was observed after approximately 14–
18 days with eruption of new anagen hairs at approximately
28 days after treating with 30 μL 2% of the four different com-
pounds. For prevention of AA in the graft model, C3h/HeJ mice
were grafted with skin from an AA-affected mouse. The tested
JAK3 inhibitors or control was administrated by i.p. injection
(0.5 mg in 50 μL ethylene glycol) daily for 8 weeks. After 6 weeks,
the control treated mice lost their hair as expected in the AA
model. In contrast, the three tested compounds treated mice all
retained their hair.
Figure 2. (A) JAK3 inhibitors from Aclaris Therapeutics. (B) JAK3 inhibitors developed by CAMS and CPU.
EXPERT OPINION ON THERAPEUTIC PATENTS 5
3.2.2. CAMS and China Pharmaceutical University
Chinese Academy of Medical Sciences (CAMS) and China
Pharmaceutical University (CPU) described 28 examples of pyr-
azolo[3,4-d] pyrimidines in the patent WO2020052489 [42]. The
authors designed and synthesized selective JAK3 inhibitors
based on the structural differences between JAK3 subtype with
a cysteine residue at position 909 and other JAK subtypes with
a serine residue at kinase domain. The biochemical inhibitions of
JAKs were provided for all examples. Many of the analogs were
selective JAK3 inhibitors with IC
50
values of less than 1 nM
(compounds 58, see Figure 2). Compound 1 irreversibly binds
to Cys909 residue with a potent activity (IC
50
= 0.1 nM) for JAK3
and over 13,000-fold selectivity than other three JAK subtypes. In
vivo, compound 1 was evaluated in an adjuvant-induced arthritis
(AIA) model. Example 1 exhibited dose-dependent effects at 50
and 100 mg/kg, with tofacitinib as a positive control drug [43].
3.2.3. Pfizer
Prior to 2016, Pfizer researchers modified the non-covalent
pan-JAK inhibitor tofacitinib with the aim of converting it
into a covalent inhibitor [27]. JAK3-selective pyrrolopyrimi-
dines and pyrrolopyrazines were substituted with an acry-
lamide moiety as described in the patent WO2015083028
[44]. Example 5, ritlecitinib was identified as the first orally
active JAK3 inhibitors that achieved JAK isoform specificity
through a covalent interaction with the unique JAK3 residue
Cys909 (Figure 3(a)). Pfizer researchers then performed
further studies on this class of irreversible inhibitors.
Another library of pyrrolo[2,3-d] pyrimidine derivatives was
patented in WO2016178110 [45], the majority of which had
an N-1-acryloylpiperidin-3-yl-amino function on C4, analo-
gously to the previous generation of compounds (Figure 3
(b)). Two of the most active compounds on JAK3 were
compounds 9 and 10, with an IC
50
value of 25 nM or
16 nM at 1 mM ATP concentration. The stability of both
these JAK3 covalent inhibitors in rat and human whole
blood was determined. The compounds were tested for
their ability to inhibit IL-15 induced STAT5 phosphorylation
in peripheral blood mononuclear cells and heparin-treated
human whole blood.
Figure 3. (A) Comparison data of ritlecitinib to tofacitinib is presented in WO2015083028. (B) JAK3 inhibitors from Pfizer in WO2016178110. (C) Aromatic
heterocyclic JAK3 inhibitors from Shenzhen Chipscreen Biosciences Ltd (ND means no data).
6C. CHEN ET AL.
3.2.4. Shenzhen chipscreen biosciences
Shenzhen Chipscreen Biosciences Ltd. has published 123
examples of aromatic heterocyclic compounds [46]. Some
of the compounds were evaluated for enzymatic inhibitory,
cell proliferation, and regulation of signaling pathways
in vitro. Compounds 1113 (Figure 3(c)) exhibited a JAK3
IC
50
of < 21 nM and were the only compounds to have
biological effects on a limited panel of 5 kinases (ITK, BLK,
TBK1, FLT1, FLT4). Findings revealed that compounds 11
and 13 exhibited inhibitory effects against TBK1. In addi-
tion, all three compounds had a good level of specific cell-
inhibitory effect on CTLL-2. Examination of intracellular
inhibitory activity indicated that all three compounds had
intracellular selective JAK3 and/or JAK1 inhibitory effects,
which was consistent with the results of in vitro enzyme
evaluation. Although compounds 1113 showed potent
inhibition of JAK3, the selectivity is about 10-fold for
JAK3 versus for JAK1. Compounds 11 and 12 were active
at 40 mg/kg twice per day in a rat adjuvant-induced arthri-
tis model. Compared to the solvent group, compound 11
could significantly inhibit the degree of arthritis swelling in
rats, and the inhibition rate of each index was greater than
the positive control drug, methotrexate. Compound 11 was
named CS12192 and is reportedly undergoing a phase 1
trial for rheumatoid arthritis [47].
3.2.5. Theravance Biopharma
Theravance Biopharma focuses on the research and devel-
opment of organ selective JAKs inhibitors [48–50].
Approximately 440 examples of pyrazolo and triazolo bicyc-
lic compounds were reported as JAK inhibitors, particularly
as JAK3 inhibitors in enzyme-binding assays, in the patent
WO2019027960 [48]. The disclosed compounds have been
shown to have potent functional activity for JAK3 in cellular
assays by testing IL-2 stimulated pSTAT5 in Tall-1 T cells and
human peripheral blood mononuclear cells (PBMC) CD4
+
T cells. According to the patents, new JAK3 inhibitors that
can be administered orally and achieve therapeutically rele-
vant exposure in the gastrointestinal tract with minimal
systemic exposure were developed. Co-crystal structures
were obtained for compounds 14, 15, and 16 (Figure 4),
with each bound to human JAK3. Twenty compounds,
including compounds 1416, exhibited an efficacy in oxa-
zolone-induced colitis animals at a dose of 3 mg/kg (p.o.,
bid). A murine model with IL-2 induced pSTAT5 in the
thymus showed that compounds 1416 did not significantly
inhibit IL-2 induced pSTAT5, indicating these JAK3 inhibitors
have gut-selective property.
In WO2020154350 [50], the invention is directed to imi-
dazole and triazole containing bicyclic compounds as selec-
tive JAK3 inhibitors. In WO2021108803 [49], fused
Figure 4. Pyrazolo and triazolo bicyclic compounds from Theravance Biopharma.
EXPERT OPINION ON THERAPEUTIC PATENTS 7
pyrimidine pyridinone compounds were disclosed to use as
JAK3 inhibitors. The disclosed 316 compounds displayed
potently inhibitory activity for JAK3 in cellular assays of IL-
2 stimulated pSTAT5 in Tall-1 T cells and human PBMC CD4
+
T cells. In CellTiter-Glo luminescent cell cytotoxicity assay,
the test compounds had less likelihood to cause cytotoxi-
city. The compounds 17–21 (Figure 4) exhibited
a statistically significant decrease in combined stool score
endpoint as compared with vehicle treated animals in the
oxazolone-induced colitis model at a dose of 1 mg/kg (p.
o., bid).
3.3. Nonselective JAK3 inhibitors
This section describes recently patented JAK3 inhibitors that
exhibit some degree of activity against other JAK family mem-
bers. The content was classified by patents application.
3.3.1. Boragen Inc
Boragen Inc has developed several chemical compounds as
JAK inhibitors [51]. Most of the compounds described in this
patent appear to be active against JAK1 and JAK2. But accord-
ing to the provided JAK enzyme data, compounds 22, 23, and
24 (Figure 5(a)) also displayed potent inhibition activity for
JAK3 with IC
50
value of 0.66 nM, 1.2 nM, and 0.29 nM, respec-
tively. The cellular assay also showed that 21, 22 and 23
obviously inhibited the expression of the pSTAT6 induced by
IL-4 in peripheral blood mononuclear cell and the pSTAT5
induced by GM-CSF in human whole blood.
3.3.2. Celon pharma
Celon Pharma has developed novel pyrazolo[1,5-a] pyrimi-
dine derivatives with more potent JAK1/JAK3 kinases inhibi-
tion than kinase JAK2 inhibition (compounds 25–27, Figure 5
(b)) [52]. Cellular assays using IL-3 (JAK2 activation) or IL-4
(JAK1/JAK3 activation) stimulation of TF-1 cells were con-
ducted to measure the inhibition of cell viability.
Furthermore, assays for TNF-α inhibition and IFN-γ produc-
tion by T lymphocytes were described. Compounds 25, 26,
and 27 were also reported to inhibit in vitro STAT6
phosphorylation.
Figure 5. (A) JAK inhibitors from Boragen Inc. (B) JAK inhibitors from Celon Pharma in patent WO2018206739. (C) Pyrrolopyrimidine five-membered azacyclic from
Huadong Medicine Co., Ltd.
8C. CHEN ET AL.
3.3.3. Huadong Medicine Co., Ltd
Huadong Medicine Co., Ltd. developed a pyrrolopyrimidine five-
membered azacyclic derivative as a novel JAK kinase inhibitor
[53]. Only two compounds, 28 and 29 (Figure 5(c)) similar to
barictinib in structure, were published that they inhibited JAK3
with IC
50
of 27 nM and 31 nM, respectively. In the rat AIA model,
compound 28 showed a 100% inhibition rate at 10 mg/kg.
Administration of compounds 28 and 29 once daily at 30 mg/
kg for 14 days was well tolerated in rat toxic side effects tests.
3.3.4. Jiangsu Hansoh Pharmaceutical Group Co., Ltd
Jiangsu Hansoh Pharmaceutical Group Co., Ltd. described 319
examples of heteroaromatic derivatives [54]. Most of the
examples showed that compounds exhibited strong inhibitory
activity against JAK1 and JAK2. Several analogs, compounds
30–35 with a 7–8 membered bridge heterocyclic group, dis-
played in Figure 6(a) were very potent JAK3 inhibitors with
IC
50
values of less than 10 nM. In IFN-α-stimulated cellular
assay, compounds of 3035 all significantly inhibited the
phosphorylation of STAT3 (with IC
50
from 0.29 to 11.05 nM),
the downstream effector molecule of JAKs. The PK of com-
pounds 30–35 in Balb/c mice was also determined, with
results revealing good exposure levels in the colon and
ileum. In the DSS-induced C57BL/6 mouse colitis model, com-
pounds 30–35 significantly reduced the daily disease index
(DAI) and had obvious drug effects.
Figure 6. (A)Heteroaromatic derivatives from Jiangsu Hansoh Pharmaceutical Group Co Ltd. (B) Heteroaromatic derivatives from Jiangsu Vcare Pharmatech Co Ltd.
EXPERT OPINION ON THERAPEUTIC PATENTS 9
3.3.5. Jiangsu Vcare Pharmatech Co., Ltd
Jiangsu Vcare Pharmatech Co., Ltd. developed 23 com-
pounds with a 7-azaindole core [55]. A minimal amount of
structure–activity relationship (SAR) was conducted at the
C-1 and C-6 positions of pyridine. Data for the biochemical
inhibition of JAKs was published, with most compounds
shown to exhibit IC
50
values of < 10 nM. However, no
cellular data was reported. There were two developed com-
pounds, 36 and 37 (Figure 6(b)), that had significant inhibi-
tory effects in DSS-induced colitis in mice at 5 mg/kg.
Furthermore, both compounds had superior efficacy than
the positive control drug tofacitinib at the same dose. In
addition, compound 37 displayed a significant inhibitory
effect on rat DNBS induced colitis at a lower dose of
1 mg/kg.
3.3.6. Nanchang Helioeast Pharmaceutical Co., Ltd
Nanchang Helioeast Pharmaceutical Co Ltd. has filed two
patents for JAK3 inhibitors. A serious of pyrimidine-amino-
imidazol compounds with a range of substituents at posi-
tion N-1 imidazol and C-4 pyrimidine were described in
patent WO2017020428 (compounds 38–42) [56]. And
4,7-diamino-pyrido[2,3-d] pyrimidine derivative JAK inhibitor
was described in patent WO201788289 (compounds 4345,
Figure 7) [57]. In vitro enzymatic-level assay, compounds
3744 were all displayed potent inhibitory activity for
JAK3 with IC
50
of < 20 nM. In cellular assays, compounds
3845 inhibited STAT5 phosphorylation by IC
50
value in the
200–2000 nM rang in IL-3-induced TF-1 cells. In IL-4-induced
THP-1 cells, they inhibited STAT6 phosphorylation by IC
50
value of < 200 nM. In vivo collagen-induced mouse
Figure 7. JAK family kinase inhibitors from Nanchang Helioeast Pharmaceutical Co Ltd.
10 C. CHEN ET AL.
rheumatoid arthritis (CIA) model assay, the selected com-
pounds were all significantly reduced the clinical inflamma-
tory symptom scores.
3.3.7. Novartis
Novartis developed novel diamino pyridine derivatives exhibit-
ing JAK modulating properties [58]. Only two compounds, 46
and 47 (Figure 8(a)), that inhibited the JAK enzyme were
disclosed. There were no reported cellular and in vivo data.
3.3.8. Shenzhen Amazing Genetech Co., Ltd
Shenzhen Amazing Genetech Co., Ltd. developed 416 pyr-
rolo pyridine derivatives as selective inhibitors of JAK1
[59]. According to the kinase data, several compounds
displayed JAK3 inhibition effects. The majority of the com-
pounds that inhibited JAK3 activity had a pyrrolo substi-
tuent at C-5 pyridine and a range of substituents at C-4
pyridine. The selected activities of the compounds
(Figure 8(b)), including 48, were investigated in a mice
CIA model and exhibited an obvious therapeutic effect
on mouse arthritis. The compounds were added to an
emulsifier to form a 2% ointment, with results showing
that compounds 48 and 49 have obvious hair growth-
promoting effects in mice assay.
3.3.9. Takeda Pharmaceutical Co., Ltd
A patent of Takeda Pharmaceutical Co., Ltd. described hetero-
cyclic compounds as JAK inhibitors [60]. The JAK inhibition
percentages at 100 nM were provided for all 23 examples,
Figure 8. (A) Inhibitors of JAK family kinases from Novartis.(B) Pyrrolo pyridine derivatives from Shenzhen Amazing Genetech Co Ltd. (C) Heterocyclic compounds
from Takeda Pharmaceutical Co Ltd.
EXPERT OPINION ON THERAPEUTIC PATENTS 11
with most compounds exhibiting > 95% inhibition.
Compounds 50–52 (Figure 8(c)) were the more potent exam-
ples of JAK3 inhibitors. There were no reported cellular and
in vivo data.
3.3.10. Theravance Biopharma
3.3.10.1. Gut-selective JAK inhibitor. Theravance
Biopharma has filed several patents for gut-selective JAKs
inhibitors. In 2016, Theravance Biopharma described
naphthyridine compounds as JAKs inhibitors in patent
WO2016191524 [61]. In biochemical assays, compounds 53
56 displayed Ki values < 100 nM for JAK3 (Figure 9(a)). The oral
bioavailability of compounds 53 and 54 were all less than 5%
in rats, and the concentration of 53 in the colon is 450 times
higher than in the plasma. The results show that these com-
pounds had gut-selective properties. Compounds 52, 53, and
56, also exhibited a significant decrease in DAI score in the
oxazolone model as compared to the vehicle-treated animals
at 1, 3, and/or 10 mg/kg (p.o., bid). There were no effects of
B and T cell populations on the immunosuppressive properties
Figure 9. (A) Gut-selective JAK inhibitors from Theravance Biopharma. (B) Skin-selective JAK inhibitors from Theravance Biopharma.
12 C. CHEN ET AL.
of mouse splenic natural killer (NK) with the treatment of
compound 53 at doses of up to 100 mg/kg (p.o., bid). In
a human study, compound 53 was evaluated for safety, toler-
ability, and PK in healthy subjects. Compound 53 has now
been named TD-1473 or izencitinib, a recent phase 2b, multi-
center, randomized, double-blind, vehicle-controlled, parallel-
group clinical trial in patients with ulcerative colitis showed
that izencitinib had not significant difference at any dose in
the change in total score at week 8 relative to placebo
(NCT03758443) [62]. In patent WO2017189822 [63],
a pyrimidine with heterocyclyl containing 4 to 6 ring atoms,
including one nitrogen atom, was described as a JAKs kinase
inhibitor. The structure of the potent JAK3 inhibitor, com-
pound 57, is shown in Figure 9(a).
3.3.10.2. Skin-selective JAK inhibitor. Theravance
Biopharma has also filed two further patents for compounds
with similar structures to those described in patent
WO2017189822 [63]. The compounds described in both patents
possess advantageous solubility properties in aqueous and/or
organic excipients that facilitate formulation into topical
compositions. There were 10 ester and carbonate pyrimidine
compounds described in patent WO2020219639 [64]. The 46
pyrimidine compounds described in patent WO2020219640
[65] were expected to sustain dermal levels in the absence of
significant systemic levels with potent local anti-inflammatory
and anti-pruritic activity in the skin without systemically driven
adverse effects. Compounds 5864 (Figure 9(b)) potently inhib-
ited the activity of JAK1/JAK2/JAK3/Tyk2 with Ki value < 1 nM in
biochemical assays. In cellular assays, 5864 showed significant
inhibitory effect on STAT5 phosphorylation induced by IL-2 with
pIC
50
≥ 8.0. In metabolic stability assay, compound 58 exhibited
an HLM Clint of 132 µL/min/mg, compound 61 exhibited over
2500 µL/min/mg, and compound 62 exhibited 1250–3000 µL/
min/mg in a human liver microsome. As topical skin application
compounds, the PK of the epidermal, dermal, and plasma in the
ointment (0.25%) were determined following 24 hours of topical
exposure to intact male rat skin. Compounds 61 exhibited
a conversion over 30% in this assay.
3.3.10.3. Lung-selective JAK inhibitor. Since 2017,
Theravance Biopharma has claimed a series of fused imidazo-
Figure 10. Lung -selective JAK inhibitors from Theravance Biopharma.
EXPERT OPINION ON THERAPEUTIC PATENTS 13
piperidine compounds with a core structure of indazol-
4,5,6,7-tetrahydro-imidazol-pyridine as JAKs inhibitors for the
treatment of respiratory diseases. In patent WO2017079205
[66], compounds 65–67 (Figure 10) exhibited potent inhibitory
activity against JAK3 with pKi IC
50
≥ 10 nM in biochemical
assay and with pIC
50
8 nM in cellular assay. In patent
WO2018165392 [67] and WO2018165395 [68], the compounds
were described as having the same core structure as com-
pounds in patent WO2017079205, exemplified by compounds
6872 as JAKs inhibitors with Ki < 0.25 nM for JAK3 and <
0.1 nM for JAK1.
In 2020, Theravance Biopharma filed patent
WO2020051105 for new series of dimethyl amino azetidine
amides compounds [69]. Compounds 73 and 74 (Figure 11(a))
possessed JAK3 inhibitory activity with pKi IC
50
values of 10.2
and 10.1 nM, respectively. Compound 73 also exhibited good
PK properties in the plasma and lung, as well as good meta-
bolic stability in human lung S9. In human 3D airway cultures
derived from asthmatic donors, compound 73 inhibited spon-
taneous periostin and IL-6 secretion by 62% ± 25 (at 10 mM)
and 91% ± 9.0 (at 10 mM), respectively, when compared to
vehicle. Assays of lung s9 metabolism and PK in mice plasma
and lungs demonstrated that the compounds exhibited expo-
sure in lungs that is one to two orders of magnitude greater
than exposure in plasma in mice. The compounds caused IL-
13 induced pSTAT6 activation and alternata-induced eosino-
philic inflammation of the murine lung tissue. Furthermore,
the compounds inhibited bronchoalveolar lavage fluid
Figure 11. (A) Lung -selective JAK inhibitors from Theravance Biopharma. (B) JAK inhibitors from Topivert Pharma Ltd.
14 C. CHEN ET AL.
eosinophil counts 48 hours after alternaria challenge.
Compound 73, also named as TD-0903 or nezulcitinib, was
performed a phase2 randomized, double-blind, placebo-
controlled clinical trial (NCT04402866) to treat symptomatic
acute lung injury associated with COVID-19 [70]. The com-
pleted clinical trial showed that the study did not meet the
primary endpoint (Respiratory Failure-Free Days, RFDs) from
randomization htrough day 18 in the intent-to-treat (ITT)
population [71]. Although inhaled nezulcitinib had no statis-
tically significant difference in RFDs from randomization
through Day 28 between nezulcitinib and placebo in ITT,
nezulcitinib demonstrated a favorable trend in improvement
when compared to placebo for 28-day all-cause mortality
rate [72].
Patents WO2020051135 [73] and WO2020051139 [74]
claimed 13 compounds and some of their fluorine substituted
compounds o treat respiratory diseases. Compounds 7577
(Figure 11(a)) possessed JAK3 inhibitory activity with pKi IC
50
values of < 10 nM. In IL-13 induced mouse model, 7577
significantly inhibited the expression of pSTAT6 in lung tissue.
In mice, the lung-to-plasma ratio at 5 hours revealed that
compounds 7577 exhibited significantly more exposure in
the lung than in plasma. The potency (pIC
50
) values of com-
pounds 7577 were all less than 7.5 for the inhibition of
thymic stromal lymphopoietin (TSLP) evoked thymus and acti-
vation-regulated chemokine (TARC) release in hPBMC.
3.3.11. Topivert Pharma Ltd
Topivert Pharma Ltd. has filed two patents on JAKs inhibitors
[75,76]. A total of 47 indazole substituents with similar struc-
tures to the phase I discontinued pan-JAK inhibitor
PF06263276 (compounds 78, Figure 11(b)) were disclosed
[77]. It was surprisingly discovered that compounds bearing
certain aminoheteroaryl substituents inhibited one or more
JAK enzymes, thus possessing good anti-inflammatory
Figure 12. (A) Tricyclic compounds in patent WO2021043850 of Universität Bern. (B) JAK inhibitors from Vimalan Biosciences Inc.
EXPERT OPINION ON THERAPEUTIC PATENTS 15
properties. The chemotype consisted of a central biaryl com-
posed of an indazole and phenyl ring, with the other ring
being tetrahydro imidazole-pyridine. Compounds 79–81
(Figure 11(b)) were all potent JAK3 inhibitors with IC
50
< 0.1 nM. Compound 79 was substantially more potent than
PF06263276 in inhibiting IFN-γ release from CD3/IL2-
stimulated PBMC cells and/or potentially displaying enhanced
viability in the Jurkat cell cytotoxicity assay.
3.3.12. Universität Bern
Universität Bern described two tricyclic compounds of
KMC420 (compound 82) and KMC423 (compound 83,
Figure 12(a)) in patent WO2021043850 [78]. Compound 83
showed potent inhibitory activity against JAK3 with 9 nM of
IC
50
value, and selectivity is 3.5-fold, 6-fold, and 20-fold for
JAK1, JAK2, and Tyk2, respectively. Now, there is no further
information available about this patented compound.
3.3.13. Vimalan Biosciences Inc
Vimalan Biosciences Inc. has published three patents describ-
ing pyrrolo pyrimidin compounds. Much of the peripheral SAR
has remained constant: for example, the pyrimidine is gener-
ally substituted at the C-2 position with an N-pyrazole amide.
The selected compounds 8490 (Figure 12(b)) were evaluated
for their ability to inhibit JAK3 with IC
50
< 100 Nm. The IL-2
(JAK1/JAK3-dependent) stimulation and measurement of
STAT5 phosphorylation cellular assays were performed on
PBMCs. Meanwhile, GM-CSF (JAK1/Tyk2 or JAK2-dependent)
stimulation and measurement of STAT1/5 phosphorylation
cellular assays were performed in hWBs. However, there were
no reported data in patents WO2021026465 [79] and
WO2020227563 [80]. Cellular data was disclosed for com-
pounds 89 and 90 in patent WO2021062036 [81].
4. Conclusion
Over the past 5 years, many patent applications for JAK3
inhibitors have been filed. In order to avoid the side effects
caused by non-selectivity, selective JAK inhibitors are the
direction of the new generation of JAK inhibitors develop-
ment. Although there are many JAK inhibitors being
approved or in development, no truly selective JAK3 inhibi-
tion is approved and only a few are in preclinical or clinical
development. Ritlecitinib, developed by Pfizer, is under-
going several clinical trials in multiple indications. Recently
completed a phase 2a randomized, placebo-controlled study
to evaluate the efficacy and safety clinical trial for AA
showed that ritlecitinib treatment was efficacious and gen-
erally well tolerated at 24 weeks. TD-5202 also a JAK3-
selective inhibitor was developed by Theravance
Biopharma, an interesting is that it was first designed as
gut-targeted selective JAK3 inhibitor to specially treat
inflammatory bowel disease [34].
5. Expert opinion
In view of the side effects caused by nonselective inhibiting
JAKs in clinic, such as infection, hematological and cardiovas-
cular effects, and malignancies [82,83], there is increasing
research focus on selective JAK inhibitors to overcome these
issues. For four isoforms of JAK kinase, JAK3 is predominantly
expressed in hematopoietic cells against to extensive expres-
sion of other three subtypes. JAK3 possess a unique Cys909
residue at kinase domain, and the residue replaced by a serine
in equivalent position in the other three JAK isoforms. The
specificity of JAK3 isoform made it a potentially ideal target for
discovering selective JAK3 inhibitors [84].
The 909-cysteine residue of JAK3 allows for the develop-
ment of a selective JAK3 inhibitor over other JAK family
members. Compounds containing electrophilic warheads
are capable of forming a covalent bond with the nucleo-
philic thiol of the cysteine residue. However, further studies
are required to understand whether a covalent JAK3 inhibi-
tor can be used as an effective disease treatment. The dual
activity of ritlecitinib against JAK3 and the TEC kinase family
may provide a beneficial inhibitory profile and generally
well tolerated during 24-week therapeutic intervention
[85]. TEC family kinase consists of five members (Bruton’s
tyrosine kinase (BTK), bone marrow tyrosine kinase on chro-
mosome X (BMX), interleukin 2-inducible T cell kinase (ITK),
resting lymphocyte kinase (RLK), and TEC). The TEC kinase
family of tyrosine kinases is primarily expressed in immune
cells, and activated TEC kinase by a variety of signals are
involved in signal transduction pathways regulating various
immunological processes in health [86]. The clinical trials of
ritlecitinib showed positive efficacy on RA and AA, and no
treatment-related serious AEs, severe AEs, or deaths were
reported. The efficacy and safety of JAK3-selective inhibitor
as an ideal target need to be further confirmed by more
clinical trials.
Although the JAK3 subtype has the appeal of a target that
is functionally different from other JAK subtypes, no selective
JAK3 inhibitor has yet been approved for the treatment of
inflammatory and autoimmune diseases. Although further
work is required to understand the specific mechanism by
which JAK3 signaling is inhibited and the therapeutic potential
of targeting JAK3, ongoing clinical and pre-clinical trials of
selective JAK3 inhibitors may provide insight and therapeutic
advantages.
Declaration of interests
The authors have no relevant affiliations or financial involvement with any
organization or entity with a financial interest in or financial conflict with
the subject matter or materials discussed in the manuscript. This includes
employment, consultancies, honoraria, stock ownership or options, expert
testimony, grants or patents received or pending, or royalties.
Reviewer disclosures
Peer reviewers on this manuscript have no relevant financial or other
relationships to disclose.
Funding
This work was supported by the National Natural Science Foundation of
China (81973338, 82104189).
16 C. CHEN ET AL.
References
Papers of special note have been highlighted as either of interest (•) or of
considerable interest (••) to readers.
1. Villarino AV, Kanno Y, O’Shea JJ. Mechanisms and consequences of
Jak-STAT signaling in the immune system. Nat Immunol. 2017;18
(4):374–384.
2. Shuai K, Liu B. Regulation of JAK-STAT signalling in the immune
system. Nat Rev Immunol. 2003;3(11):900–911.
•• (References: 1&2) Comprehensive review of JAKs-STAT physiol-
ogy and pathophysiology
3. Rane SG, Reddy EP. JAK3: a novel JAK kinase associated with
terminal differentiation of hematopoietic cells. Oncogene. 1994;9
(8):2415–2423.
4. Geokden ER, Argiriadi MA, Banach DL, et al. Tricyclic covalent
inhibitors selectively target JAK3 through an active site thiol.
J Biol Chem. 2015;290(8):4573–4589.
5. Liu Q, Sabnis Y, Zhao Z, et al. Developing irreversible inhibitors of
the protein kinase cysteinome. Chem Biol. 2013;20(2):146–159.
6. Russell SM, Tayebi N, Nakajima H, et al. Mutation of Jak3 in
a patient with SCID: essential role of Jak3 in lymphoid
development. Science. 1995;270(5237):797–800.
7. Nosaka T, Vandeursen JMA, Tripp RA, et al. Defective lymphoid
development in mice lacking Jak3. Science. 1995;270
(5237):800–802.
8. de Medeiros AKA, Speeckaert R, Desmet E, et al. JAK3 as an emer-
ging target for topical treatment of inflammatory skin diseases.
Plos One. 2016;11(10):e0164080.
9. Casanova JL, Holland SM, Notarangelo LD. Inborn errors of human
JAKs and STATs. Immunity. 2012;36(4):515–528.
10. Clark JD, Flanagan ME, Telliez JB. Discovery and development of
Janus kinase (JAK) inhibitors for inflammatory diseases. J Med
Chem. 2014;57(12):5023–5038.
11. Basquiera AL, Soria NW, Ryser R, et al. Clinical significance of V617F
mutation of the JAK2 gene in patients with chronic myeloprolifera-
tive disorders. Hematology. 2009;14(6):323–330.
12. R R Jr. Janus kinase (JAK) inhibitors in the treatment of inflamma-
tory and neoplastic diseases. Pharmacol Res. 2016;111:784–803.
13. Xu PF, Shen P, Yu B, et al. Janus kinases (JAKs): the efficient
therapeutic targets for autoimmune diseases and myeloprolifera-
tive disorders. Eur J Med Chem. 2020;192:112155.
14. Mesa RA, Yasothan U, Kirkpatrick P. Ruxolitinib. Nat Rev Drug
Discov. 2012;11(2):103–104.
15. Zeiser R, Von Bubnoff N, Butler J, et al. Ruxolitinib for
glucocorticoid-refractory acute graft-versus-host disease. N Engl
J Med. 2020;382(19):1800–1810.
16. Sandborn WJ, Su C, Sands BE, et al. Tofacitinib as induction and
maintenance therapy for ulcerative colitis. N Engl J Med. 2017;376
(18):1723–1736.
17. Lee EB, Fleischmann R, Hall S, et al. Tofacitinib versus methotrexate
in rheumatoid arthritis. N Engl J Med. 2014;370(25):2377–2386.
18. Kalil AC, Patterson TF, Mehta AK, et al. Baricitinib plus remdesivir
for hospitalized adults with COVID-19. N Engl J Med. 2021;384
(9):795–807.
19. Markham A, Keam SJ. Peficitinib: first global approval. Drugs.
2019;79(8):887–891.
20. Talpaz M, Kiladjian JJ. Fedratinib, a newly approved treatment for
patients with myeloproliferative neoplasm-associated
myelofibrosis. Leukemia. 2021;35(1):1–17.
21. van der Heijde D, Song I-H, Pangan IH, et al. Efficacy and safety of
upadacitinib in patients with active ankylosing spondylitis
(SELECT-AXIS 1): a multicentre, randomised, double-blind,
placebo-controlled, phase 2/3 trial. Lancet. 2019;394
(10214):2108–2117.
22. Rubbert-Roth A, Enejosa J, Pangan AL, et al. Trial of upadacitinib or
Abatacept in rheumatoid arthritis. N Engl J Med. 2020;383
(16):1511–1521.
23. Dhillon S. Delgocitinib: first approval. Drugs. 2020;80(6):609–615.
24. Dhillon S, Keam SJ. Filgotinib: first approval. Drugs. 2020;80
(18):1987–1997.
25. Dai ZP, Chen J, Chang YQ, et al. Selective inhibition of JAK3
signaling is sufficient to reverse alopecia areata. JCI Insight.
2021;6(7):e142205.
26. Dai J, Yang L, Addison G. Current status in the discovery of covalent
Janus kinase 3 (JAK3) inhibitors. Mini Rev Med Chem. 2019;19
(18):1531–1543.
27. Telliez JB, Dowty ME, Wang L, et al. Discovery of a JAK3-selective
inhibitor: functional differentiation of JAK3-selective inhibition over
pan-JAK or JAK1-selective inhibition. ACS Chem Biol. 2016;11
(12):3442–3451.
28. Thorarensen A, Dowty ME, Banker ME, et al., Design of a Janus
kinase 3 (JAK3) specific inhibitor 1-(2s,5R)-5-((7H-Pyrrolo[2,3-d]pyr-
imidin-4-yl)amino)-2- methylpiperidin-1-y1)prop-2-en-1-one
(PF-06651600) allowing for the interrogation of JAK3 signaling in
humans. J Med Chem. 60(5): 1971–1993. 2017.
•• Details the discovery of the covalent JAK3 inhibitor PF-
06651600.
29. Xu H, Jesson MI, Seneviratne UI, et al. PF-06651600, a dual JAK3/
TEC family kinase inhibitor. ACS Chem Biol. 2019;14
(6):1235–1242.
30. Robinson MF, Damjanov N, Stamenkovic B, et al., Efficacy and
safety of PF-06651600 (Ritlecitinib), a novel JAK3/TEC inhibitor, in
patients with moderate-to-severe rheumatoid arthritis and an
inadequate response to methotrexate. Arthritis Rheumatol. 72(10):
1621–1631. 2020.
Study showing effectiveness of the most promising JAK3 inhi-
bitor (Ritlecitinib) in clinical trials.
31. King B, Guttman-Yassky E, Peeva E, et al. A phase 2a rando-
mized, placebo-controlled study to evaluate the efficacy and
safety of the oral Janus kinase inhibitors ritlecitinib and brepo-
citinib in alopecia areata: 24-week results. J Am Acad Dermatol.
2021;85(2):379–387.
32. Aclaris Therapeutics Completes Enrollment in its Phase 2a Clinical
Trial of ATI-1777 for Moderate to Severe Atopic Dermatitis [cited
2021 March 15]. Available from https://www.globenewswire.com/
en/news-release/2021/03/15/2192631/37216/en/Aclaris-
Therapeutics-Completes-Enrollment-in-its-Phase-2a-Clinical-Trial-of
-ATI-1777-for-Moderate-to-Severe-Atopic-Dermatitis.html
33. Aclaris Therapeutics Completes Enrollment in its Phase 2a
Clinical Trial of ATI-1777 for Moderate to Severe Atopic
Dermatitis [cited 2021 June 08]. Available from https://www.
globenewswire.com/en/news-release/2021/06/08/2243460/
37216/en/Aclaris-Therapeutics-Announces-Positive-Preliminary-
Topline-Data-from-Phase-2a-Trial-of-ATI-1777-for-Moderate-to-
Severe-Atopic-Dermatitis.html
34. Theravance Biopharma Pipeline [cited 2020 July 20]. Available from
https://www.theravance.com/our-pipeline.
35. Elwood F, Witter DJ, Piesvaux J, et al. Evaluation of JAK3 biology in
autoimmune disease using a highly selective, irreversible JAK3
inhibitor. J Pharmacol Exp Ther. 2017;361(2):229–244.
36. Genovese MC, van Vollenhoven RF, Pacheco-Tena C, et al. VX-509
(Decernotinib), an oral selective janus kinase 3 inhibitor, in combi-
nation with methotrexate in patients with rheumatoid arthritis.
Arthritis Rheumatol. 2016;68(1):46–55.
37. Fleischmann RM, Damjanov NS, Kivitz AJ, et al. A randomized,
double-blind, placebo-controlled, twelve-week, dose-ranging
study of decernotinib, an oral selective JAK-3 inhibitor, as mono-
therapy in patients with active rheumatoid arthritis. Arthritis
Rheumatol. 2015;67(2):334–343.
38. Kettle JG, Åstrand A, Catley M, et al. Inhibitors of JAK-family kinases:
an update on the patent literature 2013-2015, part 1. Expert Opin
Ther Pat. 2017;27(2):127–143.
39. Kettle JG, Åstrand A, Catley M, et al. Inhibitors of JAK-family kinases:
an update on the patent literature 2013-2015, part 2. Expert Opin
Therc Pat. 2017;27(2)145–161.
History of reported JAKs inhibitors patents filed before
the year 2016
40. He X, Chen X, Zhang H, et al., Selective Tyk2 inhibitors as potential
therapeutic agents: a patent review (2015–2018). Expert Opin Ther
Pat. 29(2): 137–149. 2019.
EXPERT OPINION ON THERAPEUTIC PATENTS 17
Information on the TYK2 inhibitors patents filed between the
years 2015-2018.
41. Aclaris Therapeutics. Selective kinase inhibitors. WO2017091681A1.
42. China Pharmaceutical University, Institute of Mataria Medica,
Chinese Academy of Medical Sciences & Peking Union Medical
College. Preparation for 6-amino-1h-pyrazolo[3,4-d]pyrimidine-
based jak kinase inhibitor and application thereof.
WO2020052489A1.
43. Yin Y, Chen CJ, Yu RN, et al. Novel 1H-pyrazolo[3,4-d]pyrimidin-
6-amino derivatives as potent selective Janus kinase 3 (JAK3) inhi-
bitors. Evaluation of their improved effect for the treatment of
rheumatoid arthritis. Bioorg Chem. 2020;98:103720.
44. Pfizer Inc. Pyrrolo[2,3-d]pyrimidinyl, pyrrolo[2,3-b]pyrazinyl and
pyrrolo[2,3-d]pyridinyl acrylamides. WO2015083028A1.
45. Pfizer Inc. Pyrrolo [2, 3-D] pyrimidinyl, pyrrolo [2, 3-B] pyrazinyl,
pyrrolo [2, 3-B] pyridinyl acrylamides and epoxides thereof.
WO2016178110A1.
46. Shenzhen chipscreen biosciences, ltd. Preparation method for aro-
matic heterocyclic compound used as selective jak3 and/or jak1
kinase inhibitor and application of aromatic heterocyclic
compound. WO2016041472A1.
47. Shan S, Zhou Y, Yu JD, et al. Therapeutic treatment of a novel
selective JAK3/JAK1/TBK1 inhibitor, CS12192, in rat and mouse
models of rheumatoid arthritis. Int Immunopharmacol.
2019;77:105914.
48. Theravance biopharma. Pyrazolo and triazolo bicyclic compounds
as jak kinase inhibitors. WO2019027960A1.
49. Theravance biopharma. Fused pyrimidine pyridinone compounds
as jak inhibitors. WO2021108803A1.
50. Theravance biopharma. Imidazo[1,5-a]pyridine, 1,2,4-triazolo[4,3-a]
pyridine and imidazo[1,5-a]pyrazine as jak inhibitors.
WO2020154350A1.
51. Boragen, Inc. Chemical compounds. WO2020232470A1.
52. Celon pharma. Pyrazole[1,5-a]pyrimidine derivatives as kinase jak
inhibitors. WO2018206739A1.
53. Huadong Medicine Co., Ltd. Pyrrolopyrimidine five-membered
azacyclic derivative and application thereof. WO2017129116A1.
54. Jiangsu hansoh pharmaceutical group Co., Ltd. Heteroaromatic
derivatives for use as regulator, preparation method therefor and
use thereof. WO2020108613A1.
55. Jiangsu vcare pharmatech co., ltd. Janus kinase (jak) family inhibitor,
preparation of same, and applications thereof. WO2020156271A1.
56. Nanchang Helioeast Pharmaceutical Co., Ltd. Novel compounds for
use as jak inhibitors. WO2017020428A1.
57. Nanchang Helioeast Pharmaceutical Co., Ltd. 4,7-diamino-pyrido
[2,3-d] pyrimidine derivative for use as jak inhibitor.
WO2017088289A1.
58. Novartis. Diamino pyridine derivatives. WO2017089985A1.
59. Shenzhen Amazing Genetech Co., Ltd. Pyrrolo[2,3-b]pyridine deri-
vatives as selective inhibitors of janus kinase 1. WO2021000785A1.
60. Takeda Pharmaceutical Company Limited. Heterocyclic compound.
WO2019069844A1.
61. Theravance biopharma. Naphthyridine compounds as jak kinase
inhibitors. WO2016191524A1.
62. Theravance Biopharma Pipeline [cited 2021 August 23]. Available
from https://investor.theravance.com/static-files/3361c656-0a21-
416e-8997-1d65725affea
63. Theravance biopharma. Pyrimidine compounds as jak kinase
inhibitors. WO2017189822A1.
64. Theravance Biopharma. Ester and carbonate pyrimidine com-
pounds as jak kinase inhibitors. WO2020219639A1.
65. Theravance Biopharma. Pyrimidine jak inhibitors for the treatment
of skin diseases. WO2020219640A1.
66. Theravance Biopharma. Jak kinase inhibitor compounds for treat-
ment of respiratory disease. WO2017079205A1.
67. Theravance Biopharma. Jak inhibitors containing a 4-membered
heterocyclic amide. WO2018165392A1.
68. Theravance Biopharma. Fused imidazo-piperidine jak inhibitors.
WO2018165395A1.
69. Theravance Biopharma. Dimethyl amino azetidine amides as jak
inhibitors. WO2020051105A1.
70. Pfeifer ND, Lo A, Bourdet DL, et al. Phase 1 study in healthy
participants to evaluate safety, tolerability and pharmacokinetics
of inhaled nezulcitinib, a potential treatment for COVID-19. Clin
Transl Sci. 2021;14(6):2556–2565.
71. Singh D, Bogus M, Moskalenko V, et al. A phase 2 multiple
ascending dose study of the inhaled pan-JAK inhibitor nezulciti-
nib (TD-0903) in severe COVID-19. Eur Respir J. 2021;58
(4):2100673.
72. Theravance Biopharma Pipeline [cited 2021 June 21]. Available
from https://investor.theravance.com/news-releases/news-release-
details/theravance-biopharma-inc-announces-top-line-results-
phase-2
73. Theravance Biopharma. Process for preparing jak inhibitors and
intermediates thereof. Clin Transl Sci. 2021; WO2020051135A1.
accepted manuscript, 10.1111/cts.13123
74. Theravance Biopharma. 5 to 7 membered heterocyclic amides as
jak inhibitors. WO2020051139A1.
75. Topivert Pharma Limited. 4, 5, 6, 7-tetrahydro-1h-imidazo[4, 5-c]
pyridine and 1, 4, 5, 6, 7, 8-hexahydroimidazo[4, 5-d]azepine deri-
vatives as janus kinase inhibitors. WO2017077283A1.
76. Topivert Pharma Limited 4, 5, 6, 7-tetrahydro-1h-imidazo[4, 5-c]
pyridine and 1, 4, 5, 6, 7, 8-hexahydroimidazo[4, 5-d]azepine deri-
vatives as janus kinase inhibitors. WO2017077288A1.
77. Jones P, Storer RI, Sabnis YA, et al. Design and synthesis of a
Pan-Janus Kinase inhibitor clinical candidate (PF-06263276) suita-
ble for inhaled and topical delivery for the treatment of inflamma-
tory diseases of the lungs and skin. J Med Chem. 2017;60
(2):767–786.
78. Universität Bern. Tricyclic janus kinase (jak) inhibitors and their
use in the treatment of autoimmune diseases.
WO2021043850A1.
79. Vimalan Biosciences Inc. Jak inhibitors. WO2021026465A1.
80. Vimalan Biosciences Inc. Jak inhibitors. WO2021062036A1.
81. Vimalan Biosciences Inc. Jak inhibitors. WO2020227563A1.
82. Winthrop KL. The emerging safety profile of JAK inhibitors in
rheumatic disease. Nat Rev Rheumatol. 2017;13(4):234–243.
83. Changelian PS, Flanagan ME, Ball DJ, et al. Prevention of organ
allograft rejection by a specific Janus kinase 3 inhibitor. Science.
2003;302(5646):875–878.
84. Smith GA, Uchida K, Weiss A, et al. Essential biphasic role for JAK3
catalytic activity in IL-2 receptor signaling. Nat Chem Biol. 2016;12
(5):373–379.
85. Xu H, Jesson MI, Seneviratne UI, et al. PF-06651600, a dual JAK3/
TEC family kinase inhibitor. Acs Chem Biol. 2019;14(6):1235–1242.
86. Berg LJ, Finkelstein LD, Lucas JA, et al. Tec family kinases in
T lymphocyte development and function. Annu Rev Immunol.
2005;23(1):549–600.
18 C. CHEN ET AL.
... In summary, apart from the two major factors of unchangeable heredity and careless bite, the remaining major influencing factors can be classified into three aspects: nutrition, immunity, Gels 2023, 9,659 3 of 19 motifs, have garnered significant interest for their potential as bioactive compounds and pharmaceuticals. Currently, common α,α-difluoromethyl carbinols drugs include gemcitabine [29], GABAB receptor agonist [30], JAK3 inhibitors [31], etc. These drugs all have good anti-inflammatory activity. ...
... Compounds that contain difluoromethyl, particularly those with α,α-difluoromethyl carbinols and structural motifs, have garnered significant interest for their potential as bioactive compounds and pharmaceuticals. Currently, common α,α-difluoromethyl carbinols drugs include gemcitabine [29], GABAB receptor agonist [30], JAK3 inhibitors [31], etc. These drugs all have good anti-inflammatory activity. ...
Article
Full-text available
Oral ulcer is a common inflammatory disease of oral mucosa, causing severe burning pain and great inconvenience to daily life. In this study, compound 3J with anti-inflammatory activity was synthesized beforehand. Following that, an intelligent composite hydrogel supported 3J was designed with sodium alginate, carboxymethyl chitosan, and chitosan quaternary ammonium salt as the skeleton, and its therapeutic effect on the rat oral ulcer model was investigated. The results show that the composite hydrogel has a dense honeycomb structure, which is conducive to drug loading and wound ventilation, and has biodegradability. It has certain antibacterial effects and good anti-inflammatory activity. When loaded with 3J, it reduced levels of TNF-α and IL-6 in inflammatory cells by up to 50.0%. It has excellent swelling and water retention properties, with a swelling rate of up to 765.0% in a pH 8.5 environment. The existence of a large number of quaternary ammonium groups, carboxyl groups, and hydroxyl groups makes it show obvious differences in swelling in different pH environments, which proves that it has double pH sensitivity. It is beneficial to adapt to the highly dynamic changes of the oral environment. Compared with single hydrogel or drug treatment, the drug-loaded hydrogel has a better effect on the treatment of oral ulcers.
... 63 Goedken et al. aimed to leverage a unique cysteine residue on JAK3 (Cys909), which would confer unique reactivity relative to JAK1, JAK2, and TYR2 (which contain serine at the equivalent position) which makes covalent inhibition a particularly attractive approach. 63,64 Using a small collection of newly developed ICI (1−3), a known JAK3 inhibitor thought to be an ICI (4), and two previously known noncovalent JAK3 inhibitors (5 and 6) (Figure 7), Goedken et al. characterized and determined that the newly developed ICIs had excellent activity (low nM) and selectivity (>500×) for JAK3 versus other JAK kinases. 63 Additionally, for other highly similar kinases with cysteine residues at the equivalent position as JAK3, there was also high selectivity (>100×). ...
Article
Full-text available
Covalent inhibition has seen a resurgence in the last several years. Although long-plagued by concerns of off-target effects due to nonspecific reactions leading to covalent adducts, there has been success in developing covalent inhibitors, especially within the field of anticancer therapy. Covalent inhibitors can have an advantage over noncovalent inhibitors since the formation of a covalent adduct may serve as an additional mode of selectivity due to the intrinsic reactivity of the target protein that is absent in many other proteins. Unfortunately, many covalent inhibitors form irreversible adducts with off-target proteins, which can lead to considerable side-effects. By designing the inhibitor to form reversible covalent adducts, one can leverage competing on/off kinetics in complex formation by taking advantage of the law of mass action. Although covalent adducts do form with off-target proteins, the reversible nature of inhibition prevents accumulation of the off-target adduct, thus limiting side-effects. In this perspective, we outline important characteristics of reversible covalent inhibitors, including examples and a guide for inhibitor development.
... Each has unique traits and distinct cellular roles. Specifically, JAK1 is associated with signaling via the interferon-α receptor, while JAK3 primarily operates through the common gamma chain receptor [19][20][21]. JAK2, however, interfaces with a multitude of cytokines, including erythropoietin, thrombopoietin, and interleukin-6 [14,22], positioning it with a more expansive signaling capability compared to JAK1 and JAK3. Notably, JAK2's involvement has been identified in conditions like polycythemia vera, essential thrombocythemia, and myelofibrosis [23]. ...
Article
Full-text available
The increasing utilization of artificial intelligence algorithms in drug development has proven to be highly efficient and effective. One area where deep learning-based approaches have made significant contributions is in drug repositioning, enabling the identification of new therapeutic applications for existing drugs. In the present study, a trained deep-learning model was employed to screen a library of FDA-approved drugs to discover novel inhibitors targeting JAK2. To accomplish this, reference datasets containing active and decoy compounds specific to JAK2 were obtained from the DUD-E database. RDKit, a cheminformatic toolkit, was utilized to extract molecular features from the compounds. The DeepChem framework’s GraphConvMol, based on graph convolutional network models, was applied to build a predictive model using the DUD-E datasets. Subsequently, the trained deep-learning model was used to predict the JAK2 inhibitory potential of FDA-approved drugs. Based on these predictions, ribociclib, topiroxostat, amodiaquine, and gefitinib were identified as potential JAK2 inhibitors. Notably, several known JAK2 inhibitors demonstrated high potential according to the prediction results, validating the reliability of our prediction model. To further validate these findings and confirm their JAK2 inhibitory activity, molecular docking experiments were conducted using tofacitinib—an FDA-approved drug for JAK2 inhibition. Experimental validation successfully confirmed our computational analysis results by demonstrating that these novel drugs exhibited comparable inhibitory activity against JAK2 compared to tofacitinib. In conclusion, our study highlights how deep learning models can significantly enhance virtual screening efforts in drug discovery by efficiently identifying potential candidates for specific targets such as JAK2. These newly discovered drugs hold promises as novel JAK2 inhibitors deserving further exploration and investigation.
... As has been demonstrated, the JAK-STAT pathway is a double-edged sword, whereby the proper degree of activation helps the host get rid of invading pathogens, while further progression of the disease may occur due to overactivation of the pathway 56 . JAK inhibitors can achieve immunosuppression, which is obtained by decreasing the serum pro-inflammatory factor levels, It should be noted that JAKs are also used to treat rheumatoid arthritis, inflammatory bowel disease, tumors, diabetes, and skin-related diseases 57 . All this evidence suggests that the JAK/STAT pathway may be a therapeutic target for sporotrichosis. ...
Article
Full-text available
This study aimed to investigate the molecular mechanism of sporotrichosis and identify possible novel therapeutic targets. Total RNA was extracted from skin lesion samples from sporotrichosis patients and used to construct a long-chain RNA transcriptome library and miRNA transcriptome library for whole transcriptome sequencing. The differentially expressed genes (DEGs) between the groups were identified, and then Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and Gene Set Enrichment Analysis enrichment analyses were performed based on the DEGs. An lncRNA-miRNA-mRNA ceRNA network was constructed. The expressions of JAK/STAT pathway-related proteins were detected in the patient and control tissues using RT-qPCR and Western blot analysis. Enrichment analysis showed that the DEGs were mainly enriched in various infectious diseases and immune response-related signaling pathways. Competing endogenous RNA network analysis was performed and identified the hub lncRNAs, miRNAs, and mRNAs. Compared with the control group, the mRNA expressions of SOCS3, IL-6, and JAK3 were significantly upregulated, while the expression of STAT3 did not change significantly. Also, the protein expressions of SOCS3, IL-6, JAK3, and STAT3, as well as phosphorylated JAK3 and STAT3, were significantly upregulated. We identified 671 lncRNA DEGs, 3281 mRNA DEGs, and 214 miRNA DEGs to be involved in Sporothrix globosa infection. The study findings suggest that the JAK/STAT pathway may be a therapeutic target for sporotrichosis.
... This finding is of crucial importance, especially since JAK inhibitors are already well tolerated and have been approved for treating other inflammatory conditions. Although the use of JAK inhibitors in the treatment of patients with AIP has not yet been subjected to formal clinical studies, preliminary results suggest a notable therapeutic potential (66)(67)(68). Therefore, JAK/STAT inhibition represents a promising therapeutic direction for addressing AIP, and future research in this area is essential to confirm the effectiveness and safety of this therapeutic strategy. This approach opens up prospects for developing more effective and personalized treatment strategies for patients with AIP. ...
Article
Full-text available
Autoimmune pancreatitis (AIP) is a rare disease. There are two distinct types of AIP: AIP type 1 (AIP-1), a pancreatic manifestation of a multi-organ disease linked to immunoglobulin (Ig)G4, and AIP type 2 (AIP-2), a pancreas-specific disease unrelated to IgG4. The usual course of treatment for AIP is oral corticosteroid medication. Rituximab has also been recommended for recurrent AIP-1 in order to initiate remission and provide ongoing treatment. Immunomodulators such as azathioprine are used to keep certain patients in remission. Evaluation also takes into account a number of pharmacological alternatives, including biologic drugs like anti-tumor necrosis factor therapy, a safe and efficient second-line treatment for AIP-2 relapse or steroid dependence. Corticosteroids and immunosuppressants, which are poorly tolerated due to considerable side effects, are being replaced by other biologic drugs, which may offer a beneficial therapeutic alternative.
Article
Full-text available
This study presents a robust and integrated methodology that harnesses a range of computational techniques to facilitate the design and prediction of new inhibitors targeting the JAK3/STAT pathway. This methodology encompasses several strategies, including QSAR analysis, pharmacophore modeling, ADMET prediction, covalent docking, molecular dynamics (MD) simulations, and the calculation of binding free energies (MM/GBSA). An efficacious QSAR model was meticulously crafted through the employment of multiple linear regression (MLR). The initial MLR model underwent further refinement employing an artificial neural network (ANN) methodology aimed at minimizing predictive errors. Notably, both MLR and ANN exhibited commendable performance, showcasing R2 values of 0.89 and 0.95, respectively. The model's precision was assessed via leave-one-out cross-validation (CV) yielding a Q2 value of 0.65, supplemented by rigorous Y-randomization. , The pharmacophore model effectively differentiated between active and inactive drugs, identifying potential JAK3 inhibitors, and demonstrated validity with an ROC value of 0.86. The newly discovered and designed inhibitors exhibited high inhibitory potency, ranging from 6 to 8, as accurately predicted by the QSAR models. Comparative analysis with FDA-approved Tofacitinib revealed that the new compounds exhibited promising ADMET properties and strong covalent docking (CovDock) interactions. The stability of the new discovered and designed inhibitors within the JAK3 binding site was confirmed through 500 ns MD simulations, while MM/GBSA calculations supported their binding affinity. Additionally, a retrosyn-thetic study was conducted to facilitate the synthesis of these potential JAK3/STAT inhibitors. The overall integrated approach demonstrates the feasibility of designing novel JAK3/STAT inhibitors with robust efficacy and excellent ADMET characteristics that surpass Tofacitinib by a significant margin.
Article
Inhibition of Janus kinase 3 (JAK3), a member of the JAK family of tyrosine kinases, remains an essential area of research for developing treatments for autoimmune diseases, particularly cancer and rheumatoid arthritis. The recent discovery of a new JAK3 protein, PDB ID: 4Z16, offers exciting possibilities for developing inhibitors capable of forming a covalent bond with the Cys909 residue, thereby contributing to JAK3 inhibition. A powerful prediction model was constructed and validated using Monte Carlo methods, employing various internal and external techniques. This approach resulted in the prediction of eleven new molecules, which were subsequently filtered to identify six compounds exhibiting potent pIC 50 values. These candidates were then subjected to ADMET analysis, molecular docking (including reversible-reversible docking with tofa-citinib, an FDA-approved drug, and reversible-irreversible docking for the newly designed compounds), molecular dynamics (MD) analysis for 300 ns, and calculation of free binding energy. The results suggested that these compounds hold promise as JAK3 inhibitors. In summary, the new compounds have exhibited favorable outcomes compared to other compounds across various modeling approaches. The collective findings from these investigations provide valuable insights into the potential therapeutic applications of cova-lent JAK3 inhibitors, offering a promising direction for the development of novel treatments for autoimmune disorders.
Article
Full-text available
Nezulcitinib (TD‐0903), a lung‐selective pan–Janus‐associated kinase (JAK) inhibitor designed for inhaled delivery, is under development for treatment of acute lung injury associated with coronavirus disease 2019 (COVID‐19). This 2‐part, double‐blind, randomized, placebo‐controlled, single ascending dose (Part A) and multiple ascending dose (Part B) phase 1 study evaluated the safety, tolerability, and pharmacokinetics (PK) of nezulcitinib in healthy participants. Part A included 3 cohorts randomized 6:2 to receive a single inhaled dose of nezulcitinib (1, 3, or 10 mg) or matching placebo. Part B included 3 cohorts randomized 8:2 to receive inhaled nezulcitinib (1, 3, or 10 mg) or matching placebo for 7 days. The primary outcome was nezulcitinib safety and tolerability assessed from treatment‐emergent adverse events (TEAEs). The secondary outcome was nezulcitinib PK. All participants completed the study. All TEAEs were mild or moderate in severity, and none led to treatment discontinuation. Overall (area under the plasma concentration‐time curve) and peak (maximal plasma concentration) plasma exposures of nezulcitinib were low and increased in a dose‐proportional manner from 1–10 mg in both parts, with no suggestion of clinically meaningful drug accumulation. Maximal plasma exposures were below levels expected to result in systemic target engagement, consistent with a lung‐selective profile. No reductions in natural killer cell counts were observed, consistent with the lack of a systemic pharmacological effect and the observed PK. In summary, single and multiple doses of inhaled nezulcitinib at 1, 3, and 10 mg were well tolerated in healthy participants, with dose‐proportional PK supporting once‐daily administration.
Article
Full-text available
The inhaled lung-selective pan-JAK inhibitor nezulcitinib appears generally well tolerated in hospitalised patients with severe #COVID-19, with trends for improved oxygenation and clinical status, shortened hospitalisation, and fewer deaths versus placebo https://bit.ly/35Xs1Rf
Article
Full-text available
The Janus kinase/signal transducers and activators of transcription (JAK/STAT) are key intracellular mediators in the signal transduction of many cytokines and growth factors. Common γ chain cytokines and interferon-γ that use the JAK/STAT pathway to induce biological responses have been implicated in the pathogenesis of alopecia areata (AA), a T cell-mediated autoimmune disease of the hair follicle. We previously showed that therapeutic targeting of JAK/STAT pathways using the first-generation JAK1/2 inhibitor, ruxolitinib, and the pan-JAK inhibitor, tofacitinib, was highly effective in the treatment of human AA, as well as prevention and reversal of AA in the C3H/HeJ mouse model. To better define the role of individual JAKs in the pathogenesis of AA, in this study, we tested and compared the efficacy of several next-generation JAK-selective inhibitors in the C3H/HeJ mouse model of AA, using both systemic and topical delivery. We found that JAK1-selective inhibitors as well as JAK3-selective inhibitors robustly induced hair regrowth and decreased AA-associated inflammation, whereas several JAK2-selective inhibitors failed to restore hair growth in treated C3H/HeJ mice with AA. Unlike JAK1, which is broadly expressed in many tissues, JAK3 expression is largely restricted to hematopoietic cells. Our study demonstrates inhibiting JAK3 signaling is sufficient to prevent and reverse disease in the preclinical model of AA.
Article
Full-text available
Background Alopecia areata (AA) is an autoimmune form of hair loss with limited treatments. Objective To evaluate the efficacy and safety of the Janus kinase inhibitors ritlecitinib and brepocitinib in patients with AA with ≥50% scalp hair loss. Methods Patients were randomized to once-daily ritlecitinib, brepocitinib, or placebo. The primary efficacy endpoint was 24-week change from baseline in Severity of Alopecia Tool (SALT) score; the key secondary efficacy endpoint was proportion of patients achieving 30% improvement in SALT score (SALT30). Results The ritlecitinib, brepocitinib, and placebo groups included 48, 47, and 47 patients, respectively. At week 24, least-squares mean difference from placebo in SALT score change from baseline was 31.1 (95% CI, 18.8-43.5) for ritlecitinib and 49.2 (95% CI, 36.6-61.7) for brepocitinib (P < .0001 for both comparisons with placebo). SALT30 was achieved by 50% (90% CI 38-62%) of patients receiving ritlecitinib, 64% (90% CI, 51-75%) receiving brepocitinib, and 2% (90% CI, 0-9%) receiving placebo. Two patients experienced a serious adverse event (rhabdomyolysis) in the brepocitinib group only. Limitations Only a single dosage regimen of each study drug was included. Conclusion Treatment with ritlecitinib or brepocitinib for 24 weeks was efficacious and generally well tolerated.
Article
Full-text available
BACKGROUND Severe coronavirus disease 2019 (Covid-19) is associated with dysregulated inflammation. The effects of combination treatment with baricitinib, a Janus kinase inhibitor, plus remdesivir are not known. METHODS We conducted a double-blind, randomized, placebo-controlled trial evaluating baricitinib plus remdesivir in hospitalized adults with Covid-19. All the patients received remdesivir (≤10 days) and either baricitinib (≤14 days) or placebo (control). The primary outcome was the time to recovery. The key secondary outcome was clinical status at day 15. RESULTS A total of 1033 patients underwent randomization (with 515 assigned to combination treatment and 518 to control). Patients receiving baricitinib had a median time to recovery of 7 days (95% confidence interval [CI], 6 to 8), as compared with 8 days (95% CI, 7 to 9) with control (rate ratio for recovery, 1.16; 95% CI, 1.01 to 1.32; P=0.03), and a 30% higher odds of improvement in clinical status at day 15 (odds ratio, 1.3; 95% CI, 1.0 to 1.6). Patients receiving high-flow oxygen or noninvasive ventilation at enrollment had a time to recovery of 10 days with combination treatment and 18 days with control (rate ratio for recovery, 1.51; 95% CI, 1.10 to 2.08). The 28-day mortality was 5.1% in the combination group and 7.8% in the control group (hazard ratio for death, 0.65; 95% CI, 0.39 to 1.09). Serious adverse events were less frequent in the combination group than in the control group (16.0% vs. 21.0%; difference, −5.0 percentage points; 95% CI, −9.8 to −0.3; P=0.03), as were new infections (5.9% vs. 11.2%; difference, −5.3 percentage points; 95% CI, −8.7 to −1.9; P=0.003). CONCLUSIONS Baricitinib plus remdesivir was superior to remdesivir alone in reducing recovery time and accelerating improvement in clinical status among patients with Covid-19, notably among those receiving high-flow oxygen or noninvasive ventilation. The combination was associated with fewer serious adverse events. (Funded by the National Institute of Allergy and Infectious Diseases; ClinicalTrials.gov number, NCT04401579. opens in new tab.)
Article
Full-text available
Filgotinib (Jyseleca®) is an oral, ATP-competitive, reversible JAK1 preferential inhibitor that is being developed by Galapagos NV and Gilead Sciences for the treatment of inflammatory autoimmune diseases, including inflammatory arthritis and inflammatory bowel disease. The JAK-STAT signalling pathway has been implicated in the pathogenesis of inflammatory and autoimmune diseases, and filgotinib modulates this pathway by preventing the phosphorylation and activation of STATs. In September 2020, filgotinib received its first approvals in the EU and Japan. In the EU, filgotinib is indicated for the treatment of moderate to severe active rheumatoid arthritis (RA) in adults who have responded inadequately to, or who are intolerant to, one or more disease-modifying anti-rheumatic drugs (DMARDs). In Japan, filgotinib is indicated for the treatment of RA in patients who had an inadequate response to conventional therapies (including prevention of structural damage to joints). Clinical studies of filgotinib for the treatment of inflammatory autoimmune diseases are ongoing worldwide. This article summarizes the milestones in the development of filgotinib leading to this first approval.
Article
Full-text available
Myeloproliferative neoplasm (MPN)-associated myelofibrosis (MF) is characterized by cytopenias, marrow fibrosis, constitutional symptoms, extramedullary hematopoiesis, splenomegaly, and shortened survival. Constitutive activation of the janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway in MF leads to cell proliferation, inhibition of cell death, and clonal expansion of myeloproliferative malignant cells. Fedratinib is a selective oral JAK2 inhibitor recently approved in the United States for treatment of adult patients with intermediate-2 or high-risk MF. In mouse models of JAK2V617F-driven myeloproliferative disease, fedratinib blocked phosphorylation of STAT5, increased survival, and improved MF-associated disease features, including reduction of white blood cell counts, hematocrit, splenomegaly, and fibrosis. Fedratinib exerts off-target inhibitory activity against bromodomain-containing protein 4 (BRD4); combination JAK/STAT and BRD4 inhibition was shown to synergistically block NF-kB hyperactivation and inflammatory cytokine production, attenuating disease burden and reversing bone marrow fibrosis in animal models of MPNs. In patients, fedratinib is rapidly absorbed and dosed once daily (effective half-life 41 h). Fedratinib showed robust clinical activity in JAK-inhibitor-naïve patients and in patients with MF who were relapsed, refractory, or intolerant to prior ruxolitinib therapy. Fedratinib is effective regardless of JAK2 mutation status. Onset of spleen and symptom responses are typically seen within the first 1–2 months of treatment. The most common adverse events (AEs) with fedratinib are grades 1–2 gastrointestinal events, which are most frequent during early treatment and decrease over time. Treatment discontinuation due to hematologic AEs in clinical trials was uncommon (~3%). Suspected cases of Wernicke’s encephalopathy were reported during fedratinib trials in ~1% of patients; thiamine levels should be monitored before and during fedratinib treatment as medically indicated. Phase III trials are ongoing to assess fedratinib effects on long-term safety, efficacy, and overall survival. The recent approval of fedratinib provides a much-needed addition to the limited therapeutic options available for patients with MF.
Article
Full-text available
Objective To evaluate the efficacy and safety of PF‐06651600 (ritlecitinib), an irreversible inhibitor of JAK3 and the tyrosine kinase expressed in hepatocellular carcinoma (TEC) kinase family, in comparison with placebo in patients with rheumatoid arthritis (RA). Methods An 8‐week, phase II, double‐blind, parallel‐group study was conducted. Seventy patients who were seropositive for anti–citrullinated protein antibodies and/or rheumatoid factor were randomized 3:2 to receive oral PF‐06651600 (200 mg once daily) or placebo for 8 weeks. Eligible patients had an inadequate response to methotrexate, and the study design allowed up to 50% of patients to have previously received 1 tumor necrosis factor inhibitor that was inadequately effective and/or not tolerated. The primary end point was change from baseline in the Simplified Disease Activity Index (SDAI) score at week 8, assessed by Bayesian analysis using an informative prior distribution for placebo response. Results Mean change from baseline in the SDAI score at week 8 was greater in the PF‐06651600 group (−26.1 [95% credible interval −29.7, −22.4]) than in the placebo group (−16.8 [95% credible interval −20.9, −12.7]; P < 0.001). Most adverse events (AEs) were mild in severity, and no treatment‐related serious AEs, severe AEs, or deaths were reported. The most common classes of AE were infections and infestations as well as skin and subcutaneous tissue disorders; there was 1 mild case of herpes simplex in the PF‐06651600 group that was considered to be treatment related, which resolved within 3 days without study treatment discontinuation or antiviral therapy. Conclusion Treatment with the oral JAK3/TEC inhibitor PF‐06651600 (200 mg once daily) was associated with significant improvements in RA disease activity and was generally well‐tolerated in this small 8‐week study.
Article
Background Upadacitinib is an oral selective Janus kinase inhibitor to treat rheumatoid arthritis. The efficacy and safety of upadacitinib as compared with abatacept, a T-cell costimulation modulator, in patients with rheumatoid arthritis refractory to biologic disease-modifying antirheumatic drugs (DMARDs) are unclear. Methods In this 24-week, phase 3, double-blind, controlled trial, we randomly assigned patients in a 1:1 ratio to receive oral upadacitinib (15 mg once daily) or intravenous abatacept, each in combination with stable synthetic DMARDs. The primary end point was the change from baseline in the composite Disease Activity Score for 28 joints based on the C-reactive protein level (DAS28-CRP; range, 0 to 9.4, with higher scores indicating more disease activity) at week 12, assessed for noninferiority. Key secondary end points at week 12 were the superiority of upadacitinib over abatacept in the change from baseline in the DAS28-CRP and the percentage of patients having clinical remission according to a DAS28-CRP of less than 2.6. Results A total of 303 patients received upadacitinib, and 309 patients received abatacept. From baseline DAS28-CRP values of 5.70 in the upadacitinib group and 5.88 in the abatacept group, the mean change at week 12 was −2.52 and −2.00, respectively (difference, −0.52 points; 95% confidence interval [CI], −0.69 to −0.35; P<0.001 for noninferiority; P<0.001 for superiority). The percentage of patients having remission was 30.0% with upadacitinib and 13.3% with abatacept (difference, 16.8 percentage points; 95% CI, 10.4 to 23.2; P<0.001 for superiority). During the treatment period, one death, one nonfatal stroke, and two venous thromboembolic events occurred in the upadacitinib group, and more patients in the upadacitinib group than in the abatacept group had elevated hepatic aminotransferase levels. Conclusions In patients with rheumatoid arthritis refractory to biologic DMARDs, upadacitinib was superior to abatacept in the change from baseline in the DAS28-CRP and the achievement of remission at week 12 but was associated with more serious adverse events. Longer and larger trials are required in order to determine the effect and safety of upadacitinib in patients with rheumatoid arthritis. (Funded by AbbVie; SELECT-CHOICE Clinicaltrials.gov number, NCT03086343.)