You are on page 1of 14

Therapeutics

Current and future status of JAK inhibitors


Donal P McLornan, Janet E Pope, Jason Gotlib, Claire N Harrison

An enhanced understanding of the importance of Janus kinase (JAK) and signal transducer and activator of Lancet 2021; 398: 803–16
transcription (STAT) signalling in multiple disease states has led to an increasing applicability of therapeutic Department of Haematology,
intervention with JAK inhibitors. These agents have revolutionised treatments for a heterogeneous group of disorders, Guy’s and St Thomas’ NHS
Foundation Trust, London, UK
such as myeloproliferative neoplasms, rheumatoid arthritis, inflammatory bowel disease, and multiple immune-driven
(D P McLornan PhD,
dermatological diseases, exemplifying rapid bench-to-bedside translation. In this Therapeutics paper, we summarise Prof C N Harrison DM);
the currently available data concerning the successes and safety of an array of JAK inhibitors and hypothesise on how Department of Rheumatology,
these fields could develop. University of Western Ontario,
London, ON, Canada
(Prof J E Pope MD); Division of
Introduction transduction (figure). Activation and transphosphorylation Hematology, Stanford
Janus kinases (JAKs) are multidomain non-receptor of JAKs induces signal trans­ ducer and activator of University School of Medicine,
tyrosine kinases that have pivotal roles in cellular signal transcription (STAT) recruit­ment, dimerisation, nuclear Stanford Cancer Institute,
Stanford, CA, USA
transduction. The targeting of JAK-associated pathways translocation, and resultant transcriptional responses.2,5 (Prof J Gotlib MD)
through the use of JAK inhibitors has rapidly entered the JAK signalling is tightly regulated at multiple levels, cross
Correspondence to:
clinical arena for a wide array of disease states, including talk with many other intracellular pathways is evident, and Prof Claire Harrison, Department
myeloproliferative neoplasms, rheumatoid arthritis and full elucidation of these complex networks is ongoing. of Haematology, Guy’s and
other immune-mediated arthropathies, numerous inflam­ This evolutionary conserved pathway can be deregulated St Thomas’ NHS Foundation
Trust, London SE1 9RT, UK
matory dermatological disorders, and inflammatory bowel in many disease states, most frequently through acquired
claire.harrison@gstt.nhs.uk
disease, exemplifying rapid bench-to-bedside translation activating mutations or gene amplification.
and modifying many classic treatment algorithms. JAK–STAT signalling has a pivotal role in a pleotropic
Clinicians need to be up to date with this rapidly changing range of systems, including the orchestration and
field, given the unique opportunities afforded by JAK functional capability of immune responses, in particular
inhibitors in the modulation of signalling and pathogenetic T-cell polarisation, control of haematopoiesis and
immune responses in many disease areas. Furthermore,
an understanding is required of the potential safety
concerns and limitations of JAK inhibitors, which had Extracellular
EPO
halted many initially promising clinical trials. In this IL-6
Therapeutics paper, we provide an expert, up-to-date
gp130
Epo receptor IL-6
overview of JAK inhibitors and discuss how this complex receptor
therapeutic field might develop. We highlight the use
of JAK inhibitors in clinical scenarios, including
haematology-oncology, rheumatology, dermatology, and
gastro­enterology, as they are the most established in these
P
scenarios and have revolutionised many traditional PI3K JAK2 JAK2 RAS JAK2
TYK2
P
treatment pathways. Other potential applications in a P P P
P JAK1
disparate group of disorders are being developed and we P RAF P
AKT
await evaluation with interest. P P P
STAT5 dimerisation P STAT3 dimerisation
P P
P MEK P
JAK–STAT signalling in health and disease MTOR
P
P
In humans, the JAK family comprises JAK1, JAK2, JAK3, Nuclear P
ERK
and TYK2.1–4 Irrespective of type, all cytokine receptors are translocation
P
associated with one or more of the JAKs to facilitate signal
P
Nucleus
P Transcriptional
Search strategy and selection criteria activation or inhibition
P
We identified references for this Therapeutics paper by
searching MEDLINE, PubMed, and references from relevant Figure: Simplified overview of JAK–STAT signalling via EPO receptor and IL-6 receptor activation
articles using relevant search terms, such as “Jak inhibitor”, Cytokine receptors do not have intrinsic kinase or phosphatase activity. For ligands such as EPO, the receptor
“myeloproliferative”, and “rheumatoid arthritis”, and subunits are bound as homodimers, whereas, for interleukins and interferons, the receptor subunits are
searching between Jan 1, 1995, and Dec 31, 2020. Only heterodimers. JAK family members are essential to transduce cytokine-mediated signals via the JAK–STAT pathway.
After receptor–ligand binding, STATs dimerise and can translocate to the nucleus to influence transcriptional
articles published in English were included. Abstracts and activation. Prosurvival pathways, such as the PI3K–AKT and RAS–RAF–EFL pathways, can be upregulated.
reports from meetings were included only when we deemed EFL=elongation factor-like GTPase. ERK=extracellular signal-regulated kinase. JAK=Janus kinase. MEK=mitogen-
them of pivotal importance to the readership. activated protein kinase. P=phosphorylation. PI3K=phosphoinositide 3-kinase. RAF=Raf family kinases.
RAS=Ras family GTPases. STAT=signal transducer and activator of transcription.

www.thelancet.com Vol 398 August 28, 2021 803


Therapeutics

In the oncology field, many solid tumours have


Panel 1: Drug selectivity profiles aberrant JAK signalling as part of a prosurvival
Drug (suggested selectivity against Janus kinase [JAK] phenotype and to facilitate tumour migration.15 In depth
family members) review is outside the remit of this article, but clinically
• Ruxolitinib (JAK1 and JAK2 >JAK3 >TYK2) relevant examples of JAK in oncology include JAK2
• Baricitinib (JAK1 and JAK2, and moderate activity against locus amplification in gastric adenocarcinoma and
TYK2) amplification or enhanced nuclear translocation of
• Tofacitinib (JAK3 >JAK2 >JAK1) STAT5 in prostate cancer, offering the prospect of benefit
• Fedratinib (JAK2) of JAK inhibitors.15–18
• Momelotinib (JAK1 and JAK2) With regard to targeting JAK family members other
• Pacritinib (JAK2 >JAK1 and JAK3) than JAK2, TYK2 is an essential regulator of both the
• Fligotinib (JAK1 >JAK2 >JAK3 and TYK2) IL-12 and T-helper (Th)1 and IL-23 and Th17 axes and
• Upadacitinib (JAK1 >JAK2 and JAK3) the type I interferon pathways.15,19 Thus, specifically
• Itacitinib (JAK1) targeting TYK2 could be beneficial across a range of
• Decernotinib (JAK3) immune-mediated and neoplastic disorders but,
• Peficitinib (all JAK proteins) historically, TYK2 inhibitor development has been
• Deucravacitinib (BMS-986165; TYK2) challenging. TYK2 expression and dependency is
• Abrocitinib (JAK1) prevalent in anaplastic large-cell lymphoma, and TYK2
• NDI-031301 (TYK2) inhibition induces apoptosis in anaplastic large-cell
• Ritlecitinib (PF-06651600; JAK3) lymphoma models.20 The most advanced TYK2 inhibitor
in clinical use is the oral agent, deucravacitinib (Bristol
Myers Squibb, New York, NY, USA), discussed later in
inflam­mation, adipogenesis, and growth.6–8 The essential detail.21 Finally, JAK3 activation has additionally been
role of JAKs in cytokine signalling is pivotal to under­ found in a wide array of B-cell lymphoid malig­nancies,
standing how many cytokine-dependent inflammatory, highlighting several areas for potential future therapeutic
auto­immune, and neoplastic disorders associated with exploitation.22
deregulated activity can be manipulated and targeted
through therapeutic JAK inhibition. Overview of JAK inhibitors: current and future
Within haematology, the largest area of clinical expan­ perspectives
sion for JAK inhibitors has been in the myeloproliferative Currently licensed JAK inhibitors include tofacitinib
neoplasm field. The JAK2 Val617Phe mutation, leading (Pfizer, New York, NY, USA), ruxolitinib (Novartis, Basel,
to constitutive activation, is located in the JH2 domain Switzerland), fedratinib (Celgene, Summit, NJ, USA),
and is found in approximately 60% of individuals with upadacitinib (AbbVie, North Chicago, IL, USA), peficitinib
myelofibrosis, 50–60% with essential thrombocythaemia, (Astellas Pharma, Tokyo, Japan), and baricitinib (Eli Lilly,
and 97–98% with polycythaemia vera.9–12 Furthermore, Indianapolis, IN, USA; panel 1). Clinically relevant
irrespective of JAK2 mutational status, many selected agents are highlighted in panel 1 and table and
myeloproliferative neoplasm disorders are characterised are discussed in detail in relevant disease areas.
by upregulated JAK–STAT signalling via mutations in Achievement of JAK isoform specificity is difficult due
additional canonical genes, such as CALR.13 There has to structural homology between family members.
been rapid clinical development of JAK inhibitors and, Additionally, even if specificity is attempted, given the
in 2011, ruxolitinib (a JAK1 and JAK2 inhibitor) was the wide and complex roles of JAKs and the interdepend­
first JAK inhibitor to be approved for use in myelofibrosis ency of many cytokine receptors, resultant effects can be
by the US Food and Drug Administration (FDA) and pleotropic.35 More novel JAK inhibitor compounds are in
European Medicines Agency (EMA). advanced development, such as ritlecitinib (previously
Deregulated JAK–STAT signalling contributes to known as PF-06651600; Pfizer), which acts as a potent
autoimmune and chronic inflammatory phenotypes, inhibitor of JAK3 and tyrosine-protein kinase Tec (TEC)
often in a disease-independent way, and JAK-dependent family kinase members (ie, BTK, BMX, ITK, RLK, and
cytokines (eg, IL-6) are often pivotal pathological drivers, TEC) and can inhibit the functional capacity of both
thus explaining the rationale of JAK inhibitors in these CD8+ T cells and natural killer cells. The dual action of
disorders.14 Both first and later generation JAK in­ ritlecitinib might have an important role in therapeutic
hibitors have been explored across a disparate group of intervention for a plethora of inflammatory diseases.36
disorders, including atopic dermatitis and alopecia There is still much to learn about improving target
areata, immune-mediated arthropathies, inflammatory specificity in JAK–STAT signalling and how this might
bowel disease, and interferonopathies, as we will translate into clinical efficacy and the potential mitigation
discuss in this Therapeutics paper. Despite many of observed side-effects. Long-term safety data of many of
overlapping pathogenetic autoimmune mechanisms, these agents are emerging but still require robust
specific drug therapeutic efficacy can vary markedly. follow-up.

804 www.thelancet.com Vol 398 August 28, 2021


Therapeutics

Phase Agent Study population Main findings Safety and tolerability


Myelofibrosis
COMFORT-123 3 Ruxolitinib vs 309 patients with intermediate-2 59% of patients given ruxolitinib had ≥35% SVR at any Most common grade 3–4 adverse events
placebo or high-risk myelofibrosis point during the trial and impressive symptom responses; were anaemia and thrombocytopenia
evident survival advantage vs placebo
COMFORT-224 3 Ruxolitinib vs 219 patients with intermediate-2 53·4% in the ruxolitinib group had ≥35% SVR at any point; Most common grade 3–4 adverse events
BAT or high-risk myelofibrosis probability of ongoing spleen response at 5 years was 0·48; were anaemia and thrombocytopenia;
intention-to-treat analysis showed ruxolitinib associated increased risk of non-melanoma skin
with improved overall survival cancer
JUMP25 3b (expanded Ruxolitinib 1144 patients with intermediate-2 56·9% of patients had ≥50% reduction in palpable spleen Most common adverse event was drug
access) or high-risk myelofibrosis, as well length at week 24 and 62·3% at week 48; 44% to 46% and related cytopenia; herpes zoster (3·6%) and
as an analysis of 163 patients with 46% to 52% of patients had objective symptom responses influenza (3·0%) were the most common
intermediate-1 myelofibrosis viral infections
ROBUST26 2 (open label) Ruxolitinib 48 patients with intermediate-1, Treatment success was ≥50% reduction in spleen length or Most common adverse event was drug
intermediate-2, and high-risk ≥50% decrease in myelofibrosis symptoms at 48 weeks, related cytopenia, similar to other studies;
myelofibrosis and 50% of all patients and 57% of patients with one case of progressive multifocal
intermediate-1 risk had treatment success leukoencephalopathy was reported
SIMPLIFY-127 3 Momelotinib 432 patients with intermediate-2 First-line momelotinib was statistically non-inferior to Common side effects included cytopenia
vs ruxolitinib in or high-risk disease or symptomatic ruxolitinib on spleen response rates (p=0·011) but was and treatment emergent peripheral
patients with intermediate-1 myelofibrosis inferior with regard to symptom response; transfusion rates neuropathy
JAK inhibitor- and independence were better in the momelotinib group
naive disease
SIMPLIFY-228 3 Momelotinib 104 patients in the momelotinib In patients who had been given ruxolitinib, momelotinib Common side effects included cytopenia
vs BAT group and 52 in the BAT group was not superior to BAT for SVR and treatment emergent peripheral
(46 given ruxolitinib) neuropathy
PERSIST-129 3 Pacritinib vs 220 patients with intermediate-1 Primary endpoint of SVR of ≥35% was found in 42 (19%) of Common grade 3–4 adverse events were
BAT risk or above in the pacritinib group 220 patients in the pacritinib group vs five (5%) of anaemia (17%), thrombocytopenia (12%),
and 107 in the BAT group 107 patients in the BAT group (p=0·0003) and diarrhoea (5%); serious adverse events
were cardiac failure (2%); 12% of patients
died due to adverse events in the pacritinib
group and 13% of patients in the BAT
group
PERSIST-230 3 Pacritinib vs 311 patients with intermediate-1 Pacritinib (groups combined) was significantly more effective Most common grade 3–4 adverse events
BAT risk or above with a platelet count than BAT for ≥35% SVR rates and had ≥50% reduction were anaemia, thrombocytopenia, and
(two-dose of 100 × 10⁹ cells per L or less (not significant) in total symptom score; pacritinib twice daily gastrointestinal adverse events (diarrhoea
strata vs BAT) led to significant improvements in both endpoints vs BAT; and vomiting)
more patients given pacritinib had reduced red blood cell
transfusion dependence at week 24
JAKARTA31 3 Fedratinib 289 patients with intermediate-2 SVR (≥35%) was reached by 36% of patients given Gastrointestinal symptoms, anaemia,
(two-dose or high-risk disease fedratinib 400 mg and 40% of patients given fedratinib and increased concentrations of liver
strata vs 500 mg vs 1% in the placebo group (p<0·001); there was a aminotransferase; encephalopathy was
placebo) 50% reduction in total symptom score at week 24, 36% in reported in four women receiving
the fedratinib 400 mg group, 34% for the fedratinib fedratinib 500 mg; Wernicke’s
500 mg group, and 7% in the placebo group (p<0·001) encephalopathy was diagnosed in
three patients and suspected in one patient
in the treatment cohort
JAKARTA232 2 Fedratinib 97 patients with intermediate-1 31% of patients had at least ≥35% SVR by the end of the Grade 3–4 adverse events were anaemia
risk or above who were previously sixth cycle; 27% had ≥50% symptom response rate and thrombocytopenia; the trial was
treated with ruxolitinib temporarily halted due to perceived risks of
Wernicke’s encephalopathy
Polycythaemia vera
RESPONSE33 3 Ruxolitinib vs 212 patients requiring phlebotomy Primary endpoint of patients who reached both Grade 1–2 adverse events were fatigue,
BAT or who were resistant or intolerant haematocrit control (<45% without phlebotomy) and headache, and diarrhoea; herpes zoster
to hydroxycarbamide with SVR >35% at week 32 was reached by 21% of the ruxolitinib infection was more common in the
splenomegaly group vs 0·9% in the BAT group; there were fewer ruxolitinib group than in the placebo
thrombotic events for patients given ruxolitinib group; increased risk of non-melanoma
skin cancer
RESPONSE-234 3b Ruxolitinib vs 149 patients requiring phlebotomy 62% of patients in the ruxolitinib group vs 19% in the BAT Grade 3–4 adverse events were
BAT or were resistant or intolerant to group had the primary endpoint of haematocrit control hypertension and angina; two patients died
hydroxycarbamide (<45% without phlebotomy) at week 32 in the BAT group
BAT=best alternative therapy. JAK=Janus kinase. SVR=splenic volume reduction.

Table: Overview of key clinical trials of JAK inhibitors for myeloproliferative neoplasms

www.thelancet.com Vol 398 August 28, 2021 805


Therapeutics

Clinical relevance had been given ruxolitinib, showed that 30 (31%) of


Haematological disorders 97 patients had a 35% or better splenic volume reduction
There have been many trials, both completed and ongoing by the end of the sixth cycle and 26 (27%) of 97 had a
or planned, exploring the use of JAK inhibitors in 50% or better symptom response rate.32 However, in 2013,
myeloproliferative neoplasms (table). Availability of these fedratinib was temporarily placed on hold due to concerns
new oral JAK inhibitors revolutionised the field and of increased susceptibility to Wernicke’s encephalopathy,
we highlight four JAK inhibitors that have made which were subsequently revised. Fedratinib re-entered
the most impact (ruxolitinib, fedratinib, momelotinib, and the clinical arena after development by Celgene in
pacritinib). Historically, sparse therapeutic options were two large phase 3 studies (FREEDOM-1 and FREEDOM-2)
available for myelofibrosis, a disorder characterised by an for patients with myelofibrosis who have previously been
average life expectancy of 5 years, constitutional symp­ given ruxolitinib. On Aug 16, 2019, the FDA approved
toms, progressive splenomegaly, and variable cytopenias. fedratinib for first-line and second-line therapy for
Two large phase 3 trials (COMFORT-I and COMFORT-II) myelofibrosis.38 However, a black box warning remains for
supported the efficacy of ruxolitinib, leading to rapid serious and possibly fatal encephalopathy. Unaddressed
drug approval.23,24 In COMFORT-1, 309 patients with issues for use in patients with myelofibrosis are whether
intermediate-2 or high-risk myelofibrosis (according to the active thiamine monitoring is required for patients
International Prognostic Scoring System criteria) were receiving fedratinib and also how to safely transition from
randomly assigned to a placebo group or ruxolitinib group. ruxolitinib to fedratinib (or vice versa). Consideration
For the ruxolitinib group, 65 (42%) of 155 had a 35% or needs to be given to tapering dose, monitoring for with­
greater splenic volume reduction at 24 weeks after initial drawal, and use of steroids or short-term reintroduction
dose, and myelofibrosis symptom responses were of ruxolitinib or fedratinib.
impressive alongside improve­ments in quality of life. The Anaemia is common in myelofibrosis and is an unmet
median duration of spleen response was 168 weeks on clinical need.39 Momelotinib, a potent JAK1 and JAK2
ruxolitinib and there was a significant overall survival inhibitor (Sierra Oncology, Vancouver, BC, Canada,
advantage versus placebo (hazard ratio 0·69; p=0·025). previously Gilead Sciences, Foster City, CA, USA) was
Similar results were seen in COMFORT-II, comparing evaluated in two phase 3 trials for myelofibrosis
ruxolitinib with the best available therapy. Probability of (SIMPLIFY-1 and SIMPLIFY-2) with heterogeneous
ongoing spleen response 5 years after first dose was 48%, results with regard to splenic volume reduction and
highlighting long-term efficacy, and there was a survival symptom responses.27,28 These studies had several key
advantage favouring ruxolitinib.37 Rapid introduction into anaemia endpoints, including transfusion rates, which
clinical use has seen many thousands of patients with mostly favoured momelotinib in both the first-line and
myelofibrosis globally benefiting from ruxolitinib. Other second-line setting. The particular benefit of momelotinib
key trials of ruxolitinib in myelofibrosis and their main is that it can improve anaemia alongside reductions
findings, including side-effects and adverse events, are in symptoms and splenomegaly. Mechanistically,
highlighted in the table. momelotinib inhibits both JAK–STAT and the iron
With regard to polycythaemia vera, the phase 3 sensing bone morphogenic protein–activin receptor type-1
RESPONSE trial compared ruxolitinib with the best (BMP–ACTR1) pathways, reducing hepcidin expression
available therapy in patients with polycythaemia vera who and improving functional iron availability, which
required phlebotomy or were intolerant or resistant to the could induce erythroid responses. The development focus
cytoreductive agent, hydroxycarbamide.33 The primary is now on patients with second-line anaemia and
endpoint of the proportion of patients with haematocrit transfusion-dependent myelofibrosis in the international
control (<45% without phlebotomy) and splenic volume phase 3 MOMENTUM study (NCT04173494).
reduction of 35% or more at week 32 favoured Pacritinib is an oral multi-kinase inhibitor with spe­
ruxolitinib (21%) versus best available therapy (0·9%). cificity for JAK2, FLT3, IRAK1, and CSF1R. The focus of
The RESPONSE-2 study supported the superiority of pacritinib development has been for patients with
ruxolitinib to the best available therapy in patients with myelofibrosis and thrombocytopenia. Two large phase 3
inadequately controlled polycythaemia vera without trials investigated effi­cacy in myelofibrosis (PERSIST-1
splenomegaly.34 Ruxolitinib gained approval as a second- and PERSIST-2).29,30 However, both phase 3 trials were
line therapy in polycythaemia vera, a potentially important placed on full clinical hold by the FDA in 2016 due to
step in reducing long-term thrombotic complications in concerns about deaths in the group given pacritinib and
patients. initial concerns about excess cardiac or haemorrhagic
Fedratinib is a potent JAK2 inhibitor (with anti-FLT3 events in PERSIST-1. This hold was subsequently removed
properties) investigated in myelofibrosis that has shown in 2017; the dose finding PAC203 phase 2 study (with
superiority to placebo at 400 mg and 500 mg doses in patients who were previously given ruxolitinib) showed
the phase 3 trial, JAKARTA.31 Updated results from greatest efficacy at 200 mg twice daily,40 and the phase 3
JAKARTA2, a phase 2 study investigating efficacy of trial, PACIFICA, will compare pacritinib with the best
400 mg of fedratinib in 97 patients with myelofibrosis who available therapy in patients with myelofibrosis with

806 www.thelancet.com Vol 398 August 28, 2021


Therapeutics

platelet counts less than 50 × 10⁹ cells per L (NCT03165734). inhibitors for rheumatoid arthritis that is refractory to
Practically, if approved, pacritinib is a very attractive agent conventional synthetic disease modifying anti-rheumatic
for patients with thrombocytopenic myelofibrosis, in drugs (DMARDs) or biologics. As clinical knowledge
which many other agents might be avoided altogether, or accumulates, with the focus on early therapeutic inter­
if a reduced dose is needed therefore limiting efficacy. vention and treat-to-target strategies, it is unknown
It must be stressed that JAK inhibitors can have whether JAK inhibitors will become front-line treatments,
limitations within the myeloproliferative neoplasm although side-effects and the paucity of long-term data
therapeutic arena, including inadequate dose density, should be considered.
absence of true disease modification, and, ultimately, loss Many JAK inhibitors have been used in randomised
of response. Many agents are being investigated along­ trials for rheumatoid arthritis (eg, tofacitinib, baricitinib,
side ruxolitinib in myelofibrosis, including azacytidine upadacitinib, filgotinib, peficitinib, and decernotinib).47,48
in advanced phase disease, recombinant interferons, Tofacitinib and baricitinib are approved in multiple
erythropoiesis maturation agents (eg, luspatercept), countries, and in 2019, upadacitinib, a JAK1 inhibitor,
bromodomain inhibitors, and histone deacetylase in­ was approved for rheu­matoid arthritis in the USA and
hibitors, showing the potential of dual therapy, incor­ peficitinib was approved in Japan.49 Overall, there are
porating JAK inhibitors, to maximise responses.39 benefits of JAK inhibitors that might be considered a
class effect (eg, rapid onset of action in rheumatoid
Graft-versus-host disease arthritis with clinical benefit) and there are also important
Graft-versus-host disease (GVHD) represents one of side-effects within the class (eg, cytopenias, particularly
the most substantial challenges after allogeneic lymphopenia; increased risk of infection, including
haematopoietic stem-cell transplantation. Pathogenetic reactivation of herpes zoster; hypercholesterolaemia;
mechanisms are multifactorial but reliant, at least partly, and, for some JAK inhibitors, increased thromboembolic
on proinflammatory JAK–STAT signalling, not only in events, which will be discussed later; panel 2).47,48 For
alloreactive effector T cells but also in dendritic cells and rheumatoid arthritis, an important class effect appears to
B cells.41 First-line therapy, often systemic steroids, is be clinical efficacy as monotherapy alongside efficacy
effective only in inducing complete responses in a in combination with conventional synthetic DMARD
small proportion of cases and subse­quent strategies are (reviewed by Westhovens).50 These agents are contra­
heterogeneous. Historically, steroid-refractory acute indicated in pregnancy and breastfeeding and no
GVHD carries high mortality rates and is still an unmet evidence exists for safe use in children.
clinical need. Regarding JAK inhibitors, Spoerl and Clinical trial programmes of rheumatoid arthritis for
colleagues42 showed in preclinical models that ruxolitinib JAK inhibitors have followed a similar sequenced trial
reduced effector T-cell pro­ liferation, suppressed pro­ design. JAK inhibitors show efficacy in patients with
inflammatory cytokines and CD4+ cell differentiation active rheumatoid arthritis that does not inadequately
into IFN-γ and IL17A-producing cells, and increased the respond to methotrexate compared with placebo or
number of FoxP3-positive regulatory T cells. After initial a standard-of-care drug (eg, the biologic DMARD,
clinical proof of concept, a large retrospective multi­ adalimumab [a TNF inhibitor]). Often, JAK inhibitors
national study evaluated the efficacy of ruxolitinib in are combined with background methotrexate or, less
95 patients with steroid-refractory GVHD who had been frequently, with other DMARDs. Some trials evaluated
frequently heavily pretreated.43 Excellent overall response monotherapy (ie, JAK inhibitors with no background
rates, not pre­viously observed, were shown in 44 (82%) DMARDs), as discussed previously, and additional
of 54 patients with steroid-refractory acute GVHD studies have been done in patients previously untreated
(including 25 [46%] of 54 complete responses) and with methotrexate.47,48 Outcome measures vary but often
35 (85%) of 41 patients with chronic GVHD. The consist of standardised responses—eg, the American
6-month overall survival rates were 79% for patients with College of Rheumatology (ACR) criteria measures
acute GVHD and 97·4% for patients with chronic improvement by 20%, 50%, and 70% in cumulative
GVHD. Cytopenias and cytomegalovirus reactivations response scores.51 Frequently, trials report on improve­
were noted in some patients. The FDA granted approval ment in the disease activity score (ie, DAS28).52 Both
for the use of ruxolitinib in steroid-refractory acute the ACR criteria and DAS28 are composite scores
GVHD in patients who are 12 years and older, and, more studying changes in tender and swollen joint counts,
recently, both the REACH-2 and REACH-3 phase 3 trials patient global assessment, and inflammatory markers.
have shown efficacy (evaluating ruxolitinib in steroid- Frequently, the proportion of patients with low disease
refractory acute GVHD and chronic GVHD).44–46 activity or who have had remission is also reported. Low
disease activity or remission outcomes are difficult to
Rheumatological disorders reach. Results with JAK inhibitors are similar to other
Rheumatoid arthritis advanced therapies and, when compared head to
Contemporary treatment of rheumatoid arthritis has head, they are sometimes numerically and statistically
evolved rapidly, with many clinicians considering JAK better than the biologic DMARD, adalimu­mab.47,48 In

www.thelancet.com Vol 398 August 28, 2021 807


Therapeutics

Panel 2: Potential side-effects and general monitoring for Janus kinase inhibitors in rheumatological diseases*
Cytopenias Renal impairment and serious liver disease
• Monitor white blood cell count with differential, • Read product monograph for possible dose adjustments or
haemoglobin, and platelet count; for tofacitinib, baseline relative contraindications
and periodic measures every 3 months • Some JAK inhibitors need dose reduction in patients with
• Repeat measurements if absolute lymphocyte count is less moderate renal impairment (eg, baricitinib)
than 500 cells per mm³, and, if this count is persistent and • Safety in patients with severe renal impairment or
from treatment, look for the cause and consider stopping, end-stage renal disease is unknown and JAK inhibitor use is
holding, or decreasing Janus kinase (JAK) inhibitor use not recommended in patients with rheumatic disease in
• Interrupt treatment if absolute neutrophil count is these situations
500–1000 cells per mm³, or repeat or reduce dose, and if
Thromboembolism
persistently less than 500 cells per mm³ consider stopping
Venous thromboembolic disease
medication (if because of JAK inhibitor treatment)
• JAK inhibitors that are not used for haematology-oncology
• If haemoglobin concentration is less than 8 g/L, or if there
have a warning for venous thromboembolic disease risk;
is a decrease of 2 g/L or more, interrupt treatment and
a higher rate of venous thromboembolism was found in a
look for the cause
long-term study of tofacitinib in patients with rheumatoid
• Thrombocytosis can occur rarely, should be monitored
arthritis who were older than 50 years, with at least
and, in general, it is not thought to be associated with
one cardiovascular disease risk factor and taking tofacitinib
thromboembolic risk in patients taking baricitinib;
10 mg twice daily
consideration of haematology review and alternative
• For tofacitinib, the recommended dose in most countries is
causes might be required if thrombocytosis substantial
5 mg twice a day or 11 mg (for prolonged release) once a day
and persistent
• A greater frequency of venous thromboembolism was
Transaminitis shown with baricitinib 4 mg daily than with placebo or 2 mg
• Baseline and periodic liver enzyme monitoring, such as for daily, and only in the randomised controlled trials and not in
alanine aminotransferase concentration the long-term extension studies
• Investigate or repeat monitoring if a patient’s liver • Other JAK inhibitors have not shown an increased risk of
enzyme concentrations are three to five times the upper venous thromboembolic disease, but this risk might be a
limit of healthy range class effect
• For patients given tofacitinib, monitoring every 3 months is • Increased cardiovascular events do not yet appear to be a
required risk of JAK inhibitors
Hyperlipidaemia Malignancy
• Assess cholesterol profile after drug continuation (eg, 2 or • In general, we do not recommend use of JAK inhibitors for
3 months after first dose) and, depending on the results, rheumatic diseases during chemotherapy or active non-skin
treat or follow up according to the relevant lipid guidelines malignancy
• Surveillance is required for patients with non-melanoma
Increased risk of infection
skin cancers
Prevention
• For other diseases refer to product monograph
• If appropriate, vaccinate for herpes zoster virus (eg, if the
patient is older than 50 years) to prevent reactivation; Drug interactions
all vaccinations should be updated (preferably before • Some JAK inhibitors used in rheumatology have drug
starting treatment, but, in our opinion, JAK inhibitor use interactions; usually, with drugs that are not commonly used
could be paused after a risk assessment for a short period of • Ketoconazole increases tofacitinib maximal plasma
time, such as 2 weeks, if giving a live vaccine) concentrations through inhibition of cytochrome P450 3A4
• Administer influenza vaccine and pneumococcal vaccine and cytochrome P450 2C19
according to country guidelines • Other interactions with tofacitinib include fluconazole
• Test for tuberculosis with country standard; if latent, treat • Rifampin might increase tofacitinib metabolism
Mycobacterium tuberculosis infection; and, if positive, consider • Baricitinib does not appear to have clinically relevant drug
tuberculosis treatment concomitantly with JAK inhibitors as interactions
per country guidelines for tuberculosis treatment • Use of JAK inhibitors with other potent immunosuppressives
Active viraemia is not recommended due to immunosuppression
• Do not administer JAK inhibitors in patients with active (immunosuppressants such as tacrolimus and ciclosporin,
hepatitis B or HIV infection and biologics used in the rheumatology field)
• If patient has active hepatitis C viral replication refer them *For each JAK inhibitor used, we suggest clinician review of monitoring recommendations.
for curative therapy

808 www.thelancet.com Vol 398 August 28, 2021


Therapeutics

rheumatoid arthritis treatment, additional evaluation is and alongside methotrexate, and despite early signs of
needed to address topics such as: the optimal sequencing efficacy, there were negative safety signals (eg, infections)
of advanced therapies after non-responsiveness to and increased aminotransferase con­centrations in some
methotrexate; whether there is a benefit in switching to patients, and development was discontinued.61,62
alternative JAK inhibitors after a patient has not
responded or is intolerant; and how to prevent Spondyloarthropathies
attenuated efficacy of JAK inhibitors. Regarding efficacy Regarding spondyloarthritis (considering both psoriatic
and adverse events, dose response rates of each JAK arthritis and ankylosing spondylitis), JAK inhibitors
inhibitor are being clarified and long-term data are might address multiple disease manifestations.
being accumulated. For example, the DARWIN 1 (using Tofacitinib has received approval for 5 mg orally twice a
filgotinib in combination with methotrexate) and day for the treatment of psoriatic arthritis in many
DARWIN 2 (using filgotinib monotherapy) trials countries (reviewed in Virtanen and colleagues47). Data
showed that patients who were given filgotinib had have shown the positive benefits of TNF inhibitor
dose-dependent responses accompanied by dose- exposure with respect to improvement in composite
dependent leukopenia and thrombocytopenia and early scores (ie, ACR criteria responses) and skin improve­
increases in high-density and low-density lipoproteins ment. Efficacy was shown in patients who were previously
that then stabilised.53,54 DARWIN 1 and DARWIN 2 also given conventional synthetic DMARD treatment (in
reported increases in haemoglobin concentrations in whom the majority were given methotrexate as back­
patients given filgotinib. For upadacitinib, results of ground treatment). Results showed an improvement in
two trials (BALANCE-1 and BALANCE-2) showed dose- composite scores, compared with patients given the
dependent clinical responses and higher rates of adverse placebo, and were numerically very similar (ie, not non-
events at higher doses.55,56 inferior) to patients given adalimumab (the usual
The advantages of JAK inhibitors in rheumatoid standard of care).47 With respect to joint involvement,
arthritis include oral delivery (other options are there was no obvious advantage in exceeding the 5 mg
subcutaneous or intravenous), an equal or superior orally twice a day dose. The EQUATOR trial investigated
benefit compared with TNF inhibitors, and the the efficacy of filgotinib, a more JAK1 specific inhibitor
possibility of better retention than TNF inhibitors, than other JAK inhibitors, versus placebo in moderate-to-
which are subject to drug induced antibodies that severe psoriatic arthritis after intolerance or inadequate
attenuate benefit. Better durability with JAK inhibitors response to at least one conventional synthetic DMARD,
in rheumatoid arthritis in patients who inade­quately with clinically relevant responses at 16 weeks after first
respond to biologic DMARDs when switching therapies dose favouring filgotinib (80% vs 33%).63 Upadacitinib is
is of key clinical relevance; TNF inhibitors showed being investigated for psoriatic arthritis in the phase 3
shortest treatment duration and JAK inhibitors, both as SELECT-PsA 1 and SELECT-PsA 2 trials.64,65 For
monotherapy or combination, had the longest treatment SELECT-PsA 1, 1702 patients with psoriatic arthritis who
persistence.57 For example, results from the ORAL did not respond, or were intolerant, to at least one
Sequel long-term extension study, which evaluated open non-biologic DMARD were randomly assigned to 15 mg
label tofacitinib 5 mg and 10 mg twice daily (for up to or 30 mg upadacitinib daily, placebo, or adalimumab (the
9·5 years) in patients with rheumatoid arthritis who had active comparator). ACR criteria improvement by 20%
been enrolled in phase 1, phase 2, or phase 3 index (ACR20) response rates at 12 weeks were 79% for
studies, showed a consistent safety profile and sustained updacitinib 30 mg, 71% for updacitinib 15 mg, 65% for
efficacy.58 Cumulatively, 52% of patients discontinued adalimumab, and 36% for placebo.64 Moreover, a
tofacitinib, of whom, 24% discontinued due to adverse substantial proportion of patients given upadacitinib
events and 4% due to tofacitinib having little of efficacy. had improvements in enthesitis and dactylitis. The
Another area of interest is withdrawing methotrexate SELECT-PsA 2 trial also showed that updacitinib had
for patients with rheumatoid arthritis who are inad­ better efficacy than the placebo.65 Regarding ankylosing
equate responders. Two randomised controlled trials spondylitis, the TORTUGA trial showed efficacy of
have been done with tofacitinib; switching to tofactinib filgotinib versus placebo (with an early efficacy and
from methotrexate is in fact not as efficacious as actually predictable safety and tolerability), and the SELECT-AXIS 1
adding tofacitinib to methotrexate.59,60 The other strategy trial supported efficacy of upadacitinib versus placebo in
of adding tofacitinib to methotrexate and only randomly patients with biologic-naive ankylosing spondylitis.66,67
assigning patients with a very low disease state to stop or Determination of long-term efficacy and safety will be
continue methotrexate showed that the two strategies important.
were equal.59,60 Additionally, it is important to note that
not all JAK inhibitors for rheumatoid arthritis continue Systemic lupus erythematosus
development after late phase trials—eg, decernotinib The heterogeneous pathogenesis and clinical phenotype
(Vertex Pharmaceuticals, Boston, MA, USA), a selective of systemic lupus erythematosus is often therapeutically
JAK3 inhibitor, was investigated both as monotherapy challenging, necessitating novel approaches. Tofacitinib

www.thelancet.com Vol 398 August 28, 2021 809


Therapeutics

has been investigated in a phase 1b trial for mild-to- adalimumab and with the benefit of oral administration.
moderate lupus based on STAT4 risk alleles, in which the Moreover, therapy did not appear to induce substantial
presence of rs7574865A→T in individuals with lupus is haematological or liver enzyme abnormalities or raised
associated with enhanced T-cell responses to IL-12 and lipid concentrations. The most common adverse events
IFN-γ. These enhanced T-cell responses make JAK were nasopharyngitis, gastro­ intestinal upset, and
inhibitors an attractive therapeutic option.68 In a double headaches. The phase 2 efficacy of deucravacitinib led
blind, phase 2 study, baricitinib 2 mg or 4 mg was to subsequent evaluation in two large phase 3 trials,
evaluated against placebo in a 1:1:1 design of 314 adult POETYK-PSO-1 and POETYK-PSO-2 (NCT03611751), in
patients with systemic lupus erythematosus and active which deucravacitinib was compared with placebo and
skin or joint disease.69 The primary endpoint was assessed apremilast (a selective PDE4 inhibitor). For POETYK-
by improvements in the Systemic Lupus Erythematosus PSO-1, results have shown that both primary endpoints
Disease Activity Index-2000. A once daily baricitinib 4 mg were met versus placebo by week 16 (Psoriasis Activity
dose improved symp­toms and signs of systemic lupus Severity Index 75% and a static Physician’s Global
erythematosus in patients with poorly controlled disease Assessment score of clear or almost clear), in addition
(74 [67%] of 104 responded at 24 weeks vs 53% in the to the secondary endpoint of superiority to apremilast.21,73
placebo group for skin or arthritis), whereas there was no Safety data were similar to previous trials. Full results
statistically significant improve­ment in the 2 mg dose. from POETYK PSO-2 are expected in late 2021.
Serious adverse events were seen in 9·6% of patients in In alopecia areata, in which hair follicle destruction
the baricitinib 4 mg group versus 4·8% in the placebo is driven by CD8+ and NKG2+ T cells, multiple JAK
group. As raised by Yuan and colleagues70 in response to inhibitors have shown promising efficacy.74,75 Tofacitinib
the study, the 4 mg dosing strategy of baricitinib was not 5 mg twice daily for 3 months showed efficacy in a single
approved by the FDA for rheumatoid arthritis due to group trial in patients with severe disease, but responses
safety concerns of potential infectious and thrombotic did not have durability after discontinuation.76 An open
complications, hence additional follow-up and more label study of oral ruxolitinib, 20 mg twice daily for
knowledge are required for this dosing regimen. 3–6 months, showed that nine of 12 patients with alopecia
Two dosing strata are being explored in the phase 3 trial, areata had robust responses, with average hair growth of
BRAVE II, with an ongoing long-term follow-up study 90% by end of treatment.77 BRAVE-AA1 (NCT03570749)
from the earlier trials (SLE-BRAVE-X; NCT03843125). was an adaptive phase 2/3 trial investigating baricitinib
1 mg, 2 mg, or 4 mg daily in patients with severe or very
Dermatological disorders severe alopecia areata. Breakthrough therapy status was
A large volume of evidence highlights the efficacy of granted for baricitinib in patients with alopecia areata in
JAK inhibitors in multiple skin disorders. Early focus March, 2020.78 Reports from the phase 2 component of
was predominantly addressing psoriasis management, BRAVE-AA1 showed efficacy for both the 2 mg and 4 mg
but focus now extends to many other disease states, doses compared with placebo, leading to clinically
including alopecia areata, vitiligo, and atopic dermatitis, significant improvements and no new safety concerns or
and includes systemic and topical use. serious adverse events.78 The phase 3 BRAVE-AA1 and
Regarding moderate-to-severe psoriasis, Papp and BRAVE-AA2 (NCT03899259) will additionally evaluate
colleagues71 initially evaluated the role of tofacitinib in the efficacy and safety of the 2 mg and 4 mg doses.
two large phase 3 trials (OPT Pivotal 1 [n=901] and OPT Ritlecitinib (Pfizer) also had breakthrough therapy
Pivotal 2 [n=960]) showing efficacy of tofacitinib (5 mg or designation; data from the phase 2 trial in alopecia areata
10 mg twice daily) over placebo. In the OPT Pivotal 2 showed that patients given ritlecitinib significantly
phase 3 trial, the 10 mg twice daily tofacitinib regimen improved compared with those taking placebo as early as
was found to be non-inferior to etanercept with similar 6 weeks into treatment and met its primary endpoint in
serious adverse event rates during 12 weeks of treatment, improving hair growth by week 24, as did brepocitinib.79
and no new safety signals emerged.72 Despite these Additionally, CTP-543 (a JAK1 and JAK2 inhibitor,
findings, the FDA declined approval for use of tofacitinib Concert Pharmaceuticals, Lexington, MA, USA) is being
in psoriasis in 2015 on the basis of the extent of the explored in a phase 3 alopecia areata trial—THRIVE-AA1
safety data and clinical findings, and hence additional (NCT04518995). Top-line data for THRIVE-AA1 is
development for this indication was halted. expected to be reported in 2022.
Deucravacitinib (Bristol Myers Squibb) was evaluated A major focus has been exploring applicability of
in an initial phase 2 trial in patients with moderate-to- JAK inhibitors for atopic dermatitis. Abrocitinib
severe plaque psoriasis.21 Doses of 3 mg daily and higher (PF-04965842; Pfizer), which gained FDA breakthrough
resulted in greater clearing of psoriasis than did placebo therapy desig­ nation for moderate-to-severe atopic
during 12 weeks, with a favourable risk–benefit ratio. dermatitis in 2018, has shown positive results compared
For example, week 12 Psoriasis Activity Severity Index with placebo.80 In the JADE mono-1 trial,80 387 patients
scores between 67% and 75% were observed with doses were randomly assigned to abrocitinib (200 mg or 100 mg
greater than 6 mg daily, similar to those seen with daily) or placebo for 12 weeks. Coprimary endpoints

810 www.thelancet.com Vol 398 August 28, 2021


Therapeutics

assessed the proportion of patients who had an that investigated tofacitinib as induction therapy
Investigator Global Assessment response score of 0 (OCTAVE Induction-1 and OCTAVE Induction-2) in
(clear) or 1 (almost clear) with a two-grade or better patients who had non-successful responses to
improvement from baseline, and proportion achieving conventional or anti-TNF agents and one trial as
75% or better improvement in Eczema Area and Severity maintenance (Octave Sustain).91 Remission was defined
Index score (EASI-75). 62 (40%) of 156 patients in the by the Mayo score 2 or less and physician global
abrocitinib 100 mg dosing strata and 96 (63%) of 153 in assessment. For both induction studies, tofacitinib had
the 200 mg dosing strata had an EASI-75 response significantly better 8-week remission rates (18·5% in the
compared with nine (12%) of 76 in the placebo group tofacitinib group vs 8·2% in the placebo group in
(p<0·0001). Abrocitinib was well tolerated with no OCTAVE Induction-1, and 16·6% in the tofacitinib
treatment related deaths. In the JADE mono-2 trial, both group vs 3·6% in the placebo group in OCTAVE
dosing strata of abrocitinib showed efficacy in patients Induction-2) and remission rates were improved in both
with atopic dermatitis and, in general, both doses were tofacitinib dosing groups of OCTAVE Sustain, as were
well tolerated.81 The balance of efficacy or safety against glucocorticoid-free 52-week remission rates. Responses
the current standard of care (dupilumab) requires to tofacitinib often happened quickly and, when com­
additional evaluation considering the slight drop in pared with placebo, there were increases in LDL and
platelet count seen in some patients and consideration HDL concentration, which plateaued after 4 weeks, and
given to the risk of infection. Both baricitinib and increased infectious complications, including herpes
upadacitinib have been investigated for use in atopic zoster reactivation. Toxicity with tofacitinib in inflam­
dermatitis, with the EMA granting an extension of matory bowel disease appears to be dose dependent.
indication for baracitinib for moderate-to-severe atopic Tofacitinib was associated with more cases, albeit a small
dermatitis in September, 2020.82,83 Topical ruxolitinib has number, of non-melanoma skin cancers than the
also showed efficacy.84 placebo. An open extension study, OCTAVE Open
Given the dependency of vitiligo progression on IFN-γ (NCT01470612), will provide important long-term efficacy
signalling, JAK inhibitors have also been investigated in and safety data. Tofacitinib appears to be a highly active
this setting. Results from a multicentre, double blind, agent in ulcerative colitis and is currently being
phase 2 study comparing topical ruxolitinib with a control, positioned in the treatment pathway after FDA approval.
showed good tolerability of ruxolitinib topical cream, Guidelines from the American College of Gastro­
which was associated with substantial repigmentation of enterology placed tofacitinib either as first-line therapy
vitiligo lesions, suggesting another area in which these for moderate-to-severely active ulcerative colitis or as
agents could be used.85 second-line therapy after TNF inhibitors.92 Outside of
clinical trials, Weisshoff and colleagues93 reported real-
Inflammatory bowel disease world data of tofacitinib in 58 patients with moderate-to-
Disease-associated proinflammatory cytokines signal, severe inflammatory bowel disease and resistance to
either directly or indirectly, through the JAK–STAT TNF inhibitors; they described meaningful clinical
pathway hence providing rationale for exploring JAK responses in 40 (69%) of 58 patients at 8 weeks
inhibitors in the inflammatory bowel disease setting. In and steroid-free clinical remission in 7 (27%) of 26 at
ulcerative colitis, a breakdown of mucosal integrity exists 12 months after starting treatment.93 However, the FDA
with luminal microflora stimulating an uninhibited and EMA have placed warnings concerning tofacitinib
proinflammatory response. Innate lymphoid cells, Th9, 10 mg twice daily in patients with ulcerative colitis due to
and IL-13-producing natural killer cells all contribute the risk of thromboembolic events, as discussed later.
to epithelial damage and chronic inflam­ mation.86 In For Crohn’s disease, two phase 2b studies investigated
Crohn’s disease, the pathogenesis is also multifactorial, tofacitinib induction and maintenance in moderate-to-
with polarised Th1 responses under the control of IL-12, severe Crohn’s disease, yet in contrast to ulcerative
signalling via JAK2-TYK2, and IL-27, signalling via colitis, although minor treatment effects were noted,
JAK1, JAK2, and TYK2, among others.87 Genome wide efficacy endpoints were not significantly different
association studies have shown a significant association from placebo.94 This result might reflect the dif­
between Crohn’s disease and the IL23R gene, high­ ferential immune-mediated pathogenesis compared
lighting the proinflammatory role of IL-23 and the Th17 with ulcerative colitis, JAK inhibitors specificity in
cell pathway.88 Crohn’s disease, or trial design and patient inclusion;
Despite the recognised efficacy of biological agents therefore, JAK inhibitors therapy in Crohn’s disease
such as the TNF inhibitors, anti-integrins, and anti-IL-12 require additional exploration.
and IL-23 monoclonal antibodies, approximately more Regarding other JAK inhibitors, the FITZROY study
than 50% of patients with moderate-to-severe ulcerative investigated filgotinib in moderate-to-severe Crohn’s
colitis do not have sustained remissions.89,90 Efficacy of disease and showed better clinical efficacy than the placebo
oral tofacitinib in ulcerative colitis was reported after and acceptable safety in patients who had TNF inhibitor-
three large, placebo-controlled phase 3 trials—two trials treated and treatment-naive disease.95 Upadacitinib has

www.thelancet.com Vol 398 August 28, 2021 811


Therapeutics

been investigated in phase 2 trials for both moderate-to- As discussed, the FDA approved the baracitinib 2 mg
severe ulcerative colitis and Crohn’s disease. For moderate- dose but not the 4 mg dose due to thromboembolic risk.
to-severe ulcerative colitis, the phase 2b trial showed better For tofacitinib, a boxed warning was added in 2019 by the
remission induction efficacy than the placebo at 8 weeks.96 FDA for an increased risk of thromboembolism and
For moderate-to-severe Crohn’s disease (CELEST trial), death with the 10 mg twice daily dose in patients with
upadacitinib was superior to the placebo in inducing rheumatoid arthritis or ulcerative colitis, which was
endoscopic improvements in patients, most of whom added after increases in these events in individuals
were refractory to biologics.97 Upadacitinib efficacy is now older than 50 years who had at least one cardiovascular
being assessed in the phase 3 trial setting—eg, an risk factor. Direct thrombotic risk comparison between
ulcerative colitis trial, named U-Accomplish, comparing specific JAK inhibitors has been attempted but results
upadacitinib to placebo as assessed by the adapted are controversial.103 Although rare, gastrointestinal
Mayo score; and a Crohn’s disease trial delineating the perfora­tion has been described for a few patients with
efficacy and safety in patients with moderate-to-severe rheu­matoid arthritis (some of whom had other risk
disease who are non-responsive or intolerant to biologic factors) given either tofacitinib or baracitinib.104
therapies.98 Additional JAK inhibitors undergoing
assessment in the setting of inflammatory bowel disease Future developments and combinatorial
include peficitinib, ritlecitinib, brepocitinib, and TD-1473, approaches
a novel pan-JAK inhibitor that has marked gastro­ It is clear that JAK inhibitors have revolutionised our
intestinal specificity.99,100 Efficacy and longer-term data are therapeutic armamentarium across a pleiotropic range of
awaited, particularly as some cytokines that promote immune-mediated and inflammatory disorders, and
bowel mucosal integrity also rely on JAKs for signalling, across clonal myeloid disorders, such as myeloproliferative
and the exact use of these JAK inhibitors in the therapeutic neoplasm. As a class of agents, many patients are likely to
setting requires clarification. benefit from JAK inhibitors in the coming years; however,
increased clinical experience is required to establish the
Overview of safety concerns risk–benefit ratio for each agent in specific clinical
We have discussed specific side-effects of JAK inhibitors indications. Vigilance in the clinical community is required
and have highlighted them in the table (with a concerning the common and rare side-effects of these
haematology focus) and panel 2 (rheumatology focus). drugs, which include atypical infectious complications,
Frequently, side-effects of JAK inhibitors are mild-to- withdrawal syndrome, poten­ tial cytopenias and liver
moderate, predictable, and easy to manage, yet dose dysfunction, disturbed lipid metabolism, and risk of
modifications might be mandatory in some patients and thrombosis with some agents (depending on underlying
patients need close monitoring. Moreover, clinicians clinical condition), additional understanding of potential
should be vigilant for serious and sometimes life- risk of B-cell lymphoma,105 coupled with a need for
threatening complications. Complications include enhanced skin surveillance for non-melanoma skin
rebound phenomena, which is rare and can cause a cancers. Long-term efficacy data and postmarketing
severe cytokine storm and systemic inflam­ matory surveillance is required for all current JAK inhibitors.
response in patients, in particular in patients given Increasingly, combinatorial therapies with JAK inhibitors
ruxolitinib.101 On-target effects of agents such as are being explored, aiming to maximise treatment
ruxolitinib include expected cytopenias (predominantly responses after recognition that deregulated JAK–STAT
JAK2 related) and, due to immunosuppressive proper­ signalling is not the only pathogenetic mechanism in
ties, JAK inhibitors have a heterogeneous risk of disorders such as myelo­fibrosis and rheumatoid arthritis.
infectious complications.7,23,24,31,58,102 Depending on the Drugs can be given simultaneously or sequenced—eg, a
agent, these infectious complications can include upper trial of the histone deacetylase inhibitor, pracinostat, and
respiratory and urinary tract infections and, in general, ruxolitinib in patients with myelofibrosis in which patients
herpes zoster reactivation, which could be considered were given 3 months of ruxolitinib and then pracrinostat.
a class effect. Atypical infections, such as Mycobacterium Novel combinatorial approaches in myelofibrosis with JAK
tuberculosis reactivation and John Cunningham virus- inhibitors include targeting the neuro–haematopoietic axis
mediated neurological disease, have been described with and targeted inhibition of nuclear–cytoplasmic transport
ruxolitinib (reviewed by McLornan and colleagues).7 (leading to accumulation of p53).106,107 From a practical
Monitoring of renal and liver function is needed and stance, the future could bring access to generic JAK
hyperlipidaemia could be considered a potential class inhibitors within rheumatoid arthritis and myelopro­
effect of JAK inhibitors, with marked phenotypic liferative neoplasms disease areas and rapidly change the
variability, requiring lipid monitoring. As discussed, therapeutic landscape.
fedratinib has a black box warning for encephalopathy As the exciting field of JAK inhibitors progresses,
risk, after earlier cases of Wernicke’s encephalopathy, clinicians need to understand mechanisms of resistance
and momelotinib is associated with low level (usually or loss of response to JAK inhibitors, with particular
≤2 grade) sensory neuropathy.27,28,31,32,38 relevance to myeloproliferative neoplasm, and how best

812 www.thelancet.com Vol 398 August 28, 2021


Therapeutics

these can be attenuated. It is well established that surely be an attractive goal across the entire range of
myeloproliferative neoplasm cells can become persistent disorders we have discussed.
to type I JAK inhibitors (which stabilise the active kinase Contributors
conformation) and, in vitro, reversal is possible by use of The first draft was written by DPM, JEP, and CNH. JG wrote sections
a type II inhibitor (which bind to JAK2 in the inactive and made additional edits to the manuscript. All authors did the
literature search, interpreted data and evidence for this review article,
conformation), such as CHZ868; although, cell line and approved the final manuscript.
research has also shown that the Leu884Pro mutation of
Declaration of interests
JAK2 can confer resistance to type II inhibitors by DPM reports speaker and advisory board fees from JAZZ
weakening the interaction between inhibitor and pharmaceuticals and Novartis. JEP reports speaker and advisory board
target.108–110 Whether reversal of so-called persistence can fees from Pfizer, Lily, AbbVie, and Gilead Pharmaceuticals. JG reports
be clinically achieved with other similar agents is an funding for clinical trial conduct, advisory boards, and travel
reimbursement from Incyte; funding for clinical trial conduct, advisory
unknown. There are case reports of disease responsiveness board funding, and travel reimbursement from Celgene; funding for
in myelofibrosis being reset after withdrawal and clinical trial conduct from CTI Biopharma; and funding for clinical trial
introduction of ruxolitinib.111 Finally, concerning resis­ conduct and advisory boards from Gilead. CNH reports speaker and
tance, unlike BCR–ABL tyrosine kinase inhibitors, there advisory board fees from Novarti, Gilead, CTI Biopharma, and Celgene;
speaker fees from JANNSEN; advisory board fees from AOP Pharma,
have been no reports of acquired point mutations Roche Pharmaceuticals, and Sierra Oncology.
conferring resistance to clinically used JAK inhibitors.
References
It is not known whether JAK inhibitors should be 1 Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling
used earlier in the disease process (eg, in myelofibrosis pathway. J Cell Sci 2004; 117: 1281–83.
and rheumatoid arthritis) and whether JAK inhibitors 2 Haan C, Kreis S, Margue C, Behrmann I. Jaks and cytokine
receptors—an intimate relationship. Biochem Pharmacol 2006;
should be introduced in front-line settings for 72: 1538–46.
conventionally low-risk or less advanced disease in 3 Darnell JE, Kerr IM, Stark GR. JAK–STAT pathways and
an attempt to delay disease progression. Any such transcriptional activation in response to IFNs and other
extracellular signaling proteins. Science 1994; 264: 1415–21.
interventions need to be investigated in clinical trials to
4 Stark GR, Darnell JE. The JAK–STAT pathway at twenty. Immunity
gain maximum knowledge and an understanding of the 2012; 36: 503–14.
risk–benefit ratio. 5 Liongue C, Sertori R, Ward AC. Evolution of cytokine receptor
The medical community will see these agents being signaling. J Immunol 2016; 197: 11–18.
6 Igaz P, Tóth S, Falus A. Biological and clinical significance of the
increasingly used to treat autoinflammatory condi­ JAK–STAT pathway; lessons from knockout mice. Inflamm Res
tions—eg, JAK inhibitors are being assessed for use in 2001; 50: 435–41.
rare autoinflammatory interferonopathies, such as 7 McLornan DP, Khan AA, Harrison CN. Immunological
consequences of JAK inhibition: friend or foe?
STING-associated vasculopathy with onset in infancy Curr Hematol Malig Rep 2015; 10: 370–79.
syndrome and chronic atypical neutrophilic dermatoses 8 Richard AJ, Stephens JM. The role of JAK–STAT signaling in
with lipodys­ trophy and elevated temperature syn­ adipose tissue function. Biochim Biophys Acta 2014; 1842: 431–39.
drome.112 These interferonopathies possess a prominent 9 Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function
mutation of JAK2 in myeloproliferative disorders. N Engl J Med
type I IFN gene signature and, typically, do not respond 2005; 352: 1779–90.
to IL-1 and TNF blockade. Sanchez and colleagues113 10 James C, Ugo V, Le Couédic J-P, et al. A unique clonal JAK2
reported on the use of baricitinib in 18 patients mutation leading to constitutive signalling causes polycythaemia
vera. Nature 2005; 434: 1144–48.
with chronic atypical neutrophilic dermatoses with 11 Baxter EJ, Scott LM, Campbell PJ, et al. Acquired mutation of the
lipodystrophy and elevated temperature syndrome, tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet
STING-associated vasculopathy with onset in infancy 2005; 365: 1054–61.
syndrome, Aicardi-Goutières syndrome, or undefined 12 Levine RL, Wadleigh M, Cools J, et al. Activating mutation in the
tyrosine kinase JAK2 in polycythemia vera, essential
interferonopathy. Baracitinib treatment was associated thrombocythemia, and myeloid metaplasia with myelofibrosis.
with heterogeneous improve­ ments in interferon and Cancer Cell 2005; 7: 387–97.
inflammatory biomarker measure­ ments and with 13 Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations
of calreticulin in myeloproliferative neoplasms. N Engl J Med 2013;
clinical manifestations in some patients. Long-term 369: 2379–90.
safety data are awaited. It is the broad anti-inflammatory 14 Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, O’Shea JJ.
actions of JAK inhibitors that have led to these agents JAK inhibition as a therapeutic strategy for immune and
inflammatory diseases. Nat Rev Drug Discov 2017; 16: 843–62.
being evaluated in the management of SARS-CoV-2 15 O’Shea JJ, Holland SM, Staudt LM. JAKs and STATs in
with heterogeneous results.113 immunity, immunodeficiency, and cancer. N Engl J Med 2013;
Ultimately, potential disease-associated immune 368: 161–70.
signatures might aid clinicians to select which patients 16 Pencik J, Pham HTT, Schmoellerl J, et al. JAK–STAT signaling in
cancer: from cytokines to non-coding genome. Cytokine 2016;
across disease groups who will benefit most from a 87: 26–36.
particular JAK inhibitor, being of best clinical use in 17 Mohanty SK, Yagiz K, Pradhan D, et al. STAT3 and STAT5A are
disease areas such as rheumatoid arthritis and mye­ potential therapeutic targets in castration-resistant prostate cancer.
Oncotarget 2017; 8: 85997–6010.
loproliferative neoplasm, in which there are multiple 18 Stuart E, Buchert M, Putoczki T, et al. Therapeutic inhibition of
licensed products and more on the horizon. Personalised Jak activity inhibits progression of gastrointestinal tumors in mice.
stratification to appropriate JAK inhibitors therapy will Mol Cancer Ther 2014; 13: 468–74.

www.thelancet.com Vol 398 August 28, 2021 813


Therapeutics

19 Ishizaki M, Akimoto T, Muromoto R, et al. Involvement of tyrosine 40 Gerds AT, Savona M, Scott BL, et al. Results of PAC203:
kinase-2 in both the IL-12/Th1 and IL-23/Th17 axes in vivo. a randomized phase 2 dose-finding study and determination of
J Immunol 2011; 187: 181–89. the recommended dose of pacritinib. Blood 2019;
20 Prutsch N, Gurnhofer E, Suske T, et al. Dependency on the 134 (suppl 1): 667.
TYK2/STAT1/MCL1 axis in anaplastic large cell lymphoma. 41 Blazar BR, Murphy WJ, Abedi M. Advances in graft-versus-host
Leukemia 2019; 33: 696–709. disease biology and therapy. Nat Rev Immunol 2012; 12: 443–58.
21 Papp K, Gordon K, Thaçi D, et al. Phase 2 trial of selective 42 Spoerl S, Mathew NR, Bscheider M, et al. Activity of therapeutic
tyrosine kinase 2 inhibition in psoriasis. N Engl J Med 2018; JAK 1/2 blockade in graft-versus-host disease. Blood 2014;
379: 1313–21. 123: 3832–42.
22 Batista CR, Lim M, Laramée A-S, et al. Driver mutations in Janus 43 Zeiser R, Burchert A, Lengerke C, et al. Ruxolitinib in
kinases in a mouse model of B-cell leukemia induced by deletion of corticosteroid-refractory graft-versus-host disease after allogeneic
PU.1 and Spi-B. Blood Adv 2018; 2: 2798–810. stem cell transplantation: a multicenter survey. Leukemia 2015;
23 Harrison C, Kiladjian J-J, Al-Ali HK, et al. JAK inhibition with 29: 2062–68.
ruxolitinib versus best available therapy for myelofibrosis. 44 US Food and Drug Administration. FDA approves ruxolitinib for
N Engl J Med 2012; 366: 787–98. acute graft-versus-host disease. May 24, 2019. https://www.fda.gov/
24 Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo- drugs/resources-information-approved-drugs/fda-approves-
controlled trial of ruxolitinib for myelofibrosis. N Engl J Med 2012; ruxolitinib-acute-graft-versus-host-disease (accessed July 7, 2020).
366: 799–807. 45 Jagasia M, Zeiser R, Arbushites M, Delaite P, Gadbaw B,
25 Al-Ali HK, Griesshammer M, Foltz L, et al. Primary analysis of Bubnoff N. Ruxolitinib for the treatment of patients with steroid-
JUMP, a phase 3b, expanded-access study evaluating the safety and refractory GVHD: an introduction to the REACH trials.
efficacy of ruxolitinib in patients with myelofibrosis, including Immunotherapy 2018; 10: 391–402.
those with low platelet counts. Br J Haematol 2020; 189: 888–903. 46 Zeiser R, von Bubnoff N, Butler J, et al. Ruxolitinib for
26 Mead AJ, Milojkovic D, Knapper S, et al. Response to ruxolitinib in glucocorticoid-refractory acute graft-versus-host disease.
patients with intermediate-1-, intermediate-2-, and high-risk N Engl J Med 2020; 382: 1800–10.
myelofibrosis: results of the UK ROBUST trial. Br J Haematol 2015; 47 Virtanen AT, Haikarainen T, Raivola J, Silvennoinen O. Selective
170: 29–39. JAKinibs: prospects in inflammatory and autoimmune diseases.
27 Mesa RA, Kiladjian J-J, Catalano JV, et al. SIMPLIFY-1: a phase III BioDrugs 2019; 33: 15–32.
randomized trial of momelotinib versus ruxolitinib in janus kinase 48 Jegatheeswaran J, Turk M, Pope JE. Comparison of Janus kinase
inhibitor-naïve patients with myelofibrosis. J Clin Oncol 2017; inhibitors in the treatment of rheumatoid arthritis: a systemic
35: 3844–50. literature review. Immunotherapy 2019; 11: 737–54.
28 Harrison CN, Vannucchi AM, Platzbecker U, et al. Momelotinib 49 AbbVie. AbbVie receives FDA approval of RINVOQ™
versus best available therapy in patients with myelofibrosis (upadacitinib), an oral JAK inhibitor for the treatment of moderate
previously treated with ruxolitinib (SIMPLIFY 2): a randomised, to severe rheumatoid arthritis. Aug 16, 2019. https://news.abbvie.
open-label, phase 3 trial. Lancet Haematol 2018; 5: e73–81. com/news/press-releases/abbvie-receives-fda-approval-rinvoq-
29 Mesa RA, Vannucchi AM, Mead A, et al. Pacritinib versus best upadacitinib-an-oral-jak-inhibitor-for-treatment-moderate-to-severe-
available therapy for the treatment of myelofibrosis irrespective of rheumatoid-arthritis.htm (accessed July 7, 2020).
baseline cytopenias (PERSIST-1): an international, randomised, 50 Westhovens R. Clinical efficacy of new JAK inhibitors under
phase 3 trial. Lancet Haematol 2017; 4: e225–36. development. Just more of the same? Rheumatology 2019; 58: i27–33.
30 Mascarenhas J, Hoffman R, Talpaz M, et al. Pacritinib vs best 51 Felson DT, Anderson JJ, Boers M, et al. American College of
available therapy, including ruxolitinib, in patients with myelofibrosis: Rheumatology. Preliminary definition of improvement in
a randomized clinical trial. JAMA Oncol 2018; 4: 652–59. rheumatoid arthritis. Arthritis Rheum 1995; 38: 727–35.
31 Pardanani A, Harrison C, Cortes JE, et al. Safety and efficacy of 52 Fransen J, Creemers MCW, Van Riel PLCM. Remission in
fedratinib in patients with primary or secondary myelofibrosis: rheumatoid arthritis: agreement of the disease activity score
a randomized clinical trial. JAMA Oncol 2015; 1: 643–51. (DAS28) with the ARA preliminary remission criteria.
32 Harrison CN, Schaap N, Vannucchi AM, et al. Fedratinib in Rheumatology (Oxford) 2004; 43: 1252–55.
patients with myelofibrosis previously treated with ruxolitinib: 53 Westhovens R, Taylor PC, Alten R, et al. Filgotinib
an updated analysis of the JAKARTA2 study using stringent criteria (GLPG0634/GS-6034), an oral JAK1 selective inhibitor, is effective
for ruxolitinib failure. Am J Hematol 2020; 95: 594–60. in combination with methotrexate (MTX) in patients with active
33 Vannucchi AM, Kiladjian JJ, Griesshammer M, et al. Ruxolitinib rheumatoid arthritis and insufficient response to MTX: results from
versus standard therapy for the treatment of polycythemia vera. a randomised, dose-finding study (DARWIN 1). Ann Rheum Dis
N Engl J Med 2015; 372: 426–35. 2017; 76: 998–1008.
34 Passamonti F, Griesshammer M, Palandri F, et al. Ruxolitinib for 54 Kavanaugh A, Kremer J, Ponce L, et al. Filgotinib
the treatment of inadequately controlled polycythaemia vera (GLPG0634/GS-6034), an oral selective JAK1 inhibitor, is effective
without splenomegaly (RESPONSE-2): a randomised, open-label, as monotherapy in patients with active rheumatoid arthritis:
phase 3b study. Lancet Oncol 2017; 18: 88–99. results from a randomised, dose-finding study (DARWIN 2).
35 Moodley D, Yoshida H, Mostafavi S, et al. Network pharmacology of Ann Rheum Dis 2017; 76: 1009–19.
JAK inhibitors. Proc Natl Acad Sci USA 2016; 113: 9852–57. 55 Kremer JM, Emery P, Camp HS, et al. A phase IIb study of
36 Xu H, Jesson MI, Seneviratne UI, et al. PF-06651600, a dual ABT-494, a selective JAK-1 inhibitor, in patients with rheumatoid
JAK3/TEC family kinase inhibitor. ACS Chem Biol 2019; 14: 1235–42. arthritis and an inadequate response to anti-tumor necrosis factor
therapy: ABT-494 in patients with ra and inadequate response to
37 Harrison CN, Vannucchi AM, Kiladjian J-J, et al. Long-term
anti-TNF therapy. Arthritis Rheumatol 2016; 68: 2867–77.
findings from COMFORT-II, a phase 3 study of ruxolitinib vs best
available therapy for myelofibrosis. Leukemia 2017; 31: 775. 56 Genovese MC, Smolen JS, Weinblatt ME, et al. Efficacy and safety
of ABT-494, a selective JAK-1 inhibitor, in a phase IIb study in
38 Celgene Corporation. Celgene Corporation (NASDAQ: CELG) today
patients with rheumatoid arthritis and an inadequate response to
announced the U.S. Food and Drug Administration (FDA) has
methotrexate. Arthritis Rheumatol 2016; 68: 2857–66.
approved INREBIC® (fedratinib) for the treatment of adult patients
with intermediate-2 or high-risk primary or secondary 57 Dore RK, Antonova J, Burudpakdee C, Wang X, Ozbay B,
(post-polycythemia vera or post-essential thrombocythemia) Genovese MC. Treatment patterns, persistence, and durability in
myelofibrosis. Aug 16, 2019. https://ir.celgene.com/press-releases- rheumatoid arthritis patients with inadequate response to biologic
archive/press-release-details/2019/US-FDA-Approves-INREBIC- disease-modifying anti-rheumatic drugs (BDMARDS).
Fedratinib-as-First-New-Treatment-in-Nearly-a-Decade-for-Patients- Ann Rheum Dis 2019; 78 (suppl 2): 1135 (abstr SAT0133).
With-Myelofibrosis/default.aspx (accessed July 7, 2020). 58 Wollenhaupt J, Lee E-B, Curtis JR, et al. Safety and efficacy of
39 Harrison CN, McLornan DP. Current treatment algorithm for the tofacitinib for up to 9·5 years in the treatment of rheumatoid
management of patients with myelofibrosis, JAK inhibitors, and arthritis: final results of a global, open-label, long-term extension
beyond. Hematology 2017; 2017: 489–97. study. Arthritis Res Ther 2019; 21: 89.

814 www.thelancet.com Vol 398 August 28, 2021


Therapeutics

59 Fleischmann R, Mysler E, Hall S, et al. Efficacy and safety of 76 Kennedy Crispin M, Ko JM, Craiglow BG, et al. Safety and efficacy
tofacitinib monotherapy, tofacitinib with methotrexate, and of the JAK inhibitor tofacitinib citrate in patients with alopecia
adalimumab with methotrexate in patients with rheumatoid areata. JCI Insight 2016; 1: e89776.
arthritis (ORAL Strategy): a phase 3b/4, double-blind, head-to-head, 77 Mackay-Wiggan J, Jabbari A, Nguyen N, et al. Oral ruxolitinib
randomised controlled trial. Lancet 2017; 390: 457–68. induces hair regrowth in patients with moderate-to-severe alopecia
60 Cohen SB, Pope J, Haraoui B, et al. Methotrexate withdrawal in areata. JCI Insight 2016; 1: e89790.
patients with rheumatoid arthritis who achieve low disease activity 78 Lilly. Lilly receives FDA breakthrough therapy designation for
with tofacitinib modified-release 11 mg once daily plus methotrexate baricitinib for the treatment of alopecia areata. March 15, 2020.
(ORAL Shift): a randomised, phase 3b/4, non-inferiority trial. https://investor.lilly.com/news-releases/news-release-details/lilly-
Lancet Rheumatol 2019; 1: e23–34. receives-fda-breakthrough-therapy-designation-baricitinib (accessed
61 Genovese MC, van Vollenhoven RF, Pacheco-Tena C, Zhang Y, July 7, 2020).
Kinnman N. VX-509 (decernotinib), an oral selective JAK-3 79 Peeva E. Oral Janus kinase inhibitors PF-06700841 and PF-06651600
inhibitor, in combination with methotrexate in patients with provide clinically evident therapeutic effect at 4 and 6 weeks in
rheumatoid arthritis. Arthritis Rheumatol 2016; 68: 46–55. patients with alopecia areata and greater efficacy over 24 weeks in
62 Fleischmann RM, Damjanov NS, Kivitz AJ, Legedza A, Hoock T, patients with a shorter duration of their current alopecia episode:
Kinnman N. A randomized, double-blind, placebo-controlled, results of a randomized phase 2a trial. American Academy of
twelve-week, dose-ranging study of decernotinib, an oral selective Dermatology Annual Meeting; Washington, DC; March 1–5, 2019.
JAK-3 inhibitor, as monotherapy in patients with active rheumatoid 80 Simpson EL, Sinclair R, Forman S, et al. Efficacy and safety of
arthritis. Arthritis Rheumatol 2015; 67: 334–43. abrocitinib in adults and adolescents with moderate-to-severe atopic
63 Mease P, Coates LC, Helliwell PS, et al. Efficacy and safety of dermatitis (JADE MONO-1): a multicentre, double-blind, randomised,
filgotinib, a selective Janus kinase 1 inhibitor, in patients with placebo-controlled, phase 3 trial. Lancet 2020; 396: 255–66.
active psoriatic arthritis (EQUATOR): results from a 81 Pfizer. Complete results from second pivotal monotherapy study of
randomised, placebo-controlled, phase 2 trial. Lancet 2018; abrocitinib published in JAMA Dermatology. June 3, 2020.
392: 2367–77. https://www.pfizer.com/news/press-release/press-release-detail/
64 Mcinnes I, Anderson J, Magrey M, et al. Efficacy and safety of complete-results-second-pivotal-monotherapy-study (accessed
upadacitinib versus placebo and adalimumab in patients with active July 7, 2020).
psoriatic arthritis and inadequate response to non-biologic disease- 82 Guttman-Yassky E, Silverberg JI, Nemoto O, et al. Baricitinib in
modifying anti-rheumatic drugs (SELECT-PsA-1): a double-blind, adult patients with moderate-to-severe atopic dermatitis: a phase 2
randomized controlled phase 3 trial. Ann Rheum Dis 2020; parallel, double-blinded, randomized placebo-controlled multiple-
79 (suppl 1): 16 (abstr LB0001). dose study. J Am Acad Dermatol 2019; 80: 913–21.e9.
65 Genovese MC, Lertratanakul A, Anderson J, et al. Efficacy and 83 Guttman-Yassky E, Thaçi D, Pangan AL, et al. Upadacitinib in
safety of upadacitinib in patients with active psoriatic arthritis and adults with moderate to severe atopic dermatitis: 16-week results
inadequate response to biologic disease-modifying anti-rheumatic from a randomized, placebo-controlled trial. J Allergy Clin Immunol
drugs (SELECT-PSA-2): a double-blind, randomized controlled 2020; 145: 877–84.
phase 3 trial. Ann Rheum Dis 2020; 79 (suppl 1): 139 (abstr OP0223). 84 Kim BS, Howell MD, Sun K, Papp K, Nasir A, Kuligowski ME.
66 van der Heijde D, Song I-H, Pangan AL, et al. Efficacy and safety of Treatment of atopic dermatitis with ruxolitinib cream (JAK1/JAK2
upadacitinib in patients with active ankylosing spondylitis inhibitor) or triamcinolone cream. J Allergy Clin Immunol 2020;
(SELECT-AXIS 1): a multicentre, randomised, double-blind, 145: 572–82.
placebo-controlled, phase 2/3 trial. Lancet 2019; 394: 2108–17. 85 Rosmarin D, Pandya AG, Lebwohl M, et al. Ruxolitinib cream for
67 van der Heijde D, Baraliakos X, Gensler LS, et al. Efficacy and safety treatment of vitiligo: a randomised, controlled, phase 2 trial. Lancet
of filgotinib, a selective Janus kinase 1 inhibitor, in patients with 2020; 396: 110–20.
active ankylosing spondylitis (TORTUGA): results from a 86 de Mattos BRR, Garcia MPG, Nogueira JB, et al. Inflammatory
randomised, placebo-controlled, phase 2 trial. Lancet 2018; bowel disease: an overview of immune mechanisms and biological
392: 2378–87. treatments. Mediators Inflamm 2015; 2015: 493012.
68 Hagberg N, Joelsson M, Leonard D, et al. The STAT4 SLE risk allele 87 Moschen AR, Tilg H, Raine T. IL-12, IL-23 and IL-17 in IBD:
rs7574865[T] is associated with increased IL-12-induced IFN-γ immunobiology and therapeutic targeting.
production in T cells from patients with SLE. Ann Rheum Dis 2018; Nat Rev Gastroenterol Hepatol 2019; 16: 185–96.
77: 1070–77.
88 Liu JZ, van Sommeren S, Huang H, et al. Association analyses
69 Wallace DJ, Furie RA, Tanaka Y, et al. Baricitinib for systemic lupus identify 38 susceptibility loci for inflammatory bowel disease and
erythematosus: a double-blind, randomised, placebo-controlled, highlight shared genetic risk across populations. Nat Genet 2015;
phase 2 trial. Lancet 2018; 392: 222–31. 47: 979–86.
70 Yuan K, Huang G, Sang X, Xu A. Baricitinib for systemic lupus 89 Danese S, Argollo M, Le Berre C, Peyrin-Biroulet L. JAK selectivity
erythematosus. Lancet 2019; 393: 402. for inflammatory bowel disease treatment: does it clinically matter?
71 Papp KA, Menter MA, Abe M, et al. Tofacitinib, an oral Janus kinase Gut 2019; 68: 1893–99.
inhibitor, for the treatment of chronic plaque psoriasis: results from 90 Danese S, Allez M, van Bodegraven AA, et al. Unmet medical needs
two randomized, placebo-controlled, phase III trials. Br J Dermatol in ulcerative colitis: an expert group consensus. Dig Dis 2019;
2015; 173: 949–61. 37: 266–83.
72 Bachelez H, van de Kerkhof PCM, Strohal R, et al. Tofacitinib 91 Sandborn WJ, Su C, Sands BE, et al. Tofacitinib as induction and
versus etanercept or placebo in moderate-to-severe chronic plaque maintenance therapy for ulcerative colitis. N Engl J Med 2017;
psoriasis: a phase 3 randomised non-inferiority trial. Lancet 2015; 376: 1723–36.
386: 552–61.
92 Rubin DT, Ananthakrishnan AN, Siegel CA, Sauer BG, Long MD.
73 Bristol Myers Squibb. Bristol Myers Squibb announces ACG clinical guideline: ulcerative colitis in adults.
deucravacitinib (BMS-986165) demonstrated superiority to placebo Am J Gastroenterol 2019; 114: 384–413.
and Otezla® (apremilast) in pivotal phase 3 psoriasis study.
93 Weisshof R, Aharoni Golan M, Sossenheimer PH, et al. Real-world
March 11, 2020. https://news.bms.com/news/details/2020/Bristol-
experience with tofacitinib in IBD at a tertiary center. Dig Dis Sci
Myers-Squibb-Announces-Deucravacitinib-BMS-986165-
2019; 64: 1945–51.
Demonstrated-Superiority-to-Placebo-and-Otezla-apremilast-in-
Pivotal-Phase-3-Psoriasis-Study/default.aspx (accessed 94 Panés J, Sandborn WJ, Schreiber S, et al. Tofacitinib for induction
July 7, 2020). and maintenance therapy of Crohn’s disease: results of two phase IIb
randomised placebo-controlled trials. Gut 2017; 66: 1049–59.
74 Islam N, Leung PSC, Huntley AC, Eric Gershwin M.
The autoimmune basis of alopecia areata: a comprehensive review. 95 Vermeire S, Schreiber S, Petryka R, et al. Clinical remission in
Autoimmun Rev 2015; 14: 81–89. patients with moderate-to-severe Crohn’s disease treated with
filgotinib (the FITZROY study): results from a phase 2, double-
75 Phan K, Sebaratnam DF. JAK inhibitors for alopecia areata:
blind, randomised, placebo-controlled trial. Lancet 2017;
a systematic review and meta-analysis. J Eur Acad Dermatol Venereol
389: 266–75.
2019; 33: 850–56.

www.thelancet.com Vol 398 August 28, 2021 815


Therapeutics

96 Sandborn WJ, Ghosh S, Panes J, et al. Efficacy of upadacitinib in a 105 Rumi E. Zibellini S, Boveri E, et al. Ruxolitinib treatment and risk
randomized trial of patients with active ulcerative colitis. of B-cell lymphomas in myeloproliferative neoplasms.
Gastroenterology 2020; 158: 2139–49.e14. Am J Hematol 2019; 94: e185–88.
97 Sandborn WJ, Feagan BG, Loftus EV, et al. Efficacy and safety of 106 Herlihy N, Harrison CN, McLornan DP. Exploitation of the neural-
upadacitinib in a randomized trial of patients with Crohn’s disease. hematopoietic stem cell niche axis to treat myeloproliferative
Gastroenterology 2020; 158: 2123–38.e8. neoplasms. Haematologica 2019; 104: 639–41.
98 Sandborn WJ, Schreiber S, Lee SD, et al. Improved endoscopic 107 Yan D, Pomicter AD, Tantravahi S, et al. Nuclear-cytoplasmic
outcomes and mucosal healing of upadacitinib as an induction transport is a therapeutic target in myelofibrosis. Clin Cancer Res
therapy in adults with moderately to severely active ulcerative 2019; 25: 2323–35.
colitis: data from the U-ACHIEVE study. J Crohns Colitis 2019; 108 Koppikar P, Bhagwat N, Kilpivaara O, et al. Heterodimeric
13 (suppl 1): S009 JAK–STAT activation as a mechanism of persistence to JAK2
99 D’Amico F, Fiorino G, Furfaro F, Allocca M, Danese S. Janus kinase inhibitor therapy. Nature 2012; 489: 155–59.
inhibitors for the treatment of inflammatory bowel diseases: 109 Kong X, Sun H, Pan P, et al. How does the L884P mutation confer
developments from phase I and phase II clinical trials. resistance to type-II inhibitors of JAK2 kinase: a comprehensive
Expert Opin Investig Drugs 2018; 27: 595–99. molecular modeling study. Sci Rep 2017; 7: 9088.
100 Ma C, Lee JK, Mitra AR, et al. Systematic review with meta-analysis: 110 Meyer SC, Keller MD, Chiu S, et al. CHZ868, a type II JAK2
efficacy and safety of oral Janus kinase inhibitors for inflammatory inhibitor, reverses type I JAK inhibitor persistence and demonstrates
bowel disease. Aliment Pharmacol Ther 2019; 50: 5–23. efficacy in myeloproliferative neoplasms. Cancer Cell 2015; 28: 15–28.
101 Palandri F, Palumbo GA, Elli EM, et al. Ruxolitinib discontinuation 111 Gisslinger H, Schalling M, Gisslinger B, Skrabs C, Müllauer L,
syndrome: incidence, risk factors, and management in 251 patients Kralovics R. Restoration of response to ruxolitinib upon brief
with myelofibrosis. Blood Cancer J 2021; 11: 4. withdrawal in two patients with myelofibrosis. Am J Hematol 2014;
102 Jegatheeswaran J, Turk M, Pope JE. Comparison of Janus kinase 89: 344–46.
inhibitors in the treatment of rheumatoid arthritis: a systemic 112 Sanchez GAM, Reinhardt A, Ramsey S, et al. JAK1/2 inhibition
literature review. Immunotherapy 2019; 11: 737–54. with baricitinib in the treatment of autoinflammatory
103 Scott IC, Hider SL, Scott DL. Thromboembolism with Janus kinase interferonopathies. J Clin Invest 2018; 128: 3041–52.
(JAK) inhibitors for rheumatoid arthritis: how real is the risk? 113 D’Alessio A, Del Poggio P, Bracchi F, et al. Low-dose ruxolitinib
Drug Saf 2018; 41: 645–53. plus steroid in severe SARS-CoV-2 pneumonia. Leukemia 2020;
104 Xie F, Yun H, Bernatsky S, Curtis JR. Brief report: risk of 35: 635–38.
gastrointestinal perforation among rheumatoid arthritis patients
receiving tofacitinib, tocilizumab, or other biologic treatments. © 2021 Elsevier Ltd. All rights reserved.
Arthritis Rheumatol 2016; 68: 2612–17.

816 www.thelancet.com Vol 398 August 28, 2021

You might also like