TG101348

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 transcription (STAT) signalling in multiple disease states has led to an increasing applicability of therapeutic intervention with JAK inhibitors. These agents have revolutionised treatments for a heterogeneous group of disorders, such as myeloproliferative neoplasms, rheumatoid arthritis, inflammatory bowel disease, and multiple immune-driven dermatological diseases, exemplifying rapid bench-to-bedside translation. In this Therapeutics paper, we summarise the currently available data concerning the successes and safety of an array of JAK inhibitors and hypothesise on how these fields could develop.

Lancet 2021; 398: 803–16
Department of Haematology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
(D P McLornan PhD, Prof C N Harrison DM);
Department of Rheumatology,
University of Western Ontario, London, ON, Canada

Introduction
Janus kinases (JAKs) are multidomain non-receptor tyrosine kinases that have pivotal roles in cellular signal transduction. The targeting of JAK-associated pathways through the use of JAK inhibitors has rapidly entered the clinical arena for a wide array of disease states, including myeloproliferative neoplasms, rheumatoid arthritis and other immune-mediated arthropathies, numerous inflam- matory dermatological disorders, and inflammatory bowel disease, exemplifying rapid bench-to-bedside translation and modifying many classic treatment algorithms. Clinicians need to be up to date with this rapidly changing field, given the unique opportunities afforded by JAK inhibitors in the modulation of signalling and pathogenetic immune responses in many disease areas. Furthermore, an understanding is required of the potential safety concerns and limitations of JAK inhibitors, which had halted many initially promising clinical trials. In this Therapeutics paper, we provide an expert, up-to-date overview of JAK inhibitors and discuss how this complex 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 scenarios and have revolutionised many traditional treatment pathways. Other potential applications in a disparate group of disorders are being developed and we await evaluation with interest.

JAK–STAT signalling in health and disease
In humans, the JAK family comprises JAK1, JAK2, JAK3, and TYK2.1–4 Irrespective of type, all cytokine receptors are associated with one or more of the JAKs to facilitate signal
transduction (figure). Activation and transphosphorylation of JAKs induces signal transducer and activator of transcription (STAT) recruitment, dimerisation, nuclear translocation, and resultant transcriptional responses.2,5 JAK signalling is tightly regulated at multiple levels, cross talk with many other intracellular pathways is evident, and full elucidation of these complex networks is ongoing. This evolutionary conserved pathway can be deregulated in many disease states, most frequently through acquired activating mutations or gene amplification.
gp130
JAK–STAT signalling has a pivotal role in a pleotropic range of systems, including the orchestration and functional capability of immune responses, in particular T-cell polarisation, control of haematopoiesis and
(Prof J E Pope MD); Division of Hematology, Stanford
University School of Medicine, Stanford Cancer Institute,
Stanford, CA, USA (Prof J Gotlib MD) Correspondence to:
Prof Claire Harrison, Department
of Haematology, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 9RT, UK [email protected]

Search strategy and selection criteria
We identified references for this Therapeutics paper by searching MEDLINE, PubMed, and references from relevant articles using relevant search terms, such as “Jak inhibitor”, “myeloproliferative”, and “rheumatoid arthritis”, and searching between Jan 1, 1995, and Dec 31, 2020. Only articles published in English were included. Abstracts and reports from meetings were included only when we deemed them of pivotal importance to the readership.
Figure: Simplified overview of JAK–STAT signalling via EPO receptor and IL-6 receptor activation Cytokine receptors do not have intrinsic kinase or phosphatase activity. For ligands such as EPO, the receptor subunits are bound as homodimers, whereas, for interleukins and interferons, the receptor subunits are
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 activation. Prosurvival pathways, such as the PI3K–AKT and RAS–RAF–EFL pathways, can be upregulated.
EFL=elongation factor-like GTPase. ERK=extracellular signal-regulated kinase. JAK=Janus kinase. MEK=mitogen- activated protein kinase. P=phosphorylation. PI3K=phosphoinositide 3-kinase. RAF=Raf family kinases.
RAS=Ras family GTPases. STAT=signal transducer and activator of transcription.

Panel 1: Drug selectivity profiles
Drug (suggested selectivity against Janus kinase [JAK] family members)
⦁ Ruxolitinib (JAK1 and JAK2 >JAK3 >TYK2)
⦁ Baricitinib (JAK1 and JAK2, and moderate activity against TYK2)
⦁ Tofacitinib (JAK3 >JAK2 >JAK1)
⦁ Fedratinib (JAK2)
⦁ Momelotinib (JAK1 and JAK2)
⦁ Pacritinib (JAK2 >JAK1 and JAK3)
⦁ Fligotinib (JAK1 >JAK2 >JAK3 and TYK2)
⦁ Upadacitinib (JAK1 >JAK2 and JAK3)
⦁ Itacitinib (JAK1)
⦁ Decernotinib (JAK3)
⦁ Peficitinib (all JAK proteins)
⦁ Deucravacitinib (BMS-986165; TYK2)
⦁ Abrocitinib (JAK1)
⦁ NDI-031301 (TYK2)
⦁ Ritlecitinib (PF-06651600; JAK3)

inflammation, adipogenesis, and growth.6–8 The essential role of JAKs in cytokine signalling is pivotal to under- standing how many cytokine-dependent inflammatory, autoimmune, and neoplastic disorders associated with deregulated activity can be manipulated and targeted through therapeutic JAK inhibition.
Within haematology, the largest area of clinical expan- sion for JAK inhibitors has been in the myeloproliferative neoplasm field. The JAK2 Val617Phe mutation, leading to constitutive activation, is located in the JH2 domain and is found in approximately 60% of individuals with myelofibrosis, 50–60% with essential thrombocythaemia, and 97–98% with polycythaemia vera.9–12 Furthermore, irrespective of JAK2 mutational status, many myeloproliferative neoplasm disorders are characterised by upregulated JAK–STAT signalling via mutations in additional canonical genes, such as CALR.13 There has been rapid clinical development of JAK inhibitors and, in 2011, ruxolitinib (a JAK1 and JAK2 inhibitor) was the first JAK inhibitor to be approved for use in myelofibrosis by the US Food and Drug Administration (FDA) and European Medicines Agency (EMA).
Deregulated JAK–STAT signalling contributes to autoimmune and chronic inflammatory phenotypes, often in a disease-independent way, and JAK-dependent cytokines (eg, IL-6) are often pivotal pathological drivers, thus explaining the rationale of JAK inhibitors in these disorders.14 Both first and later generation JAK in- hibitors have been explored across a disparate group of disorders, including atopic dermatitis and alopecia areata, immune-mediated arthropathies, inflammatory bowel disease, and interferonopathies, as we will discuss in this Therapeutics paper. Despite many overlapping pathogenetic autoimmune mechanisms, specific drug therapeutic efficacy can vary markedly.
In the oncology field, many solid tumours have aberrant JAK signalling as part of a prosurvival phenotype and to facilitate tumour migration.15 In depth review is outside the remit of this article, but clinically relevant examples of JAK in oncology include JAK2 locus amplification in gastric adenocarcinoma and amplification or enhanced nuclear translocation of STAT5 in prostate cancer, offering the prospect of benefit of JAK inhibitors.15–18
With regard to targeting JAK family members other than JAK2, TYK2 is an essential regulator of both the IL-12 and T-helper (Th)1 and IL-23 and Th17 axes and the type I interferon pathways.15,19 Thus, specifically targeting TYK2 could be beneficial across a range of immune-mediated and neoplastic disorders but, historically, TYK2 inhibitor development has been challenging. TYK2 expression and dependency is prevalent in anaplastic large-cell lymphoma, and TYK2 inhibition induces apoptosis in anaplastic large-cell 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 detail.21 Finally, JAK3 activation has additionally been found in a wide array of B-cell lymphoid malignancies, highlighting several areas for potential future therapeutic exploitation.22

Overview of JAK inhibitors: current and future perspectives
Currently licensed JAK inhibitors include tofacitinib (Pfizer, New York, NY, USA), ruxolitinib (Novartis, Basel, Switzerland), fedratinib (Celgene, Summit, NJ, USA), upadacitinib (AbbVie, North Chicago, IL, USA), peficitinib (Astellas Pharma, Tokyo, Japan), and baricitinib (Eli Lilly, Indianapolis, IN, USA; panel 1). Clinically relevant selected agents are highlighted in panel 1 and table and are discussed in detail in relevant disease areas. Achievement of JAK isoform specificity is difficult due to structural homology between family members. Additionally, even if specificity is attempted, given the wide and complex roles of JAKs and the interdepend- ency of many cytokine receptors, resultant effects can be pleotropic.35 More novel JAK inhibitor compounds are in advanced development, such as ritlecitinib (previously known as PF-06651600; Pfizer), which acts as a potent inhibitor of JAK3 and tyrosine-protein kinase Tec (TEC) family kinase members (ie, BTK, BMX, ITK, RLK, and TEC) and can inhibit the functional capacity of both CD8+ T cells and natural killer cells. The dual action of ritlecitinib might have an important role in therapeutic intervention for a plethora of inflammatory diseases.36 There is still much to learn about improving target specificity in JAK–STAT signalling and how this might translate into clinical efficacy and the potential mitigation of observed side-effects. Long-term safety data of many of these agents are emerging but still require robust follow-up.

Phase Agent Study population Main findings Safety and tolerability
Myelofibrosis
COMFORT-123 3 Ruxolitinib vs
placebo 309 patients with intermediate-2 or high-risk myelofibrosis 59% of patients given ruxolitinib had ≥35% SVR at any point during the trial and impressive symptom responses;
evident survival advantage vs placebo Most common grade 3–4 adverse events were anaemia and thrombocytopenia
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 access) Ruxolitinib 1144 patients with intermediate-2 or high-risk myelofibrosis, as well as an analysis of 163 patients with
intermediate-1 myelofibrosis 56·9% of patients had ≥50% reduction in palpable spleen length at week 24 and 62·3% at week 48; 44% to 46% and 46% to 52% of patients had objective symptom responses Most common adverse event was drug related cytopenia; herpes zoster (3·6%) and influenza (3·0%) were the most common
viral infections
ROBUST26 2 (open label) Ruxolitinib 48 patients with intermediate-1, intermediate-2, and high-risk myelofibrosis Treatment success was ≥50% reduction in spleen length or
≥50% decrease in myelofibrosis symptoms at 48 weeks, and 50% of all patients and 57% of patients with
intermediate-1 risk had treatment success Most common adverse event was drug related cytopenia, similar to other studies; one case of progressive multifocal
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 JAK inhibitor-
naive disease intermediate-1 myelofibrosis inferior with regard to symptom response; transfusion rates and independence were better in the momelotinib group neuropathy
SIMPLIFY-228 3 Momelotinib
vs BAT 104 patients in the momelotinib group and 52 in the BAT group
(46 given ruxolitinib) In patients who had been given ruxolitinib, momelotinib was not superior to BAT for SVR Common side effects included cytopenia and treatment emergent peripheral
neuropathy
PERSIST-129 3 Pacritinib vs
BAT 220 patients with intermediate-1 risk or above in the pacritinib group and 107 in the BAT group Primary endpoint of SVR of ≥35% was found in 42 (19%) of 220 patients in the pacritinib group vs five (5%) of
107 patients in the BAT group (p=0·0003) Common grade 3–4 adverse events were anaemia (17%), thrombocytopenia (12%), 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 risk or above who were previously treated with ruxolitinib 31% of patients had at least ≥35% SVR by the end of the sixth cycle; 27% had ≥50% symptom response rate Grade 3–4 adverse events were anaemia and thrombocytopenia; the trial was temporarily halted due to perceived risks of
Wernicke’s encephalopathy
Polycythaemia vera
RESPONSE33 3 Ruxolitinib vs
BAT 212 patients requiring phlebotomy or who were resistant or intolerant to hydroxycarbamide with splenomegaly Primary endpoint of patients who reached both haematocrit control (<45% without phlebotomy) and
SVR >35% at week 32 was reached by 21% of the ruxolitinib group vs 0·9% in the BAT group; there were fewer thrombotic events for patients given ruxolitinib Grade 1–2 adverse events were fatigue, headache, and diarrhoea; herpes zoster infection was more common in the ruxolitinib group than in the placebo group; increased risk of non-melanoma
skin cancer
RESPONSE-234 3b Ruxolitinib vs
BAT 149 patients requiring phlebotomy or were resistant or intolerant to
hydroxycarbamide 62% of patients in the ruxolitinib group vs 19% in the BAT group had the primary endpoint of haematocrit control
(<45% without phlebotomy) at week 32 Grade 3–4 adverse events were hypertension and angina; two patients died
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

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

platelet counts less than 50 × 10⁹ cells per L (NCT03165734). Practically, if approved, pacritinib is a very attractive agent for patients with thrombocytopenic myelofibrosis, in which many other agents might be avoided altogether, or if a reduced dose is needed therefore limiting efficacy.
It must be stressed that JAK inhibitors can have limitations within the myeloproliferative neoplasm therapeutic arena, including inadequate dose density, absence of true disease modification, and, ultimately, loss of response. Many agents are being investigated along- side ruxolitinib in myelofibrosis, including azacytidine in advanced phase disease, recombinant interferons, erythropoiesis maturation agents (eg, luspatercept), bromodomain inhibitors, and histone deacetylase in- hibitors, showing the potential of dual therapy, incor- porating JAK inhibitors, to maximise responses.39

Graft-versus-host disease
Graft-versus-host disease (GVHD) represents one of the most substantial challenges after allogeneic haematopoietic stem-cell transplantation. Pathogenetic mechanisms are multifactorial but reliant, at least partly, on proinflammatory JAK–STAT signalling, not only in alloreactive effector T cells but also in dendritic cells and B cells.41 First-line therapy, often systemic steroids, is effective only in inducing complete responses in a small proportion of cases and subsequent strategies are heterogeneous. Historically, steroid-refractory acute GVHD carries high mortality rates and is still an unmet clinical need. Regarding JAK inhibitors, Spoerl and colleagues42 showed in preclinical models that ruxolitinib reduced effector T-cell proliferation, suppressed pro- inflammatory cytokines and CD4+ cell differentiation into IFN-γ and IL17A-producing cells, and increased the number of FoxP3-positive regulatory T cells. After initial clinical proof of concept, a large retrospective multi- national study evaluated the efficacy of ruxolitinib in 95 patients with steroid-refractory GVHD who had been frequently heavily pretreated.43 Excellent overall response rates, not previously observed, were shown in 44 (82%) of 54 patients with steroid-refractory acute GVHD (including 25 [46%] of 54 complete responses) and
35 (85%) of 41 patients with chronic GVHD. The 6-month overall survival rates were 79% for patients with acute GVHD and 97·4% for patients with chronic GVHD. Cytopenias and cytomegalovirus reactivations were noted in some patients. The FDA granted approval for the use of ruxolitinib in steroid-refractory acute GVHD in patients who are 12 years and older, and, more recently, both the REACH-2 and REACH-3 phase 3 trials have shown efficacy (evaluating ruxolitinib in steroid- refractory acute GVHD and chronic GVHD).44–46

Rheumatological disorders
Rheumatoid arthritis
Contemporary treatment of rheumatoid arthritis has evolved rapidly, with many clinicians considering JAK

inhibitors for rheumatoid arthritis that is refractory to conventional synthetic disease modifying anti-rheumatic drugs (DMARDs) or biologics. As clinical knowledge accumulates, with the focus on early therapeutic inter- vention and treat-to-target strategies, it is unknown whether JAK inhibitors will become front-line treatments, although side-effects and the paucity of long-term data should be considered.
Many JAK inhibitors have been used in randomised trials for rheumatoid arthritis (eg, tofacitinib, baricitinib, upadacitinib, filgotinib, peficitinib, and decernotinib).47,48 Tofacitinib and baricitinib are approved in multiple countries, and in 2019, upadacitinib, a JAK1 inhibitor, was approved for rheumatoid arthritis in the USA and peficitinib was approved in Japan.49 Overall, there are benefits of JAK inhibitors that might be considered a class effect (eg, rapid onset of action in rheumatoid arthritis with clinical benefit) and there are also important side-effects within the class (eg, cytopenias, particularly lymphopenia; increased risk of infection, including reactivation of herpes zoster; hypercholesterolaemia; and, for some JAK inhibitors, increased thromboembolic events, which will be discussed later; panel 2).47,48 For rheumatoid arthritis, an important class effect appears to be clinical efficacy as monotherapy alongside efficacy in combination with conventional synthetic DMARD (reviewed by Westhovens).50 These agents are contra- indicated in pregnancy and breastfeeding and no evidence exists for safe use in children.
Clinical trial programmes of rheumatoid arthritis for JAK inhibitors have followed a similar sequenced trial design. JAK inhibitors show efficacy in patients with active rheumatoid arthritis that does not inadequately respond to methotrexate compared with placebo or a standard-of-care drug (eg, the biologic DMARD, adalimumab [a TNF inhibitor]). Often, JAK inhibitors are combined with background methotrexate or, less frequently, with other DMARDs. Some trials evaluated monotherapy (ie, JAK inhibitors with no background DMARDs), as discussed previously, and additional studies have been done in patients previously untreated with methotrexate.47,48 Outcome measures vary but often consist of standardised responses—eg, the American College of Rheumatology (ACR) criteria measures improvement by 20%, 50%, and 70% in cumulative response scores.51 Frequently, trials report on improve- ment in the disease activity score (ie, DAS28).52 Both the ACR criteria and DAS28 are composite scores studying changes in tender and swollen joint counts, patient global assessment, and inflammatory markers. Frequently, the proportion of patients with low disease activity or who have had remission is also reported. Low disease activity or remission outcomes are difficult to reach. Results with JAK inhibitors are similar to other advanced therapies and, when compared head to head, they are sometimes numerically and statistically better than the biologic DMARD, adalimumab.47,48 In

Panel 2: Potential side-effects and general monitoring for Janus kinase inhibitors in rheumatological diseases*

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

rheumatoid arthritis treatment, additional evaluation is needed to address topics such as: the optimal sequencing of advanced therapies after non-responsiveness to methotrexate; whether there is a benefit in switching to alternative JAK inhibitors after a patient has not responded or is intolerant; and how to prevent attenuated efficacy of JAK inhibitors. Regarding efficacy and adverse events, dose response rates of each JAK inhibitor are being clarified and long-term data are being accumulated. For example, the DARWIN 1 (using filgotinib in combination with methotrexate) and DARWIN 2 (using filgotinib monotherapy) trials showed that patients who were given filgotinib had dose-dependent responses accompanied by dose- dependent leukopenia and thrombocytopenia and early increases in high-density and low-density lipoproteins that then stabilised.53,54 DARWIN 1 and DARWIN 2 also reported increases in haemoglobin concentrations in patients given filgotinib. For upadacitinib, results of two trials (BALANCE-1 and BALANCE-2) showed dose- dependent clinical responses and higher rates of adverse events at higher doses.55,56
The advantages of JAK inhibitors in rheumatoid arthritis include oral delivery (other options are subcutaneous or intravenous), an equal or superior benefit compared with TNF inhibitors, and the possibility of better retention than TNF inhibitors, which are subject to drug induced antibodies that attenuate benefit. Better durability with JAK inhibitors in rheumatoid arthritis in patients who inadequately respond to biologic DMARDs when switching therapies is of key clinical relevance; TNF inhibitors showed shortest treatment duration and JAK inhibitors, both as monotherapy or combination, had the longest treatment persistence.57 For example, results from the ORAL Sequel long-term extension study, which evaluated open label tofacitinib 5 mg and 10 mg twice daily (for up to 9·5 years) in patients with rheumatoid arthritis who had been enrolled in phase 1, phase 2, or phase 3 index studies, showed a consistent safety profile and sustained efficacy.58 Cumulatively, 52% of patients discontinued tofacitinib, of whom, 24% discontinued due to adverse events and 4% due to tofacitinib having little of efficacy. Another area of interest is withdrawing methotrexate for patients with rheumatoid arthritis who are inad- equate responders. Two randomised controlled trials have been done with tofacitinib; switching to tofactinib from methotrexate is in fact not as efficacious as actually adding tofacitinib to methotrexate.59,60 The other strategy of adding tofacitinib to methotrexate and only randomly assigning patients with a very low disease state to stop or continue methotrexate showed that the two strategies were equal.59,60 Additionally, it is important to note that not all JAK inhibitors for rheumatoid arthritis continue development after late phase trials—eg, decernotinib (Vertex Pharmaceuticals, Boston, MA, USA), a selective JAK3 inhibitor, was investigated both as monotherapy
and alongside methotrexate, and despite early signs of efficacy, there were negative safety signals (eg, infections) and increased aminotransferase concentrations in some patients, and development was discontinued.61,62

Spondyloarthropathies
Regarding spondyloarthritis (considering both psoriatic arthritis and ankylosing spondylitis), JAK inhibitors might address multiple disease manifestations. Tofacitinib has received approval for 5 mg orally twice a day for the treatment of psoriatic arthritis in many countries (reviewed in Virtanen and colleagues47). Data have shown the positive benefits of TNF inhibitor exposure with respect to improvement in composite scores (ie, ACR criteria responses) and skin improve- ment. Efficacy was shown in patients who were previously given conventional synthetic DMARD treatment (in whom the majority were given methotrexate as back- ground treatment). Results showed an improvement in composite scores, compared with patients given the placebo, and were numerically very similar (ie, not non- inferior) to patients given adalimumab (the usual standard of care).47 With respect to joint involvement, there was no obvious advantage in exceeding the 5 mg orally twice a day dose. The EQUATOR trial investigated the efficacy of filgotinib, a more JAK1 specific inhibitor than other JAK inhibitors, versus placebo in moderate-to- severe psoriatic arthritis after intolerance or inadequate response to at least one conventional synthetic DMARD, with clinically relevant responses at 16 weeks after first dose favouring filgotinib (80% vs 33%).63 Upadacitinib is being investigated for psoriatic arthritis in the phase 3 SELECT-PsA 1 and SELECT-PsA 2 trials.64,65 For SELECT-PsA 1, 1702 patients with psoriatic arthritis who did not respond, or were intolerant, to at least one non-biologic DMARD were randomly assigned to 15 mg or 30 mg upadacitinib daily, placebo, or adalimumab (the active comparator). ACR criteria improvement by 20% (ACR20) response rates at 12 weeks were 79% for updacitinib 30 mg, 71% for updacitinib 15 mg, 65% for adalimumab, and 36% for placebo.64 Moreover, a substantial proportion of patients given upadacitinib had improvements in enthesitis and dactylitis. The SELECT-PsA 2 trial also showed that updacitinib had better efficacy than the placebo.65 Regarding ankylosing spondylitis, the TORTUGA trial showed efficacy of filgotinib versus placebo (with an early efficacy and predictable safety and tolerability), and the SELECT-AXIS 1 trial supported efficacy of upadacitinib versus placebo in patients with biologic-naive ankylosing spondylitis.66,67 Determination of long-term efficacy and safety will be important.

Systemic lupus erythematosus
The heterogeneous pathogenesis and clinical phenotype of systemic lupus erythematosus is often therapeutically challenging, necessitating novel approaches. Tofacitinib

has been investigated in a phase 1b trial for mild-to- moderate lupus based on STAT4 risk alleles, in which the presence of rs7574865A→T in individuals with lupus is associated with enhanced T-cell responses to IL-12 and IFN-γ. These enhanced T-cell responses make JAK inhibitors an attractive therapeutic option.68 In a double blind, phase 2 study, baricitinib 2 mg or 4 mg was evaluated against placebo in a 1:1:1 design of 314 adult patients with systemic lupus erythematosus and active skin or joint disease.69 The primary endpoint was assessed by improvements in the Systemic Lupus Erythematosus Disease Activity Index-2000. A once daily baricitinib 4 mg dose improved symptoms and signs of systemic lupus erythematosus in patients with poorly controlled disease (74 [67%] of 104 responded at 24 weeks vs 53% in the placebo group for skin or arthritis), whereas there was no statistically significant improvement in the 2 mg dose. Serious adverse events were seen in 9·6% of patients in the baricitinib 4 mg group versus 4·8% in the placebo group. As raised by Yuan and colleagues70 in response to the study, the 4 mg dosing strategy of baricitinib was not approved by the FDA for rheumatoid arthritis due to safety concerns of potential infectious and thrombotic complications, hence additional follow-up and more knowledge are required for this dosing regimen. Two dosing strata are being explored in the phase 3 trial, BRAVE II, with an ongoing long-term follow-up study from the earlier trials (SLE-BRAVE-X; NCT03843125).

Dermatological disorders
A large volume of evidence highlights the efficacy of JAK inhibitors in multiple skin disorders. Early focus was predominantly addressing psoriasis management, but focus now extends to many other disease states, including alopecia areata, vitiligo, and atopic dermatitis, and includes systemic and topical use.
Regarding moderate-to-severe psoriasis, Papp and colleagues71 initially evaluated the role of tofacitinib in two large phase 3 trials (OPT Pivotal 1 [n=901] and OPT Pivotal 2 [n=960]) showing efficacy of tofacitinib (5 mg or 10 mg twice daily) over placebo. In the OPT Pivotal 2 phase 3 trial, the 10 mg twice daily tofacitinib regimen was found to be non-inferior to etanercept with similar serious adverse event rates during 12 weeks of treatment, and no new safety signals emerged.72 Despite these findings, the FDA declined approval for use of tofacitinib in psoriasis in 2015 on the basis of the extent of the safety data and clinical findings, and hence additional development for this indication was halted.
Deucravacitinib (Bristol Myers Squibb) was evaluated in an initial phase 2 trial in patients with moderate-to- severe plaque psoriasis.21 Doses of 3 mg daily and higher resulted in greater clearing of psoriasis than did placebo during 12 weeks, with a favourable risk–benefit ratio. For example, week 12 Psoriasis Activity Severity Index scores between 67% and 75% were observed with doses greater than 6 mg daily, similar to those seen with

adalimumab and with the benefit of oral administration. Moreover, therapy did not appear to induce substantial haematological or liver enzyme abnormalities or raised lipid concentrations. The most common adverse events were nasopharyngitis, gastrointestinal upset, and headaches. The phase 2 efficacy of deucravacitinib led to subsequent evaluation in two large phase 3 trials, POETYK-PSO-1 and POETYK-PSO-2 (NCT03611751), in
which deucravacitinib was compared with placebo and apremilast (a selective PDE4 inhibitor). For POETYK- PSO-1, results have shown that both primary endpoints were met versus placebo by week 16 (Psoriasis Activity Severity Index 75% and a static Physician’s Global Assessment score of clear or almost clear), in addition to the secondary endpoint of superiority to apremilast.21,73 Safety data were similar to previous trials. Full results from POETYK PSO-2 are expected in late 2021.
In alopecia areata, in which hair follicle destruction is driven by CD8+ and NKG2+ T cells, multiple JAK inhibitors have shown promising efficacy.74,75 Tofacitinib 5 mg twice daily for 3 months showed efficacy in a single group trial in patients with severe disease, but responses did not have durability after discontinuation.76 An open label study of oral ruxolitinib, 20 mg twice daily for 3–6 months, showed that nine of 12 patients with alopecia areata had robust responses, with average hair growth of 90% by end of treatment.77 BRAVE-AA1 (NCT03570749) was an adaptive phase 2/3 trial investigating baricitinib 1 mg, 2 mg, or 4 mg daily in patients with severe or very severe alopecia areata. Breakthrough therapy status was granted for baricitinib in patients with alopecia areata in March, 2020.78 Reports from the phase 2 component of BRAVE-AA1 showed efficacy for both the 2 mg and 4 mg doses compared with placebo, leading to clinically significant improvements and no new safety concerns or serious adverse events.78 The phase 3 BRAVE-AA1 and BRAVE-AA2 (NCT03899259) will additionally evaluate the efficacy and safety of the 2 mg and 4 mg doses. Ritlecitinib (Pfizer) also had breakthrough therapy designation; data from the phase 2 trial in alopecia areata showed that patients given ritlecitinib significantly improved compared with those taking placebo as early as 6 weeks into treatment and met its primary endpoint in improving hair growth by week 24, as did brepocitinib.79 Additionally, CTP-543 (a JAK1 and JAK2 inhibitor, Concert Pharmaceuticals, Lexington, MA, USA) is being explored in a phase 3 alopecia areata trial—THRIVE-AA1 (NCT04518995). Top-line data for THRIVE-AA1 is expected to be reported in 2022.
A major focus has been exploring applicability of JAK inhibitors for atopic dermatitis. Abrocitinib (PF-04965842; Pfizer), which gained FDA breakthrough therapy designation for moderate-to-severe atopic dermatitis in 2018, has shown positive results compared with placebo.80 In the JADE mono-1 trial,80 387 patients were randomly assigned to abrocitinib (200 mg or 100 mg daily) or placebo for 12 weeks. Coprimary endpoints

assessed the proportion of patients who had an Investigator Global Assessment response score of 0 (clear) or 1 (almost clear) with a two-grade or better improvement from baseline, and proportion achieving 75% or better improvement in Eczema Area and Severity Index score (EASI-75). 62 (40%) of 156 patients in the abrocitinib 100 mg dosing strata and 96 (63%) of 153 in the 200 mg dosing strata had an EASI-75 response compared with nine (12%) of 76 in the placebo group (p<0·0001). Abrocitinib was well tolerated with no treatment related deaths. In the JADE mono-2 trial, both dosing strata of abrocitinib showed efficacy in patients with atopic dermatitis and, in general, both doses were well tolerated.81 The balance of efficacy or safety against the current standard of care (dupilumab) requires additional evaluation considering the slight drop in platelet count seen in some patients and consideration given to the risk of infection. Both baricitinib and upadacitinib have been investigated for use in atopic dermatitis, with the EMA granting an extension of indication for baracitinib for moderate-to-severe atopic dermatitis in September, 2020.82,83 Topical ruxolitinib has also showed efficacy.84
Given the dependency of vitiligo progression on IFN-γ signalling, JAK inhibitors have also been investigated in this setting. Results from a multicentre, double blind, phase 2 study comparing topical ruxolitinib with a control, showed good tolerability of ruxolitinib topical cream, which was associated with substantial repigmentation of vitiligo lesions, suggesting another area in which these agents could be used.85

Inflammatory bowel disease
Disease-associated proinflammatory cytokines signal, either directly or indirectly, through the JAK–STAT pathway hence providing rationale for exploring JAK inhibitors in the inflammatory bowel disease setting. In ulcerative colitis, a breakdown of mucosal integrity exists with luminal microflora stimulating an uninhibited proinflammatory response. Innate lymphoid cells, Th9, and IL-13-producing natural killer cells all contribute to epithelial damage and chronic inflammation.86 In Crohn’s disease, the pathogenesis is also multifactorial, with polarised Th1 responses under the control of IL-12, signalling via JAK2-TYK2, and IL-27, signalling via JAK1, JAK2, and TYK2, among others.87 Genome wide association studies have shown a significant association between Crohn’s disease and the IL23R gene, high- lighting the proinflammatory role of IL-23 and the Th17 cell pathway.88
Despite the recognised efficacy of biological agents such as the TNF inhibitors, anti-integrins, and anti-IL-12 and IL-23 monoclonal antibodies, approximately more than 50% of patients with moderate-to-severe ulcerative colitis do not have sustained remissions.89,90 Efficacy of oral tofacitinib in ulcerative colitis was reported after three large, placebo-controlled phase 3 trials—two trials

that investigated tofacitinib as induction therapy (OCTAVE Induction-1 and OCTAVE Induction-2) in patients who had non-successful responses to conventional or anti-TNF agents and one trial as maintenance (Octave Sustain).91 Remission was defined by the Mayo score 2 or less and physician global assessment. For both induction studies, tofacitinib had significantly better 8-week remission rates (18·5% in the tofacitinib group vs 8·2% in the placebo group in OCTAVE Induction-1, and 16·6% in the tofacitinib group vs 3·6% in the placebo group in OCTAVE Induction-2) and remission rates were improved in both tofacitinib dosing groups of OCTAVE Sustain, as were glucocorticoid-free 52-week remission rates. Responses to tofacitinib often happened quickly and, when com- pared with placebo, there were increases in LDL and HDL concentration, which plateaued after 4 weeks, and increased infectious complications, including herpes zoster reactivation. Toxicity with tofacitinib in inflam- matory bowel disease appears to be dose dependent. Tofacitinib was associated with more cases, albeit a small number, of non-melanoma skin cancers than the placebo. An open extension study, OCTAVE Open (NCT01470612), will provide important long-term efficacy and safety data. Tofacitinib appears to be a highly active agent in ulcerative colitis and is currently being positioned in the treatment pathway after FDA approval. Guidelines from the American College of Gastro- enterology placed tofacitinib either as first-line therapy for moderate-to-severely active ulcerative colitis or as second-line therapy after TNF inhibitors.92 Outside of clinical trials, Weisshoff and colleagues93 reported real- world data of tofacitinib in 58 patients with moderate-to- severe inflammatory bowel disease and resistance to TNF inhibitors; they described meaningful clinical responses in 40 (69%) of 58 patients at 8 weeks and steroid-free clinical remission in 7 (27%) of 26 at 12 months after starting treatment.93 However, the FDA and EMA have placed warnings concerning tofacitinib 10 mg twice daily in patients with ulcerative colitis due to the risk of thromboembolic events, as discussed later.
For Crohn’s disease, two phase 2b studies investigated tofacitinib induction and maintenance in moderate-to- severe Crohn’s disease, yet in contrast to ulcerative colitis, although minor treatment effects were noted, efficacy endpoints were not significantly different from placebo.94 This result might reflect the dif- ferential immune-mediated pathogenesis compared with ulcerative colitis, JAK inhibitors specificity in Crohn’s disease, or trial design and patient inclusion; therefore, JAK inhibitors therapy in Crohn’s disease require additional exploration.
Regarding other JAK inhibitors, the FITZROY study investigated filgotinib in moderate-to-severe Crohn’s disease and showed better clinical efficacy than the placebo and acceptable safety in patients who had TNF inhibitor- treated and treatment-naive disease.95 Upadacitinib has

been investigated in phase 2 trials for both moderate-to- severe ulcerative colitis and Crohn’s disease. For moderate- to-severe ulcerative colitis, the phase 2b trial showed better remission induction efficacy than the placebo at 8 weeks.96 For moderate-to-severe Crohn’s disease (CELEST trial), upadacitinib was superior to the placebo in inducing endoscopic improvements in patients, most of whom were refractory to biologics.97 Upadacitinib efficacy is now being assessed in the phase 3 trial setting—eg, an ulcerative colitis trial, named U-Accomplish, comparing upadacitinib to placebo as assessed by the adapted Mayo score; and a Crohn’s disease trial delineating the efficacy and safety in patients with moderate-to-severe disease who are non-responsive or intolerant to biologic therapies.98 Additional JAK inhibitors undergoing assessment in the setting of inflammatory bowel disease include peficitinib, ritlecitinib, brepocitinib, and TD-1473, a novel pan-JAK inhibitor that has marked gastro- intestinal specificity.99,100 Efficacy and longer-term data are awaited, particularly as some cytokines that promote bowel mucosal integrity also rely on JAKs for signalling, and the exact use of these JAK inhibitors in the therapeutic setting requires clarification.

Overview of safety concerns
We have discussed specific side-effects of JAK inhibitors and have highlighted them in the table (with a haematology focus) and panel 2 (rheumatology focus). Frequently, side-effects of JAK inhibitors are mild-to- moderate, predictable, and easy to manage, yet dose modifications might be mandatory in some patients and patients need close monitoring. Moreover, clinicians should be vigilant for serious and sometimes life- threatening complications. Complications include rebound phenomena, which is rare and can cause a severe cytokine storm and systemic inflammatory response in patients, in particular in patients given ruxolitinib.101 On-target effects of agents such as ruxolitinib include expected cytopenias (predominantly JAK2 related) and, due to immunosuppressive proper- ties, JAK inhibitors have a heterogeneous risk of infectious complications.7,23,24,31,58,102 Depending on the agent, these infectious complications can include upper respiratory and urinary tract infections and, in general, herpes zoster reactivation, which could be considered a class effect. Atypical infections, such as Mycobacterium tuberculosis reactivation and John Cunningham virus- mediated neurological disease, have been described with ruxolitinib (reviewed by McLornan and colleagues).7 Monitoring of renal and liver function is needed and hyperlipidaemia could be considered a potential class effect of JAK inhibitors, with marked phenotypic variability, requiring lipid monitoring. As discussed, fedratinib has a black box warning for encephalopathy risk, after earlier cases of Wernicke’s encephalopathy, and momelotinib is associated with low level (usually
≤2 grade) sensory neuropathy.27,28,31,32,38

As discussed, the FDA approved the baracitinib 2 mg dose but not the 4 mg dose due to thromboembolic risk. For tofacitinib, a boxed warning was added in 2019 by the FDA for an increased risk of thromboembolism and death with the 10 mg twice daily dose in patients with rheumatoid arthritis or ulcerative colitis, which was added after increases in these events in individuals older than 50 years who had at least one cardiovascular risk factor. Direct thrombotic risk comparison between specific JAK inhibitors has been attempted but results are controversial.103 Although rare, gastrointestinal perforation has been described for a few patients with rheumatoid arthritis (some of whom had other risk factors) given either tofacitinib or baracitinib.104

Future developments and combinatorial approaches
It is clear that JAK inhibitors have revolutionised our therapeutic armamentarium across a pleiotropic range of immune-mediated and inflammatory disorders, and across clonal myeloid disorders, such as myeloproliferative neoplasm. As a class of agents, many patients are likely to benefit from JAK inhibitors in the coming years; however, increased clinical experience is required to establish the risk–benefit ratio for each agent in specific clinical indications. Vigilance in the clinical community is required concerning the common and rare side-effects of these drugs, which include atypical infectious complications, withdrawal syndrome, potential cytopenias and liver dysfunction, disturbed lipid metabolism, and risk of thrombosis with some agents (depending on underlying clinical condition), additional understanding of potential risk of B-cell lymphoma,105 coupled with a need for enhanced skin surveillance for non-melanoma skin cancers. Long-term efficacy data and postmarketing surveillance is required for all current JAK inhibitors.
Increasingly, combinatorial therapies with JAK inhibitors are being explored, aiming to maximise treatment responses after recognition that deregulated JAK–STAT signalling is not the only pathogenetic mechanism in disorders such as myelofibrosis and rheumatoid arthritis. Drugs can be given simultaneously or sequenced—eg, a trial of the histone deacetylase inhibitor, pracinostat, and ruxolitinib in patients with myelofibrosis in which patients were given 3 months of ruxolitinib and then pracrinostat. Novel combinatorial approaches in myelofibrosis with JAK inhibitors include targeting the neuro–haematopoietic axis and targeted inhibition of nuclear–cytoplasmic transport (leading to accumulation of p53).106,107 From a practical stance, the future could bring access to generic JAK inhibitors within rheumatoid arthritis and myelopro- liferative neoplasms disease areas and rapidly change the therapeutic landscape.
As the exciting field of JAK inhibitors progresses, clinicians need to understand mechanisms of resistance or loss of response to JAK inhibitors, with particular relevance to myeloproliferative neoplasm, and how best

these can be attenuated. It is well established that myeloproliferative neoplasm cells can become persistent to type I JAK inhibitors (which stabilise the active kinase conformation) and, in vitro, reversal is possible by use of a type II inhibitor (which bind to JAK2 in the inactive conformation), such as CHZ868; although, cell line research has also shown that the Leu884Pro mutation of JAK2 can confer resistance to type II inhibitors by weakening the interaction between inhibitor and target.108–110 Whether reversal of so-called persistence can be clinically achieved with other similar agents is an unknown. There are case reports of disease responsiveness in myelofibrosis being reset after withdrawal and introduction of ruxolitinib.111 Finally, concerning resis- tance, unlike BCR–ABL tyrosine kinase inhibitors, there have been no reports of acquired point mutations conferring resistance to clinically used JAK inhibitors.
It is not known whether JAK inhibitors should be used earlier in the disease process (eg, in myelofibrosis and rheumatoid arthritis) and whether JAK inhibitors should be introduced in front-line settings for conventionally low-risk or less advanced disease in an attempt to delay disease progression. Any such interventions need to be investigated in clinical trials to gain maximum knowledge and an understanding of the risk–benefit ratio.
The medical community will see these agents being increasingly used to treat autoinflammatory condi- tions—eg, JAK inhibitors are being assessed for use in rare autoinflammatory interferonopathies, such as STING-associated vasculopathy with onset in infancy syndrome and chronic atypical neutrophilic dermatoses with lipodystrophy and elevated temperature syn- drome.112 These interferonopathies possess a prominent type I IFN gene signature and, typically, do not respond to IL-1 and TNF blockade. Sanchez and colleagues113 reported on the use of baricitinib in 18 patients with chronic atypical neutrophilic dermatoses with lipodystrophy and elevated temperature syndrome, STING-associated vasculopathy with onset in infancy syndrome, Aicardi-Goutières syndrome, or undefined interferonopathy. Baracitinib treatment was associated with heterogeneous improvements in interferon and inflammatory biomarker measurements and with clinical manifestations in some patients. Long-term safety data are awaited. It is the broad anti-inflammatory actions of JAK inhibitors that have led to these agents being evaluated in the management of SARS-CoV-2 with heterogeneous results.113
Ultimately, potential disease-associated immune signatures might aid clinicians to select which patients across disease groups who will benefit most from a particular JAK inhibitor, being of best clinical use in disease areas such as rheumatoid arthritis and mye- loproliferative neoplasm, in which there are multiple licensed products and more on the horizon. Personalised stratification to appropriate JAK inhibitors therapy will

surely be an attractive goal across the entire range of disorders we have discussed.
Contributors
The first draft was written by DPM, JEP, and CNH. JG wrote sections and made additional edits to the manuscript. All authors did the literature search, interpreted data and evidence for this review article, and approved the final manuscript.
Declaration of interests
DPM reports speaker and advisory board fees from JAZZ pharmaceuticals and Novartis. JEP reports speaker and advisory board fees from Pfizer, Lily, AbbVie, and Gilead Pharmaceuticals. JG reports funding for clinical trial conduct, advisory boards, and travel reimbursement from Incyte; funding for clinical trial conduct, advisory board funding, and travel reimbursement from Celgene; funding for clinical trial conduct from CTI Biopharma; and funding for clinical trial conduct and advisory boards from Gilead. CNH reports speaker and advisory board fees from Novarti, Gilead, CTI Biopharma, and Celgene; speaker fees from JANNSEN; advisory board fees from AOP Pharma, Roche Pharmaceuticals, and Sierra Oncology.
References
⦁ Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J Cell Sci 2004; 117: 1281–83.
⦁ Haan C, Kreis S, Margue C, Behrmann I. Jaks and cytokine receptors—an intimate relationship. Biochem Pharmacol 2006; 72: 1538–46.
⦁ Darnell JE, Kerr IM, Stark GR. JAK–STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 1994; 264: 1415–21.
⦁ Stark GR, Darnell JE. The JAK–STAT pathway at twenty. Immunity
2012; 36: 503–14.
⦁ Liongue C, Sertori R, Ward AC. Evolution of cytokine receptor signaling. J Immunol 2016; 197: 11–18.
⦁ Igaz P, Tóth S, Falus A. Biological and clinical significance of the JAK–STAT pathway; lessons from knockout mice. Inflamm Res 2001; 50: 435–41.
⦁ McLornan DP, Khan AA, Harrison CN. Immunological consequences of JAK inhibition: friend or foe?
Curr Hematol Malig Rep 2015; 10: 370–79.
⦁ Richard AJ, Stephens JM. The role of JAK–STAT signaling in adipose tissue function. Biochim Biophys Acta 2014; 1842: 431–39.
⦁ Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med 2005; 352: 1779–90.
⦁ James C, Ugo V, Le Couédic J-P, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 2005; 434: 1144–48.
⦁ Baxter EJ, Scott LM, Campbell PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 2005; 365: 1054–61.
⦁ Levine RL, Wadleigh M, Cools J, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 2005; 7: 387–97.
⦁ Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 2013; 369: 2379–90.
⦁ Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, O’Shea JJ. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov 2017; 16: 843–62.
⦁ O’Shea JJ, Holland SM, Staudt LM. JAKs and STATs in immunity, immunodeficiency, and cancer. N Engl J Med 2013; 368: 161–70.
⦁ Pencik J, Pham HTT, Schmoellerl J, et al. JAK–STAT signaling in cancer: from cytokines to non-coding genome. Cytokine 2016;
87: 26–36.
⦁ Mohanty SK, Yagiz K, Pradhan D, et al. STAT3 and STAT5A are potential therapeutic targets in castration-resistant prostate cancer. Oncotarget 2017; 8: 85997–6010.
⦁ Stuart E, Buchert M, Putoczki T, et al. Therapeutic inhibition of Jak activity inhibits progression of gastrointestinal tumors in mice. Mol Cancer Ther 2014; 13: 468–74.

⦁ Ishizaki M, Akimoto T, Muromoto R, et al. Involvement of tyrosine kinase-2 in both the IL-12/Th1 and IL-23/Th17 axes in vivo.
J Immunol 2011; 187: 181–89.
⦁ Prutsch N, Gurnhofer E, Suske T, et al. Dependency on the TYK2/STAT1/MCL1 axis in anaplastic large cell lymphoma. Leukemia 2019; 33: 696–709.
⦁ Papp K, Gordon K, Thaçi D, et al. Phase 2 trial of selective tyrosine kinase 2 inhibition in psoriasis. N Engl J Med 2018; 379: 1313–21.
⦁ Batista CR, Lim M, Laramée A-S, et al. Driver mutations in Janus kinases in a mouse model of B-cell leukemia induced by deletion of PU.1 and Spi-B. Blood Adv 2018; 2: 2798–810.
⦁ Harrison C, Kiladjian J-J, Al-Ali HK, et al. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis.
N Engl J Med 2012; 366: 787–98.
⦁ Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo- controlled trial of ruxolitinib for myelofibrosis. N Engl J Med 2012; 366: 799–807.
⦁ Al-Ali HK, Griesshammer M, Foltz L, et al. Primary analysis of JUMP, a phase 3b, expanded-access study evaluating the safety and efficacy of ruxolitinib in patients with myelofibrosis, including those with low platelet counts. Br J Haematol 2020; 189: 888–903.
⦁ Mead AJ, Milojkovic D, Knapper S, et al. Response to ruxolitinib in patients with intermediate-1-, intermediate-2-, and high-risk myelofibrosis: results of the UK ROBUST trial. Br J Haematol 2015; 170: 29–39.
⦁ Mesa RA, Kiladjian J-J, Catalano JV, et al. SIMPLIFY-1: a phase III randomized trial of momelotinib versus ruxolitinib in janus kinase inhibitor-naïve patients with myelofibrosis. J Clin Oncol 2017;
35: 3844–50.
⦁ Harrison CN, Vannucchi AM, Platzbecker U, et al. Momelotinib versus best available therapy in patients with myelofibrosis previously treated with ruxolitinib (SIMPLIFY 2): a randomised, open-label, phase 3 trial. Lancet Haematol 2018; 5: e73–81.
⦁ Mesa RA, Vannucchi AM, Mead A, et al. Pacritinib versus best available therapy for the treatment of myelofibrosis irrespective of baseline cytopenias (PERSIST-1): an international, randomised, phase 3 trial. Lancet Haematol 2017; 4: e225–36.
⦁ Mascarenhas J, Hoffman R, Talpaz M, et al. Pacritinib vs best available therapy, including ruxolitinib, in patients with myelofibrosis: a randomized clinical trial. JAMA Oncol 2018; 4: 652–59.
⦁ Pardanani A, Harrison C, Cortes JE, et al. Safety and efficacy of fedratinib in patients with primary or secondary myelofibrosis: a randomized clinical trial. JAMA Oncol 2015; 1: 643–51.
⦁ Harrison CN, Schaap N, Vannucchi AM, et al. Fedratinib in patients with myelofibrosis previously treated with ruxolitinib:
an updated analysis of the JAKARTA2 study using stringent criteria for ruxolitinib failure. Am J Hematol 2020; 95: 594–60.
⦁ Vannucchi AM, Kiladjian JJ, Griesshammer M, et al. Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N Engl J Med 2015; 372: 426–35.
⦁ Passamonti F, Griesshammer M, Palandri F, et al. Ruxolitinib for the treatment of inadequately controlled polycythaemia vera without splenomegaly (RESPONSE-2): a randomised, open-label, phase 3b study. Lancet Oncol 2017; 18: 88–99.
⦁ Moodley D, Yoshida H, Mostafavi S, et al. Network pharmacology of JAK inhibitors. Proc Natl Acad Sci USA 2016; 113: 9852–57.
⦁ Xu H, Jesson MI, Seneviratne UI, et al. PF-06651600, a dual JAK3/TEC family kinase inhibitor. ACS Chem Biol 2019; 14: 1235–42.
⦁ Harrison CN, Vannucchi AM, Kiladjian J-J, et al. Long-term findings from COMFORT-II, a phase 3 study of ruxolitinib vs best available therapy for myelofibrosis. Leukemia 2017; 31: 775.
⦁ Celgene Corporation. Celgene Corporation (NASDAQ: CELG) today announced the U.S. Food and Drug Administration (FDA) has approved INREBIC® (fedratinib) for the treatment of adult patients with intermediate-2 or high-risk primary or secondary
(post-polycythemia vera or post-essential thrombocythemia) myelofibrosis. Aug 16, 2019. https://ir.celgene.com/press-releases- archive/press-release-details/2019/US-FDA-Approves-INREBIC- Fedratinib-as-First-New-Treatment-in-Nearly-a-Decade-for-Patients- With-Myelofibrosis/default.aspx (accessed July 7, 2020).
⦁ Harrison CN, McLornan DP. Current treatment algorithm for the management of patients with myelofibrosis, JAK inhibitors, and beyond. Hematology 2017; 2017: 489–97.

⦁ Gerds AT, Savona M, Scott BL, et al. Results of PAC203:
a randomized phase 2 dose-finding study and determination of the recommended dose of pacritinib. Blood 2019;
134 (suppl 1): 667.
⦁ Blazar BR, Murphy WJ, Abedi M. Advances in graft-versus-host disease biology and therapy. Nat Rev Immunol 2012; 12: 443–58.
⦁ Spoerl S, Mathew NR, Bscheider M, et al. Activity of therapeutic JAK 1/2 blockade in graft-versus-host disease. Blood 2014;
123: 3832–42.
⦁ Zeiser R, Burchert A, Lengerke C, et al. Ruxolitinib in corticosteroid-refractory graft-versus-host disease after allogeneic stem cell transplantation: a multicenter survey. Leukemia 2015; 29: 2062–68.
⦁ US Food and Drug Administration. FDA approves ruxolitinib for acute graft-versus-host disease. May 24, 2019. ⦁ https://www.fda.gov/ drugs/resources-information-approved-drugs/fda-approves- ruxolitinib-acute-graft-versus-host-disease (accessed July 7, 2020).
⦁ Jagasia M, Zeiser R, Arbushites M, Delaite P, Gadbaw B, Bubnoff N. Ruxolitinib for the treatment of patients with steroid- refractory GVHD: an introduction to the REACH trials. Immunotherapy 2018; 10: 391–402.
⦁ Zeiser R, von Bubnoff N, Butler J, et al. Ruxolitinib for glucocorticoid-refractory acute graft-versus-host disease. N Engl J Med 2020; 382: 1800–10.
⦁ Virtanen AT, Haikarainen T, Raivola J, Silvennoinen O. Selective JAKinibs: prospects in inflammatory and autoimmune diseases. BioDrugs 2019; 33: 15–32.
⦁ Jegatheeswaran J, Turk M, Pope JE. Comparison of Janus kinase inhibitors in the treatment of rheumatoid arthritis: a systemic literature review. Immunotherapy 2019; 11: 737–54.
⦁ AbbVie. AbbVie receives FDA approval of RINVOQ™ (upadacitinib), an oral JAK inhibitor for the treatment of moderate to severe rheumatoid arthritis. Aug 16, 2019. https://news.abbvie. com/news/press-releases/abbvie-receives-fda-approval-rinvoq- upadacitinib-an-oral-jak-inhibitor-for-treatment-moderate-to-severe- rheumatoid-arthritis.htm (accessed July 7, 2020).
⦁ Westhovens R. Clinical efficacy of new JAK inhibitors under development. Just more of the same? Rheumatology 2019; 58: i27–33.
⦁ Felson DT, Anderson JJ, Boers M, et al. American College of Rheumatology. Preliminary definition of improvement in rheumatoid arthritis. Arthritis Rheum 1995; 38: 727–35.
⦁ Fransen J, Creemers MCW, Van Riel PLCM. Remission in rheumatoid arthritis: agreement of the disease activity score (DAS28) with the ARA preliminary remission criteria. Rheumatology (Oxford) 2004; 43: 1252–55.
⦁ Westhovens R, Taylor PC, Alten R, et al. Filgotinib (GLPG0634/GS-6034), an oral JAK1 selective inhibitor, is effective in combination with methotrexate (MTX) in patients with active
rheumatoid arthritis and insufficient response to MTX: results from a randomised, dose-finding study (DARWIN 1). Ann Rheum Dis 2017; 76: 998–1008.
⦁ Kavanaugh A, Kremer J, Ponce L, et al. Filgotinib
(GLPG0634/GS-6034), an oral selective JAK1 inhibitor, is effective as monotherapy in patients with active rheumatoid arthritis: results from a randomised, dose-finding study (DARWIN 2).
Ann Rheum Dis 2017; 76: 1009–19.
⦁ Kremer JM, Emery P, Camp HS, et al. A phase IIb study of
ABT-494, a selective JAK-1 inhibitor, in patients with rheumatoid arthritis and an inadequate response to anti-tumor necrosis factor therapy: ABT-494 in patients with ra and inadequate response to anti-TNF therapy. Arthritis Rheumatol 2016; 68: 2867–77.
⦁ 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 patients with rheumatoid arthritis and an inadequate response to methotrexate. Arthritis Rheumatol 2016; 68: 2857–66.
⦁ Dore RK, Antonova J, Burudpakdee C, Wang X, Ozbay B, Genovese MC. Treatment patterns, persistence, and durability in rheumatoid arthritis patients with inadequate response to biologic disease-modifying anti-rheumatic drugs (BDMARDS).
Ann Rheum Dis 2019; 78 (suppl 2): 1135 (abstr SAT0133).
⦁ Wollenhaupt J, Lee E-B, Curtis JR, et al. Safety and efficacy of tofacitinib for up to 9·5 years in the treatment of rheumatoid arthritis: final results of a global, open-label, long-term extension study. Arthritis Res Ther 2019; 21: 89.

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

⦁ Sandborn WJ, Ghosh S, Panes J, et al. Efficacy of upadacitinib in a randomized trial of patients with active ulcerative colitis. Gastroenterology 2020; 158: 2139–49.e14.
⦁ Sandborn WJ, Feagan BG, Loftus EV, et al. Efficacy and safety of upadacitinib in a randomized trial of patients with Crohn’s disease. Gastroenterology 2020; 158: 2123–38.e8.
⦁ Sandborn WJ, Schreiber S, Lee SD, et al. Improved endoscopic outcomes and mucosal healing of upadacitinib as an induction therapy in adults with moderately to severely active ulcerative colitis: data from the U-ACHIEVE study. J Crohns Colitis 2019; 13 (suppl 1): S009
⦁ D’Amico F, Fiorino G, Furfaro F, Allocca M, Danese S. Janus kinase inhibitors for the treatment of inflammatory bowel diseases: developments from phase I and phase II clinical trials.
Expert Opin Investig Drugs 2018; 27: 595–99.
⦁ Ma C, Lee JK, Mitra AR, et al. Systematic review with meta-analysis: efficacy and safety of oral Janus kinase inhibitors for inflammatory bowel disease. Aliment Pharmacol Ther 2019; 50: 5–23.
⦁ Palandri F, Palumbo GA, Elli EM, et al. Ruxolitinib discontinuation syndrome: incidence, risk factors, and management in 251 patients with myelofibrosis. Blood Cancer J 2021; 11: 4.
⦁ Jegatheeswaran J, Turk M, Pope JE. Comparison of Janus kinase inhibitors in the treatment of rheumatoid arthritis: a systemic literature review. Immunotherapy 2019; 11: 737–54.
⦁ Scott IC, Hider SL, Scott DL. Thromboembolism with Janus kinase (JAK) inhibitors for rheumatoid arthritis: how real is the risk?
Drug Saf 2018; 41: 645–53.
⦁ Xie F, Yun H, Bernatsky S, Curtis JR. Brief report: risk of gastrointestinal perforation among rheumatoid arthritis patients receiving tofacitinib, tocilizumab, or other biologic treatments. Arthritis Rheumatol 2016; 68: 2612–17.

⦁ Rumi E. Zibellini S, Boveri E, et al. Ruxolitinib treatment and risk of B-cell lymphomas in myeloproliferative neoplasms.
Am J Hematol 2019; 94: e185–88.
⦁ Herlihy N, Harrison CN, McLornan DP. Exploitation of the neural- hematopoietic stem cell niche axis to treat myeloproliferative neoplasms. Haematologica 2019; 104: 639–41.
⦁ Yan D, Pomicter AD, Tantravahi S, et al. Nuclear-cytoplasmic transport is a therapeutic target in myelofibrosis. Clin Cancer Res 2019; 25: 2323–35.
⦁ Koppikar P, Bhagwat N, Kilpivaara O, et al. Heterodimeric JAK–STAT activation as a mechanism of persistence to JAK2 inhibitor therapy. Nature 2012; 489: 155–59.
⦁ Kong X, Sun H, Pan P, et al. How does the L884P mutation confer resistance to type-II inhibitors of JAK2 kinase: a comprehensive molecular modeling study. Sci Rep 2017; 7: 9088.
⦁ Meyer SC, Keller MD, Chiu S, et al. CHZ868, a type II JAK2 inhibitor, reverses type I JAK inhibitor persistence and demonstrates efficacy in myeloproliferative neoplasms. Cancer Cell 2015; 28: 15–28.
⦁ Gisslinger H, Schalling M, Gisslinger B, Skrabs C, Müllauer L, Kralovics R. Restoration of response to ruxolitinib upon brief withdrawal in two patients with myelofibrosis. Am J Hematol 2014; 89: 344–46.
⦁ Sanchez GAM, Reinhardt A, Ramsey S, et al. JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies. J Clin Invest 2018; 128: 3041–52.
⦁ D’Alessio A, Del Poggio P, Bracchi F, et al. Low-dose ruxolitinib plus steroid in severe SARS-CoV-2 pneumonia. Leukemia 2020; 35: 635–38.TG101348

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>