Abstract
Asthma is an inflammatory disease of the airways characterized by intermittent episodes of wheezing, chest tightness, and cough. Many of the inflammatory pathways implicated in asthma involve cytokines and growth factors that activate Janus kinases (JAKs). The discovery of the JAK/signal transducer and activator of transcription (STAT) signaling pathway was a major breakthrough that revolutionized our understanding of cell growth and differentiation. JAK inhibitors are under active investigation for immune and inflammatory diseases, and they have demonstrated clinical efficacy in diseases such as rheumatoid arthritis and atopic dermatitis. Substantial preclinical data support the idea that inhibiting JAKs will ameliorate airway inflammation and hyperreactivity in asthma. Here, we review the rationale for use of JAK inhibitors in different asthma endotypes as well as the preclinical and early clinical evidence supporting such use. We review preclinical data from the use of systemic and inhaled JAK inhibitors in animal models of asthma and safety data based on the use of JAK inhibitors in other diseases. We conclude that JAK inhibitors have the potential to usher in a new era of anti-inflammatory treatment for asthma.
Keywords: Asthma, severe asthma, Janus kinase, JAK/STAT pathway, JAK Inhibitor
RATIONALE FOR JAK INHIBITORS IN ASTHMA
Asthma is an inflammatory disease of the airways characterized by intermittent episodes of wheezing, chest tightness, and cough. Asthma affects approximately 300 million people world-wide and more than 25 million people in the United States. Severe asthma affects 5% to 15% of the population with adult asthma and is associated with increased mortality, increased hospitalizations, significant burden of symptoms, health care costs, and missed work and school. In the United States alone, more than $80 billion in direct and indirect costs are spent on asthma annually, with most of that amount spent on patients with severe asthma. Severe asthma represents a subset of difficult-to-treat asthma and occurs in patients whose disease remains uncontrolled despite the use of high doses of inhaled corticosteroids (ICSs) combined with long-acting β-agonists or other controllers.1 Even with the approval of several targeted biologics in the past few years, many patients continue to experience exacerbations or uncontrolled disease, indicating a need for more novel therapies.
Over the past several decades, it has become clear that asthma is a heterogeneous disease in terms of its clinical presentation, physiologic characteristics, and responses to therapy. Much of this variability is due to differences in gene-by-environment interactions, resulting in complex biochemical pathways underlying the disease. As we have gained a better understanding of pathophysiologic mechanisms of asthma, we have shifted from a phenotypic classification system focused on specific triggers and clinical presentation to a classification system focused on endotypes, particular pathophysiologic mechanisms, and related clinical biomarkers associated with a given patient.2 There are 2 major endotypes in asthma: type 2 and non–type 2. The type 2 pathway is defined by activation of cytokines derived from TH2 cells and group 2 innate lymphoid cells (ILC2s); these include IL-4, IL-5, and IL-13 that cause airway inflammation by activating eosinophils, B cells, airway epithelial cells, and other cell types. Biomarkers of type 2 asthma include blood/sputum eosinophilia and elevated levels of fractional exhaled nitric oxide (Feno) and IgE. The type 2–low pathway is characterized by absence of type 2–high cytokines and biomarkers, and it manifests either increased levels of neutrophils in the airways or a paucigranulocytic profile, with normal levels of airway neutrophils and eosinophils.3 Type 2–low asthma is currently not well understood, and it likely encompasses multiple distinct endotypes.
Most of the inflammatory pathways implicated in different asthma endotypes involve cytokines and growth factors that signal via receptors coupled to Janus kinases (JAKs). The JAK family comprises the following 4 members of intracellular tyrosine kinases: JAK1, JAK2, JAK3, and Tyk2. JAK family members phosphorylate and activate intracellular signal transducer and activator of transcription (STAT) proteins, which regulate the transcription of their downstream target genes. The discovery of the JAK/STAT signaling pathway was a major breakthrough that revolutionized our understanding of cell growth and differentiation.4 Experiments using cell culture and genetically modified mouse models revealed an essential role for JAK/STAT family members in immune-mediated diseases, including asthma.
Cytokines and growth factors implicated in asthma pathophysiology that signal via JAK-coupled receptors include IL-4, IL-5, IL-6, IL-9, IL-13, GM-CSF, IFN-α, IFN-β, INF-γ, and thymic stromal lymphopoietin (TSLP). Notable exceptions to this list include TNF, IL-1, IL-17 members, and IL-33.5,6 Different JAK family members couple to cytokine receptors in a combinatorial manner. However, it is important to note that not all JAK family members participate equally in signal transduction, despite association with a given receptor. For example, IL-13–induced phosphorylation of STAT6 was preferentially inhibited by compounds targeting JAK1 (but not JAK2 or Tyk2) in the BEAS-2B airway epithelial cell line.7 Similarly, IL-6 recruits and activates JAK1, JAK2, and Tyk2, but IL-6–dependent STAT3 phosphorylation was preferentially inhibited by JAK1 (but not Tyk2) inhibitors in human cell lines.8
Small molecule JAK inhibitors are under active development and have been approved to treat rheumatoid arthritis (RA) and other conditions.9,10 In this review, we summarize the rationale for targeting JAK-dependent signaling pathways in patients with type 2–high and type 2–low asthma, review available literature reporting preclinical studies in asthma models of asthma, and review a recently published human clinical trial that demonstrated proof of concept of target engagement in asthma.11 We conclude by considering the safety profile of JAK inhibitors and speculate on the positioning of these therapies in the context of other asthma treatments.
JAK INHIBITOR SPECIFICITY AND ROUTE OF DELIVERY
Rapid progress is being made in the development of orally available small molecule JAK inhibitors.9,12,13 There are currently 5 US Food and Drug Administration (FDA)-approved compounds for clinical use, 2 for use in hematologic disorders (ruxolitinib and fedratinib) and 3 for use in inflammatory diseases (tofacitinib, baricitinib, and upadicitinib [Table I]), and many others are in phase II/III studies for a variety of conditions, including RA, ulcerative colitis, psoriasis, and atopic dermatitis. Ruxolitinib and tofacitinib are considered pan-JAK inhibitors (inhibiting JAK1, JAK 2, and JAK3), although tofacitinib has lower affinity for JAK2 than ruxolitinib does. All of the currently approved JAK inhibitors cause dose-dependent myelosuppression, which is usually mild and reversible and thought to be due to inhibition of JAK2-dependent hematopoietic growth factors.14 For treatment of inflammatory diseases, there has been recent emphasis on developing targeted antagonists that spare JAK2-dependent growth factors in order to avoid bone marrow suppression. However, there is significant overlap in the chemical structure of JAK isoforms, and although inhibitors with selectivity for certain JAK family members have been developed, it is not clear whether narrow specificity is desirable for a complex inflammatory disease such as asthma. Substrate specificity for a given JAK inhibitor can be determined by in vitro biochemical assays (ie, the inhibitory constant [Ki] for ATP-competitive inhibition with use of purified substrates) or cell-based assays (eg, the inhibitory concentration of 50% [IC50] for cytokine-induced STAT phosphorylation in reference cell lines). Although these approaches usually result in comparable and reproducible rank order potencies under controlled conditions,8 it is clear that specificity for a given JAK family member can also vary depending on cell type and cytokine(s) studied.15,16 Therefore, the idea of JAK isoform specificity should be considered a relative concept. It is clear that we have more to learn about how these compounds work in human subjects, and clinicians and investigators need to remain vigilant for unexpected and off-target effects in human clinical trials.
TABLE I.
Compound | Indication |
---|---|
Tofacitinib | RA, PsA, UC |
Ruxolitinib | PCV, MF |
Baricitinib | RA |
Upadacitinib | RA |
Fedratinib | MF |
MF, Myelofibrosis; PCV, polycythemia vera; PsA, psoriatic arthritis; UC, ulcerative colitis.
Particularly exciting for asthma is the development of inhaled JAK inhibitors, with chemical structures that facilitate lung retention and minimize systemic exposure.16–20 This is an active area of research that was recently reviewed comprehensively.21 Local deposition and retention of inhaled JAK inhibitors might obviate some of the concerns raised with systemic administration, such as JAK2-dependent myelosuppression. We review some of the preclinical studies with these novel compounds in asthma in Table II16,18,19,22–27 and in the section on decreased lymphocyte and neutrophil counts later in this article.
TABLE II.
Compound | JAK specificity | Route | Species | Allergen/model | Findings | Reference |
---|---|---|---|---|---|---|
Tofacitinib | Pan-JAK | Subcutaneous via osmotic minipumps (1.5–15 mg/kg/d) | Mouse | OVA i.p. + alum, then OVA aerosol | Reduced BAL eosinophilia, eotaxin, and IL-13 levels when administered during both sensitization and challenge or during the challenge phase alone | 22 |
P6 | Pan-JAK | i.p.* | Mouse | OVA i.p. + alum, then OVA aerosol | Decreased airway resistance, BAL eosinophilia, and Muc5AC, but only if PLGA-encapsulated and administered during challenge No effect if administered during sensitization and challenge |
23 |
R256† | JAK1/3 | Oral gavage (10–50 mg/kg) | Mouse | OVA i.p. + alum then OVA aerosol | Decreased airway resistance, BAL eosinophilia, mucus production Decreased TH2 cytokines only if administered during sensitization |
24 |
Ruxolitnib | Pan-JAK | Intragastric (180 mg/kg)‡ | Mouse | OVA + low-dose LPS inhaled then OVA aerosol | Reduced BAL eosinophil, neutrophil, and IL-17 levels | 25 |
iJak-381 | JAK1 | i.n., dry powder inhaler | Mouse Guinea pig | OVA i.p. + alum then OVA aerosol Aspergillus, Alternaria, and house dust mite (AAH) | Reduced BAL eosinophilia, CCL11, airway resistance, and Muc5AC in OVA-challenged mice Reduced BAL eosinophilia, neutrophilia, CCL11, and CXCL1 in in AAH-challenged mice Reduced lung inflammation in OVA-challenged guinea pigs |
18 |
LAS194046 | Pan-JAK§ | i.t. (0.3–1 mg/kg) | Rat | OVA i.p. + alum then OVA aerosol | Reduced BAL eosinophilia and neutrophila, and AHR (Penh) | 16 |
iJAK-001 | Not stated | i.t. (0.03–1 mg/kg) or dry powder inhaler (10 mg) | Rat Sheep | Alternaria extract i.t. Ascaris suum | Reduced Alternaria-induced BAL eosinophilia in mice Reduced Ascaris-induced airway resistance in sheep |
19 |
Tofacitinib | Aerosol (1–2 mg/kg) | Mouse | House dust mite extract | Reduced eosinophil and total protein levels | 26 | |
Tofacitinib | i.p. (5 mg/kg) | Mouse | OVA i.p. + alum then OVA i.n. | Reduced numbers of tissue eosinophils, polypoid-like lesions, TH2 cytokines, and phospho- STAT6 | 27 |
AAH, Aspergillus, Alternaria, and house dust mite; i.n., intranasal; i.p., intraperitoneal; i.t., intratracheal, PLGA, poly d,l-lactic-co-glycolic acid.
Alone (3 mg/kg) or in PLGA-encapsulated nanoparticles (60 mg/kg).
This compound is apparently now known as AZD0449.
Timing of ruxolitinib administration is not clear.
This compound was also found to inhibit Flt3 and TrkA.
JAK-STAT AND TYPE 2–HIGH ASTHMA
The JAK/STAT pathway has a critical role in the development and signaling of TH2 cytokine–driven immune responses. IL-4 and IL-13 activate STAT6 in their target cells in a JAK-dependent manner, and experiments using gene-targeted knockout mice confirmed a key role for STAT6 in IgE production, bronchial hyperresponsiveness, airway remodeling, and mucus metaplasia after allergen sensitization.28,29 Other type 2 asthma–associated cytokines also signal through JAK/STAT proteins, including IL-5 and TSLP. Because each of these key type 2 asthma cytokines (IL4, IL5, IL13, and TSLP) signal via JAK-dependent pathways, there is a strong potential role for JAK inhibitors in the treatment of type 2 asthma.
Development and maintenance of type 2 immune responses in asthma
JAK signaling pathways are critically important for the differentiation of CD4+ TH2 cells from naive precursors. Key cytokines implicated in this process include IL-2 and IL-4, which activate the transcription factors STAT5 and STAT6, respectively, via cytokine receptors coupled to JAK1 and JAK3.30 However, the acute production of cytokines from already-differentiated TH2 cells occurs in a JAK-independent manner via signals from the T-cell receptor (TCR) and costimulatory molecules. Proof of principle that inhibiting JAK1/3 attenuates TH2 cell differentiation was reported by Ashino et al, who used a compound called R526.24 Interestingly, R526 had no effect on the differentiation of TH1 or TH17 cells, possibly reflecting the requirement for Tyk2-dependent signals downstream of IL-12 and/or IL-23 receptors in these cells. As expected, R526 did not inhibit TH2 cytokine production from already-differentiated T cells restimulated via the TCR. This compound also blocked eosinophilic airway inflammation in a mouse model of asthma (see later and Table II). A phase I clinical trial of R526, now known as AZD0449, was recently completed, with (it is hoped) results to be reported soon (ClinicalTrials.gov identifier NCT03766399).
Although less is currently known about the molecular regulation of TH2 cell gene expression in ILC2s, a recent study demonstrated that JAK inhibitors can block cytokine production from these cells. Human lung ILC2s were obtained by bronchoalveolar lavage (BAL) and cultured in the presence of the pan-JAK inhibitor tofacitinib. Interestingly, the production of IL-5 by ILC2s was resistant to the effects of dexamethasone, whereas tofacitinib (alone and in combination with dexamethasone) suppressed IL-5 production by ILC2s, probably by attenuating the effects of TSLP present in BAL fluids.31 This and other studies suggest the possibility that JAK inhibitors may be particularly effective in subjects with steroid-resistant TH2 cell–high asthma, especially if characterized by expansion of pathogenic ILC2s.32,33
In addition to blocking the differentiation or activation of TH2 cells and ILC2s, JAK-dependent signals may be important for the homeostasis of TH2 cells and ILC2s. In support of this possibility, IL-2 (which also signals via JAK1 and JAK3) was recently shown to play a critical role in the maintenance of allergen-specific tissue-resident memory (Trm) TH2 cells in the lung in a mouse model of asthma.34 JAK inhibitors might therefore decrease the persistence of these pathogenic T cells (which contribute to mucus metaplasia and airway hyperresponsiveness (AHR)] in the lung.35 However, this potential beneficial effect of JAK inhibitors in asthma would need to be balanced with the inhibition of protective tissue-resident memory T cells that reside in the lung following respiratory viral infections.36 It is worth noting that IL-2 is a pleiotropic cytokine and regulates the differentiation and survival of regulatory T (Treg) cells in a JAK/STAT-dependent manner.37 Although this raises the possibility that JAK inhibitors might interfere with the function of Treg cells, tofacitinib was found to spare Treg cell function in kidney transplant recipients.38 In summary, JAK inhibitors may be useful in subjects with TH2 cell–high asthma by inhibiting the development, activation, or homeostasis of CD4+ TH2 cells, as well as ILC2s. Potentially of greater importance will be the ability of JAK inhibitors to block the downstream signals of TH2 cytokines on their target cells.
Type 2 cytokine signal transduction as a therapeutic target in asthma
IL-4 binds IL-4Rα and γc, which couple to JAK1 and JAK3, whereas IL-4 and IL-13 bind IL-4Rα and IL-13Rα1, which couple to JAK1, JAK2, and Tyk2. Support for the idea that blocking IL-4Rα signal transduction is an effective therapeutic strategy in type 2–high asthma comes from the clinical experience with the IL-4Rα antagonist dupilumab.39 IL-4 and IL-13 have pleiotropic effects on virtually every cell type relevant to asthma pathophysiology; particularly important targets in the lung include airway epithelial cells and ASM cells.40 In airway epithelial cells, IL-4 and IL-13 induce a characteristic pattern of gene expression in a STAT6-dependent manner, resulting in the production of eosinophil-attracting chemokines, mucus metaplasia, and inducible nitric oxide synthase (iNOS).28,29,41,42 Guided by structure-activity relationships, Zak et al developed a family of new specific JAK1 inhibitors that blocked IL-13–induced STAT6 phosphorylation in the BEAS-2B airway epithelial cell line.7 TSLP-induced eotaxin-1 (CCL11) was also inhibited by the JAK1/2 inhibitor CYT387 in human nasal epithelial cells (HNECs) from subjects with chronic rhinosinusitis with nasal polyps.43 In another study, tofacitinib blocked CCL5 production (also known as RANTES) in BEAS2B cells induced by the double-stranded RNA polyI:C, and it did so more potently than fluticasone did.44 Thus, JAK inhibitors can block the production of epithelial chemokines relevant to asthma in response to a variety of stimuli. Taken together, these exciting results suggest that JAK inhibitors will inhibit cytokine-driven epithelial activation in asthma. STAT6 does not appear to be a major target of glucocorticoids in the airway,45 which raises the possibility that JAK inhibitors will also be effective in subjects with type 2–high and steroid-unresponsive disease.46 Because IL-4 and IL-13 also induce production of iNOS in airway epithelial cells, Feno can be used as a pharmacodynamic biomarker of target engagement. Interestingly, Braithwaite et al recently conducted a phase I clinical trial of the JAK inhibitor GDC-0214; they showed proof of concept that Feno levels are indeed reduced by this compound11 (see later).
In addition to inducing epithelial chemokine production, mucus metaplasia, and iNOS expression, TH2 cytokines also cause epithelial barrier dysfunction. Barrier dysfunction in this context refers to dysfunction of tight junction complexes that normally help cells form a tight monolayer, and it is increasingly recognized as a feature of severe asthma and other diseases associated with chronic mucosal inflammation.47,48 In 16HBE epithelial cells, IL-4– and IL-13–induced barrier dysfunction was attenuated by a pan-JAK inhibitor.49 Support for the idea that inhibiting JAK signaling can potentiate barrier function in vivo comes from a study by Amano et al, who reported that the JAK inhibitor JTE-052 improved skin barrier function in a mouse model.50 It will be interesting in future studies to determine whether JAK inhibitors facilitate the restoration of barrier function in asthma, which might help break the “vicious cycle of leak.”47
ASM cells are another potential target of IL-4 and IL-13 in the airway.51–54 Manson et al recently reported that IL-4 and IL-13 induced AHR in a dexamethasone-insensitive manner in isolated human small airways.55 This was associated with increased expression of the histamine H1 receptor, but interestingly, it was not blocked by a STAT6 inhibitor.55 Future studies will be needed to define the role of specific JAK family members in type 2 cytokine–driven ASM responses, and it remains to be seen whether JAK inhibitors might have direct effects on ASM contractility and AHR, independent of their anti-inflammatory effects.
IL-5 plays a critical role in eosinophil biology, and it promotes the growth and survival of eosinophils via the IL-5α and IL-5Rβc (CSF2RB) receptor complex, which signals via JAK2. The IL-5 pathway–targeted biologics mepolizumab, reslizumab, and benralizumab are now widely used to treat severe and exacerbation-prone asthma.56 Therefore, it stands to reason that JAK2 antagonists might have antieosinophilic effects that are potentially of clinical utility in asthma. However, JAK2 is also involved in signal transduction downstream of erythropoietin (EPO), thrombopoietin (TPO), and myeloid growth factors (ie, GM-CSF and G-CSF), and bone marrow suppression is a recognized side effect of JAK2 inhibition.14 Inhaled JAK2 inhibitors that are not active systemically might be able to suppress cytokine-dependent eosinophil activation within the lung without suppressing hematopoiesis. It is important to note that the JAK inhibitors tofacitinib, ruxolitinib, and baricitinib have all demonstrated efficacy in the treatment of idiopathic hypereosinophilic syndrome, eosinophilic esophagitis, and eosinophilic fasciitis.57–59 Efficacy in hypereosinophilic syndrome and related conditions could be due to inhibition of the JAK-dependent production of cytokines that drive pathologic eosinophilia, as well as JAK-coupled cytokine receptors on eosinophils. In future studies of JAK inhibitors in asthma, it will be important to investigate whether inhibiting airway eosinophils is associated with, or necessary for, clinical efficacy.
JAK-STAT AND TYPE 2–LOW ASTHMA
Although the immune and inflammatory mechanisms underlying type 2–low asthma are not currently well understood,1–3 it seems likely that JAK inhibitors may also be useful in some subjects with type 2–low asthma. Here we review and speculate on the potential role for JAK inhibitors in endotypes potentially associated with type 2–low asthma.
IL-6
Circulating levels of IL-6 are increased in some subjects with type 2–low asthma in association with metabolic disturbances and systemic inflammation.60–62 A gene signature of IL-6–driven epithelial activation is apparent in many individuals with severe asthma independent of type 2 inflammation.63 An IL-6 antagonist is currently under investigation in individuals with severe asthma and elevated plasma IL-6 levels in the National Heart, Lung and Blood Institute–sponsored PrecISE Asthma Network64 (ClinicalTrials.gov identifier NCT04129931). IL-6 receptor signaling activates JAK1, JAK2, and Tyk2, but IL-6–dependent STAT3 phosphorylation was preferentially inhibited by JAK1 inhibitors in studies using reference human cell lines.8 Therefore, JAK1 inhibitors might be beneficial in subjects with IL-6–high asthma. Measuring IL-6–dependent biomarkers such as C-reactive protein (CRP) could provide a biomarker of target engagement in these subjects.
IFN-γ
Increased frequency of IFN-γ–expressing lymphocytes has been observed in subjects with severe asthma, and IFN-γ signaling was required for AHR in a mouse model of severe asthma.65–67 Taken together, these observations suggest that blocking JAK1 or JAK2 activation downstream of IFN-γ receptors may be useful in subjects with this asthma endotype. At present, whether these subjects can be identified with an easily measured biomarker is not clear, but reduced expression of secretory leukocyte peptidase inhibitor (SLPI) in sputum or lung lavage samples might be useful in this regard.
IL-17
In mouse models, TH17 cells are important for airway neutrophilia and AHR, which can be inhibited by neutralizing IL-17A.68 However, the IL-17 receptor antagonist brodalumab did not meet the clinical end points in a controlled trial in individuals with moderate-to-severe asthma,69 and whether blocking IL-17 family members in subsets of individuals with asthma is clinically useful remains to be seen. Although IL-17A signals independently of JAK family members, IL-17A–secreting TH17 cells develop in response to JAK-coupled cytokines.8,70 In mouse models, there is heterogeneity among TH17 cells, with some subsets having potentially protective effects in promoting tissue integrity, whereas pathologic TH17 cell subsets cause tissue inflammation and injury.71–73 Because pathologic TH17 cell subsets are dependent on IL-23, which preferentially signals via Tyk2,8 Tyk2 antagonists might be effective in diseases mediated by pathologic TH17 cells.
Mast cells
Mast cell activation characterizes many subjects with severe asthma, and the tyrosine kinase inhibitor imatinib decreased AHR, mast cell counts, and tryptase release in a pilot study of individuals with severe and refractory asthma,74 probably by targeting the receptor tyrosine kinase c-Kit, which activates JAK2 and STAT1.75 STAT5 is critical for mast cell growth and survival,76 and it was recently shown to be involved in TSLP-induced skin mast cell degranulation.77 The role of JAK family members in mast cell biology is under active investigation. In human mast cell lines, ruxolitinib blocked degranulation and cytokine production.78 These observations raise the possibility that JAK inhibitors might block mast cell activation in asthma, which can be monitored by measuring serum and lung tryptase levels and other readouts.
PRECLINICAL STUDIES IN ANIMAL MODELS
Several preclinical studies have demonstrated efficacy of systemic or topical JAK inhibitors in animal models of asthma (Table II).16,18,19,22–27,79 In an early study, Kudlacz et al administered tofacitinib (known then as CP-690550) via subcutaneous osmotic minipumps (1.5–15 mg/kg per day) to mice during either sensitization with ovalbumin (OVA) plus alum intraperitoneally, or only during challenge with an OVA aerosol. Tofacitinib inhibited BAL levels of eosinophils, eotaxin, and IL-13 in a dose-dependent manner when administered during sensitization and challenge, as well as during challenge alone.22 Matsunaga et al studied the effect of the pan-JAK inhibitor P6 on airway inflammation and AHR in OVA-challenged mice.23 P6 inhibited AHR and some aspects of airway inflammation, especially if administered by intraperitoneal injection encapsulated with poly d,l-lactic-co-glycolic acid (PLGA) during the challenge phase with OVA aerosols. Surprisingly, P6 was not effective when administered during both sensitization and challenge with aerosolized OVA.
Ashino et al administered the JAK1/3 inhibitor R256 to mice and compared its effects when administered during the sensitization or challenge phases with use of OVA as a model allergen.24 When administered by oral gavage only during sensitization with OVA plus alum administered intraperitoneally, R256 inhibited the AHR, BAL eosinophilia, periodic acid–Schiff–positive airway cells, and TH2 cytokine production that developed during OVA aerosol challenge 21 days later. When administered only during OVA challenge, R256 also inhibited AHR, BAL eosinophilia, and mucus metaplasia; however, TH2 cytokine production was unaffected. This mimicked the effect of this compound on TH2 cell differentiation and cytokine production in vitro, in which case R526 inhibited cytokine-induced T-cell differentiation but not OVA-driven recall cytokine production in already-differentiated cells.
Dengler et al reported on the development of the novel inhaled pyrazolopyrimidine JAK inhibitor iJAK-381, which was very effective in different animal models of allergic airway inflammation.18 In kinase inhibition assays, iJAK-381 inhibited JAK1 (Ki = 0.26 nM) more than JAK2 (Ki = 0.62 nM) did, and it was less active against JAK3 (Ki = 20.8 nM) and Tyk2 (Ki = 3.15 nM). Although iJAK-381 inhibited IL-13–induced STAT6 phosphorylation, as well as JAK2-dependent EPO and IL-5 signaling in cell lines, when administered in vivo it was specific for STAT6 inhibition in the lung. Inhaled iJAK-381 blocked eosinophilic and neutrophilic airway inflammation and reactivity in 2 different mouse models with either OVA or a combination of Aspergillus, Alternaria, and house dust mite extracts used as model allergens. Inhaled iJAK-381 had no apparent systemic effects on spleen cellularity or natural killer cell count, which is a sensitive marker of systemic JAK inhibition.80 Inhaled iJak-381 also blocked OVA-induced airway inflammation in guinea pigs. Taken together, the data from this important study demonstrated proof of concept that a selective inhaled JAK inhibitor has therapeutic potential in asthma.
A new class of pyridone JAK inhibitors was recently developed; it demonstrated good selectivity, long lung retention time, and low oral bioavailability, suggesting that these compounds would be effective when administered via inhalation.20 Calbet et al reported that 1 member of this class, LAS194046, suppressed OVA-driven airway inflammation and airway reactivity (as measured by enhanced pause [Penh]) in rats.16 LAS194046 was a potent pan-JAK inhibitor, with IC50 values of 5.46, 0.4, and 2.07 nM, respectively, for JAK1, JAK2, and JAK3, and 21.8 nM for Tyk2, in in vitro kinase inhibition assays. LAS194046 demonstrated target engagement in the lung, as indicated by reduced levels of phospo-STAT1 and phospho-STAT6 in lung homogenates in the absence of pharmacologically relevant systemic plasma levels.
Caniga et al studied a new inhaled JAK inhibitor called iJAK-001 in a rat model of Alternaria extract–induced lung inflammation and found that when dosed intratracheally before challenge, it inhibited BAL eosinophilia in a dose-dependent manner.19 Substrate specificity for iJAK-001 was not described in the report. A single dose of iJAK-001 delivered 48 hours before challenge was also effective. iJAK-001–treated animals demonstrated improved lung retention and a greater lung-to-plasma ratio than did the more water-soluble JAK inhibitor tofacitinib. In sheep challenged with Ascaris suum, iJAK-001 (when delivered via a dry powder inhaler) inhibited the increase in airway resistance during both the early- and late-phase airway responses. Importantly, intravenously administered iJAK-001 (delivered to match the systemic exposure with the inhaled compound) had no effect in this model. This study adds further support to the idea that lung-restricted JAK inhibition is sufficient to achieve physiologically relevant end points in asthma models.
Younis et al developed a method to aerosolize tofacitinib, with a particle size estimated to deposit efficiently in the lung (1.2 ± 0.2-μm median mass aerodynamic diameter). They found that nebulized tofacitinib was significantly effective in reducing house dust mite–induced airway eosinophilia and also reduced BAL protein levels, a marker of barrier dysfunction.26 Their findings demonstrate that tofacitinib can be successfully nebulized at a droplet size suitable for apparent therapeutic efficacy.
Joo et al developed a mouse model of chronic rhinosinusitis by topical application of intranasal OVA in sensitized mice and compared the effect of intraperitoneally administered tofacitinib with that of triamcinolone.27 Tofacitinib effectively inhibited nasal inflammation, as determined by reduced numbers of tissue eosinophils and TH2 cytokines, and it was associated with target engagement, as determined by reduction in phospho-STAT6. Thus, JAK inhibitors can block both upper and lower airway pathology in mouse models of allergic airway inflammation.
In addition to these studies of allergen-driven airway inflammation, which involves effector cells of the adaptive immune system, JAK inhibitors have also been shown to dampen LPS-induced lung inflammation, which is mediated by the innate immune system. For example, Calama et al administered aerosolized LPS (0.1 mg/mL) and studied the effect that orally administered tofacitinib (3–30 mg/kg) administered 1 hour prior has on lung inflammation.81 Tofacitinib effectively suppressed lung phospho-STAT3 levels and BAL neutrophilia, and it selectively attenuated production of IL-6, MIP-1α, IL-1β, and MIP-3α without inhibiting TNF-α or CXCL1 levels in BAL fluids. Therefore, JAK inhibitors have the potential to suppress innate and adaptive immunity in the lung.
HUMAN CLINICAL STUDIES
The inhaled selective, small molecule JAK inhibitor GDC-0214 was recently evaluated by Braithwaite et al in a double-blind, randomized, placebo-controlled, phase I proof-of-activity study.11 GDC-0214 is a relative JAK1 inhibitor of the pyrazolopyrimidine class. In biochemical studies, GDC-0214 has a Ki for JAK1 inhibition of 0.40 nM, which is 2.3-fold more selective than for JAK2, 20-fold more selective than for JAK3, and 3-fold more selective than for Tyk2. In cell-based assays of cytokine-induced STAT phosphorylation, GDC-0214 has an IC50 value of 17 nM for JAK1 (IL-13–induced phosphorylation of STAT6), which is 12-fold more selective than for JAK2 (EPO-induced phosphorylation of JAK2). The trial included adults with mild asthma and a baseline Feno level greater than 40 ppb. All subjects completed a 4-day, single-blind placebo period followed by a 10-day, double-blind treatment period of either placebo or GDC-0214. The treatment arm comprised 4 sequential ascending dose cohorts of patients who were given 1 mg once daily, 4 mg once daily, 15 mg once daily, and 15 mg twice daily. Each dose-allocated cohort completed the treatment period before the subsequent higher-dosed cohort was treated. Mild asthma was defined as a physician-made diagnosis of 6 months or more and an FEV1 value greater than 70% predicted. Patients who had used ICSs within the past 60 days, had poor asthma control, or had a significant smoking history were excluded. A total of 36 patients were enrolled; their mean age was 28 years and their ages ranged from 18 to 56 years. The majority of patients were female (56%), predominately White (64%), and not of Hispanic or Latino origin (89%). Their mean baseline Feno level, established during the blinded placebo period, was 93 ppb, with a wide range of variation from 32 to 190 ppb. The primary outcome to establish drug activity was placebo-corrected percent reduction in Feno level from baseline to the end of the 10-day active treatment period.
Although changes in Feno level were nonsignificant for the 2 lowest doses of 1 mg daily and 4 mg daily,11 significant reductions in Feno level were observed in the cohorts treated with the 2 highest doses of 15 mg daily and 15 mg twice daily. At the end of the active treatment period, the placebo-corrected absolute reductions in Feno level for the 15-mg daily group and 15-mg twice-daily group were 18 ppb and 44 ppb, respectively. The changes in treatment group Feno level were placebo-corrected by 1.1 ppb (±20), which was the small difference observed in the pooled placebo sample between baseline and end of the treatment period. Following the treatment period, there was a 28-day safety monitoring follow-up period during which no deaths or serious adverse events were noted. Most patients (88.9%) reported at least 1 adverse event, but only 1 adverse event (thirst) was thought to be attributable to GDC-0214. Upper respiratory tract infections were the most commonly reported adverse outcome (30.6%), but the infection rate was not significantly different between the placebo and treatment groups. Low systemic exposure to GDC-0214 was expected on the basis of preclinical data and demonstrated by unaffected eosinophil counts, as well as by low plasma levels of the drug. Plasma levels of GDC-0214 measured at multiple time points during the study period were at least 15-fold less than concentrations expected to inhibit systemic JAK1. This important study established that local targeting of JAK1 is a feasible approach, and it opens the door to a new era of pharmacotherapy in asthma.
Ongoing studies of other JAK inhibitors in asthma that are currently listed on ClinicalTrials.gov are reported in Table III.
TABLE III.
Name of compound (route) | Study phase | Sponsor | Target | ClinicalTrials.gov identifier |
---|---|---|---|---|
AZD0449 (inhaled) | Completed | AstraZeneca | JAK1 | NCT03766399 |
VR588 (in vitro study) | Completed | Imperial College London | Pan-JAK | NCT02740049 |
AZD4604 (inhaled) | Not yet recruiting | AstraZeneca | Not specified | NCT04769869 |
TD-8236 (inhaled) | Completed | Theravance Biopharma | Pan-JAK | NCT04150341 |
TD-8236 (inhaled) | Completed | Theravance Biopharma | Pan-JAK | NCT03652038 |
SAFETY
An important aspect of considering JAK inhibitors in asthma is balancing their potential efficacy in reducing lung inflammation and AHR with safety concerns. The safety of JAK inhibitors has been intensively studied and scrutinized, especially in subjects with rheumatic diseases treated with tofacitinib,82–84 with ongoing postmarketing and surveillance studies that will lead to a better understanding of the long-term safety profile of these compounds in the coming years. Although early studies of tofacitinib were conducted in subjects treated with other disease-modifying antirheumatic drugs (DMARDs [eg, methotrexate]), which have their own potential side effects, subsequent clinical experience with JAK inhibitors has indicated serious potential safety concerns that will need to be carefully considered in clinical trials in asthma. Here we review 6 potential safety concerns of systemic JAK inhibitors: (1) opportunistic and respiratory viral infections, (2) viral reactivation, (3) malignancy and lymphoproliferative disorders, (4) decreased lymphocyte and neutrophil counts, (5) lipid profile and cardiovascular disease, and (6) venous thromboembolism (VTE). To date, there is insufficient experience with the use of topical JAK inhibitors, but it seems likely that most of these concerns will be significantly lessened in patients treated with inhaled JAK inhibitors.
Opportunistic and respiratory viral infections.
Because JAK inhibitors are immunosuppressive, infection should be considered as a potential risk of these agents. Serious and sometimes fatal infections due to bacterial, mycobacterial, invasive fungal, viral, or other opportunistic pathogens have been reported in patients with RA who are receiving tofacitinib.82–84 Infections were also among the most common adverse events in studies with baricitinib.85 The risk of serious infections in patients treated with JAK inhibitors needs to be considered in context with other populations. In patients with RA treated with tofacitinib the rate of serious infections is 2.7 per 100 patient years, whereas in general asthma populations the risk of serious infections is 1.8 per 100 patient years, and in individuals with asthma who are taking oral steroids the rate is 7.8 per 100 patient years.86 Therefore, the risk of serious infections in patients with RA treated with tofacitinib is less than that observed in individuals with severe, corticosteroid-dependent asthma. Because of the potential risk of infections with the administration of any JAK inhibitor, subjects with acute and chronic infections or a history of recurrent infections and/or latent infections should be excluded from studies with these compounds. All subjects treated with JAK inhibitors should be monitored for signs and symptoms of infection during treatment, with appropriate antimicrobial therapies initiated promptly when indicated.
Respiratory viral infections are of special concern in asthma given their association with asthma exacerbations. Neither respiratory viral infections nor asthma exacerbations have been reported with increased frequency in clinical trials of JAK inhibitors in other diseases to date, although these studies did not specifically enroll subjects with severe asthma or carefully monitor asthma exacerbations. Additionally, the striking clinical efficacy of different JAK inhibitors in patients with COVID-19 pneumonia is encouraging87 and indicates that these compounds can suppress deleterious lung inflammation associated with a life-threatening respiratory virus. On the other hand, individuals with severe asthma may be particularly susceptible to respiratory viral infections, and future clinical trials in asthma will need to carefully monitor viral infections and exacerbation rates in asthma.
Viral reactivation.
Viral reactivation has been reported with JAK inhibitors and appears to be a class effect with these compounds. Cases of herpes virus reactivation (eg, herpes zoster), including disseminated disease, were observed in clinical studies with tofacitinib and other JAK inhibitors.84,88 In patients with RA, the risk of herpes zoster is increased by concomitant use of prednisone (>7.5 mg per day), as well as other immunosuppressive agents (eg, methotrexate). In the general population, the incidence rate of herpes zoster per 100 person years ranges from 0.31 to 0.55, which is similar to the incidence rate in individuals with asthma who are not taking oral corticosteroids (0.47). In individuals with asthma who are taking 5 mg or more of prednisone per day, the incidence rate is 0.72 per 100 person years.86 In patients with RAwho are taking tofacitinib alone, the incidence rate is 2.2, whereas it increases to 3.8 in subjects using tofacitinib and oral corticosteroids and to between 4.4 and 5.4 in subjects using tofacitinib plus other immunosuppressive agents. Thus, subjects experiencing recurrent or disseminated herpes zoster, as well as subjects with other active viral infections (eg, hepatitis B and C), should be excluded from studies with JAK inhibitors. Appropriate antiviral vaccines (including zoster) should be administered to subjects before initiation of JAK inhibitor therapy where appropriate.
Malignancy and lymphoproliferative disorders.
Lymphoma and other malignancies have been observed in patients treated with tofacitinib.83 EBV-associated posttransplant lymphoproliferative disorder has been observed at an increased rate in renal transplant patients treated with tofacitinib and concomitant immunosuppressive medications.89 In controlled clinical trials and postmarketing studies in subjects with RA and subjects with psoriatic arthritis, higher rates of malignancy were observed in subjects treated with tofacitinib with or without another DMARD than in subjects treated with placebo with or without a DMARD.83 Although malignancies were observed in these observational studies, a meta-analysis comparing the risk of malignancies in patients with RA treated with tofacitinib and other DMARDs concluded that tofacitinib use did not increase the risk of malignancies.90 Furthermore, it is estimated that the rate of malignancy in the general asthma population (1.6–1.9 per 100 patient years) is comparable to the rate of malignancy in patients with RA treated with tofacitinib (0.8–1.8 per 100 patient years).91 The ORAL Surveillance study (ClinicalTrials.gov identifier NCT02092467) is an FDA-mandated postmarketing safety study intended to demonstrate noninferiority of tofacitinib to TNF inhibitors among older patients with RA from the standpoint of risk of cardiovascular events and malignancies. In February 2021, on the basis of preliminary results from the ORAL Surveillance study, the FDA issued a safety alert warning about the risk of cancer in older subjects taking tofacitinib for RA. The final results of this and other studies will need to be monitored to help assess the risk-benefit ratio of using JAK inhibitors in other inflammatory diseases.
Decreased lymphocyte and neutrophil counts.
Tofacitinib has been associated with initial lymphocytosis at 1 month of exposure, followed by a gradual decrease in mean lymphocyte counts of approximately 10% during 12 months of therapy. Counts less than 500 cells/mm3 were associated with an increased incidence of treated and serious infections. Tofacitinib has also been associated with an increased incidence of neutropenia (<2000 cells/mm3) versus the incidence with placebo.84 In 2 phase 3 clinical studies of ruxolitinib, 1% of patients had to have their dose reduced or their treatment stopped because of neutropenia.92 Dose-dependent decreases in neutrophil counts were also observed in patients treated with baricitinib, but infections were rare even in subjects with neutropenia.93 Lymphocyte and neutrophil counts should be monitored closely in subjects taking JAK inhibitors.
Lipid profile and cardiovascular disease.
JAK blockade by tofacitinib was associated with increases in lipid parameters, including total cholesterol, low-density lipoprotein, and high-density lipoprotein levels. Maximum effects were generally observed within 6 weeks. Elevated lipid levels were also observed in subjects treated with ruxolitinib and baricitinib. In ongoing studies in subjects with RA and psoriasis, the alterations in lipid profiles have not translated into an increased incidence of major cardiac adverse events.94–96 However, in 2021 the FDA announced that on the basis of preliminary analyses of the ORAL Surveillance study (ClinicalTrials.gov identifier NCT02092467), older RA subjects treated with tofacitinib experienced more major cardiovascular events than did those treated with TNF inhibitors. RA is known to be an independent risk factor for cardiovascular disease, and final results from this study will be needed to help us understand the cardiovascular risk profile of JAK inhibitors in different patient populations.
VTE.
Concerns were raised about the risk of VTE in subjects treated with higher doses of JAK inhibitors, and upadicitinib, tofacitinib, and baricitinib all carry black box warnings about the risk of thrombosis. Whether or how JAK inhibitors induce a prothrombotic state is not known, and observational studies are confounded by the fact that many immune and inflammatory diseases are associated with increased risk of VTE. Transient increases in platelet counts have been observed following initiation of JAK inhibitors,97 but these increases were not associated with thrombotic events. A recent metanalysis concluded that the risk of VTE associated with JAK inhibitor use was low.98 More information clarifying the potential prothrombotic risks of systemic JAK inhibition should be forthcoming soon.
Other safety considerations.
Other safety considerations with JAK inhibitors include anemia and elevation of liver enzyme levels, which occurred in 1% to 2% of patients. Most of these abnormalities have occurred in studies with background DMARD (primarily methotrexate) therapy.
CONCLUSIONS
JAK inhibitors are a new class of anti-inflammatory agents that have strong therapeutic potential in asthma. On the basis of their broad mechanism of action, JAK inhibitors should be clinically useful in multiple different asthma endotypes, including in subjects with type 2–high and type 2–low asthma. An obvious unmet need is in individuals with severe asthma whose disease is poorly controlled despite high-dose ICSs plus other controllers, in which case JAK inhibitors could represent a more convenient (via pill or inhaler) and less expensive therapy than biologics. JAK inhibitors have the potential to improve symptom control, reduce exacerbations. and minimize the use of systemic corticosteroids. The benefits of JAK inhibitors will need to be carefully balanced with potential risks. The ongoing development of inhaled JAK inhibitors is particularly exciting because these compounds should have a better safety profile than systemic JAK inhibitors in asthma.
As we await more efficacy and safety data from studies of JAK inhibitors in other inflammatory diseases, we can speculate about where to position these therapies in the asthma armamentarium. Systemic JAK inhibitors should be investigated in individuals with severe and/or exacerbation-prone asthma, who require maintenance or frequent bursts of systemic steroids to achieve control. In these subjects, even a single course of systemic glucocorticoids is associated with serious side effects, and maintenance steroids are associated with substantial morbidity and mortality.99–102 The safety profile of systemic JAK inhibitors reported to date seems to be at least comparable to that of systemic glucocorticoids. Because JAK inhibitors have a completely different mechanism of action, they may be particularly effective in individuals with asthma who are refractory or resistant to the anti-inflammatory effects of glucocorticoids. Inhaled JAK inhibitors should be studied in subjects with severe and mild-to-moderate asthma, both as maintenance inhalers and on demand during periods of loss of control. As JAK inhibitors enter clinical trials in asthma, these compounds should also provide insights into disease pathobiology. An important area for future research will be identifying biomarkers that predict response to therapy and can be used to monitor response to therapy. This will help us tailor therapy to the best-responding subset of patients and achieve the goal of personalized medicine in asthma.
Disclosure of potential conflict of interest:
S. N. Georas reports receiving grants from the National Institutes of Health for asthma research and personal fees from Merck outside the submitted work; his spouse reports grants and personal fees from AbbVie, Allakos, AstraZeneca, Benevolent AIBio, DermTech, Galderma, Incyte, Janssen, Kiniksa, LEO Pharma, Lilly, Novartis, Pfizer, Principia Biopharma, Rapt Therapeutics, Regeneron, Sanofi/Genzyme, Sanofi-Aventis, and Stealth Biotherapeutics, for the design and conduct of clinical trials in atopic dermatitis, including some with JAK inhibitors. M. E. Wechsler reports consulting honoraria/fees from GlaxoSmithKline, AstraZeneca, Sanofi, Regeneron, Boehringer Ingelheim, Teva, Sentien, Cohero, Genzyme, Novartis, Genentech, Restorbio, and Equillium outside of the submitted work. The rest of the authors declare that they have no relevant conflicts of interest.
S.N.G. was supported by grants from the NIH including R01 AI144241 and U24 HL138998. P.D. and M.C. were supported by NIH T32 HL066988. M.E.W. was supported by NIH UG1 HL139123 and the Jin Hua Foundation. The authors gratefully acknowledge helpful discussion with the PrecISE Asthma Network Jak Inhibitor Working Group (Praveen Akuthota, Anastasia Ivanova, Jonathan M. Gaffin, James Moy, Sally J. Wenzel).
Abbreviations used
- AHR
Airway hyperresponsiveness
- ASM
Airway smooth muscle
- BAL
Bronchoalveolar lavage
- DMARD
Disease-modifying antirheumatic drug
- EPO
Erythropoietin
- FDA
US Food and Drug Administration
- Feno
Fractional exhaled nitric oxide
- IC50
Inhibitory concentration of 50%
- ICS
Inhaled corticosteroid
- ILC2
Group 2 innate lymphoid cell
- iNOS
Inducible nitric oxide synthase
- JAK
Janus kinase
- Ki
Inhibitory constant
- OVA
Ovalbumin
- RA
Rheumatoid arthritis
- STAT
Signal transducer and activator of transcription
- Treg
Regulatory T
- TSLP
Thymic stromal lymphopoietin
- VTE
Venous thromboembolism
REFERENCES
- 1.Israel E, Reddel HK. Severe and difficult-to-treat asthma in adults. N Engl J Med 2017;377:965–76. [DOI] [PubMed] [Google Scholar]
- 2.Wenzel SE. Severe adult asthmas: integrating clinical features, biology, and therapeutics to improve outcomes. Am J Respir Crit Care Med 2021;203:809–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Fahy JV. Type 2 inflammation in asthma - present in most, absent in many. Nat Rev Immunol 2014;15:57–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.O’Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, Laurence A. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu Rev Med 2015;66:311–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Li X, Bechara R, Zhao J, McGeachy MJ, Gaffen SL. IL-17 receptor-based signaling and implications for disease. Nat Immunol 2019;20:1594–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Liew FY, Girard JP, Turnquist HR. Interleukin-33 in health and disease. Nat Rev Immunol 2016;16:676–89. [DOI] [PubMed] [Google Scholar]
- 7.Zak M, Hanan EJ, Lupardus P, Brown DG, Robinson C, Siu M, et al. Discovery of a class of highly potent Janus kinase 1/2 (JAK1/2) inhibitors demonstrating effective cell-based blockade of IL-13 signaling. Bioorg Med Chem Lett 2019;29:1522–31. [DOI] [PubMed] [Google Scholar]
- 8.Sohn SJ, Barrett K, Van Abbema A, Chang C, Kohli PB, Kanda H, et al. A restricted role for TYK2 catalytic activity in human cytokine responses revealed by novel TYK2-selective inhibitors. J Immunol 2013;191:2205–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.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. [DOI] [PubMed] [Google Scholar]
- 10.Howell MD, Fitzsimons C, Smith PA. JAK/STAT inhibitors and other small molecule cytokine antagonists for the treatment of allergic disease. Ann Allergy Asthma Immunol 2018;120:367–75. [DOI] [PubMed] [Google Scholar]
- 11.Braithwaite IE, Cai F, Tom JA, Galanter JM, Owen RP, Zhu R, et al. Inhaled JAK inhibitor GDC-0214 reduces exhaled nitric oxide in patients with mild asthma: a randomized, controlled, proof-of-activity trial. J Allergy Clin Immunol 2021. [DOI] [PubMed] [Google Scholar]
- 12.Gadina M, Johnson C, Schwartz D, Bonelli M, Hasni S, Kanno Y, et al. Translational and clinical advances in JAK-STAT biology: the present and future of jakinibs. J Leukoc Biol 2018. [DOI] [PubMed] [Google Scholar]
- 13.Howell MD, Kuo FI, Smith PA. Targeting the Janus kinase family in autoimmune skin diseases. Front Immunol 2019;10:2342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Strober B, Buonanno M, Clark JD, Kawabata T, Tan H, Wolk R, et al. Effect of tofacitinib, a Janus kinase inhibitor, on haematological parameters during 12 weeks of psoriasis treatment. Br J Dermatol 2013;169:992–9. [DOI] [PubMed] [Google Scholar]
- 15.McInnes IB, Byers NL, Higgs RE, Lee J, Macias WL, Na S, et al. Comparison of baricitinib, upadacitinib, and tofacitinib mediated regulation of cytokine signaling in human leukocyte subpopulations. Arthritis Res Ther 2019;21:183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Calbet M, Ramis I, Calama E, Carreno C, Paris S, Maldonado M, et al. Novel inhaled pan-JAK inhibitor, LAS194046, reduces allergen-induced airway inflammation, late asthmatic response, and pSTAT activation in brown Norway rats. J Pharmacol Exp Ther 2019;370:137–47. [DOI] [PubMed] [Google Scholar]
- 17.Jones P, Storer RI, Sabnis YA, Wakenhut FM, Whitlock GA, England KS, et al. Design and synthesis of a pan-janus kinase inhibitor clinical candidate (PF-06263276) suitable for inhaled and topical delivery for the treatment of inflammatory diseases of the lungs and skin. J Med Chem 2017;60:767–86. [DOI] [PubMed] [Google Scholar]
- 18.Dengler HS, Wu X, Peng I, Rinderknecht CH, Kwon Y, Suto E, et al. Lung-restricted inhibition of Janus kinase 1 is effective in rodent models of asthma. Sci Transl Med 2018;10:eaao2151. [DOI] [PubMed] [Google Scholar]
- 19.Caniga M, Yu H, Lee HH, Wang M, Witter D, Salmon M, et al. Estimation of fraction dissolved after intratracheal delivery of a potent janus kinase inhibitor, iJAK-001, with low solubility in rat and sheep: impact of preclinical PKPD on inhaled human dose projection. J Aerosol Med Pulm Drug Deliv 2019;32:251–65. [DOI] [PubMed] [Google Scholar]
- 20.Bach J, Eastwood P, Gonzalez J, Gomez E, Alonso JA, Fonquerna S, et al. Identification of 2-imidazopyridine and 2-aminopyridone purinones as potent pan-Janus kinase (JAK) inhibitors for the inhaled treatment of respiratory diseases. J Med Chem 2019;62:9045–60. [DOI] [PubMed] [Google Scholar]
- 21.Zak M, Dengler HS, Rajapaksa NS. Inhaled Janus kinase (JAK) inhibitors for the treatment of asthma. Bioorg Med Chem Lett 2019;29:126658. [DOI] [PubMed] [Google Scholar]
- 22.Kudlacz E, Conklyn M, Andresen C, Whitney-Pickett C, Changelian P. The JAK-3 inhibitor CP-690550 is a potent anti-inflammatory agent in a murine model of pulmonary eosinophilia. Eur J Pharmacol 2008;582:154–61. [DOI] [PubMed] [Google Scholar]
- 23.Matsunaga Y, Inoue H, Fukuyama S, Yoshida H, Moriwaki A, Matsumoto T, et al. Effects of a Janus kinase inhibitor, pyridone 6, on airway responses in a murine model of asthma. Biochem Biophys Res Commun 2011;404:261–7. [DOI] [PubMed] [Google Scholar]
- 24.Ashino S, Takeda K, Li H, Taylor V, Joetham A, Pine PR, et al. Janus kinase 1/3 signaling pathways are key initiators of TH2 differentiation and lung allergic responses. J Allergy Clin Immunol 2014;133:1162–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Li RF, Wang GF. JAK/STAT5 signaling pathway inhibitor ruxolitinib reduces airway inflammation of neutrophilic asthma in mice model. Eur Rev Med Pharmacol Sci 2018;22:835–43. [DOI] [PubMed] [Google Scholar]
- 26.Younis US, Vallorz E, Addison KJ, Ledford JG, Myrdal PB. Preformulation and evaluation of tofacitinib as a therapeutic treatment for asthma. AAPS PharmSci-Tech 2019;20:167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Joo YH, Cho HJ, Jeon YJ, Kim JH, Jung MH, Jeon SY, et al. Therapeutic effects of intranasal tofacitinib on chronic rhinosinusitis with nasal polyps in mice. Laryngoscope 2021;131:E1400–7. [DOI] [PubMed] [Google Scholar]
- 28.Kuperman DA, Huang X, Koth LL, Chang GH, Dolganov GM, Zhu Z, et al. Direct effects of interleukin-13 on epithelial cells cause airway hyperreactivity and mucus overproduction in asthma. Nat Med 2002;8:885–9. [DOI] [PubMed] [Google Scholar]
- 29.Kuperman DA, Lewis CC, Woodruff PG, Rodriguez MW, Yang YH, Dolganov GM, et al. Dissecting asthma using focused transgenic modeling and functional genomics. J Allergy Clin Immunol 2005;116:305–11. [DOI] [PubMed] [Google Scholar]
- 30.Ruterbusch M, Pruner KB, Shehata L, Pepper M. In vivo CD4(+) T cell differentiation and function: revisiting the Th1/Th2 paradigm. Annu Rev Immunol 2020;38:705–25. [DOI] [PubMed] [Google Scholar]
- 31.Liu S, Verma M, Michalec L, Liu W, Sripada A, Rollins D, et al. Steroid resistance of airway type 2 innate lymphoid cells from patients with severe asthma: the role of thymic stromal lymphopoietin. J Allergy Clin Immunol 2018;141:257–68.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.van der Ploeg EK, Golebski K, van Nimwegen M, Fergusson JR, Heesters BA, Martinez-Gonzalez I, et al. Steroid-resistant human inflammatory ILC2s are marked by CD45RO and elevated in type 2 respiratory diseases. Sci Immunol 2021;6:eabd3489. [DOI] [PubMed] [Google Scholar]
- 33.Verma M, Michalec L, Sripada A, McKay J, Sirohi K, Verma D, et al. The molecular and epigenetic mechanisms of innate lymphoid cell (ILC) memory and its relevance for asthma. J Exp Med 2021;218:e20201354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hondowicz BD, An D, Schenkel JM, Kim KS, Steach HR, Krishnamurty AT, et al. Interleukin-2-dependent allergen-specific tissue-resident memory cells drive asthma. Immunity 2016;44:155–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Rahimi RA, Nepal K, Cetinbas M, Sadreyev RI, Luster AD. Distinct functions of tissue-resident and circulating memory Th2 cells in allergic airway disease. J Exp Med 2020;217:e20190865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Pruner KB, Pepper M. Local memory CD4 T cell niches in respiratory viral infection. J Exp Med 2021;218:e20201733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ross SH, Cantrell DA. Signaling and function of interleukin-2 in T lymphocytes. Annu Rev Immunol 2018;36:411–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sewgobind VD, Quaedackers ME, van der Laan LJ, Kraaijeveld R, Korevaar SS, Chan G, et al. The Jak inhibitor CP-690,550 preserves the function of CD4CD25FoxP3 regulatory T cells and inhibits effector T cells. Am J Transplant 2010;10:1785–95. [DOI] [PubMed] [Google Scholar]
- 39.Corren J, Castro M, O’Riordan T, Hanania NA, Pavord ID, Quirce S, et al. Dupilumab efficacy in patients with uncontrolled, moderate-to-severe allergic asthma. J Allergy Clin Immunol Pract 2020;8:516–26. [DOI] [PubMed] [Google Scholar]
- 40.McKnight CG, Potter C, Finkelman FD. IL-4Ralpha expression by airway epithelium and smooth muscle accounts for nearly all airway hyperresponsiveness in murine allergic airway disease. Mucosal Immunol 2020;13:283–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Matsukura S, Stellato C, Georas SN, Casolaro V, Plitt JR, Miura K, et al. Inter-leukin-13 upregulates eotaxin expression in airway epithelial cells by a STAT6-dependent mechanism. Am J Respir Cell Mol Biol 2001;24:755–61. [DOI] [PubMed] [Google Scholar]
- 42.Woodruff PG, Modrek B, Choy DF, Jia G, Abbas AR, Ellwanger A, et al. T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med 2009;180:388–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wang WW, Lu DM, Zheng M, Zhang JG, Zhang B. TSLP regulates eotaxin-1 production by nasal epithelial cells from patients with eosinophilic CRSwNP. Rhinology 2018;56:370–7. [DOI] [PubMed] [Google Scholar]
- 44.Sada M, Watanabe M, Inui T, Nakamoto K, Hirata A, Nakamura M, et al. Ruxolitinib inhibits poly(I:C) and type 2 cytokines-induced CCL5 production in bronchial epithelial cells: a potential therapeutic agent for severe eosinophilic asthma. Immun Inflamm Dis 2021;9:363–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Heller NM, Matsukura S, Georas SN, Boothby MR, Stellato C, Schleimer RP. Assessment of signal transducer and activator of transcription 6 as a target of glucocorticoid action in human airway epithelial cells. Clin Exp Allergy 2004; 34:1690–700. [DOI] [PubMed] [Google Scholar]
- 46.Peters MC, Kerr S, Dunican EM, Woodruff PG, Fajt ML, Levy BD, et al. Refractory airway type 2 inflammation in a large subgroup of asthmatic patients treated with inhaled corticosteroids. J Allergy Clin Immunol 2019;143:104–13.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Georas SN, Rezaee F. Epithelial barrier function: at the front line of asthma immunology and allergic airway inflammation. J Allergy Clin Immunol 2014; 134:509–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Akdis CA. Does the epithelial barrier hypothesis explain the increase in allergy, autoimmunity and other chronic conditions? Nat Rev Immunol 2021. [DOI] [PubMed] [Google Scholar]
- 49.Saatian B, Rezaee F, Desando S, Emo J, Chapman T, Knowlden S, et al. Interleukin-4 and interleukin-13 cause barrier dysfunction in human airway epithelial cells. Tissue Barriers 2013;1:e24333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Amano W, Nakajima S, Kunugi H, Numata Y, Kitoh A, Egawa G, et al. The Janus kinase inhibitor JTE-052 improves skin barrier function through suppressing signal transducer and activator of transcription 3 signaling. J Allergy Clin Immunol 2015;136:667–77.e7. [DOI] [PubMed] [Google Scholar]
- 51.Grunstein MM, Hakonarson H, Leiter J, Chen M, Whelan R, Grunstein JS, et al. IL-13-dependent autocrine signaling mediates altered responsiveness of IgE-sensitized airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2002; 282:L520–8. [DOI] [PubMed] [Google Scholar]
- 52.Moore PE, Church TL, Chism DD, Panettieri RA Jr, Shore SA. IL-13 and IL-4 cause eotaxin release in human airway smooth muscle cells: a role for ERK. Am J Physiol Lung Cell Mol Physiol 2002;282:L847–53. [DOI] [PubMed] [Google Scholar]
- 53.Eum SY, Maghni K, Tolloczko B, Eidelman DH, Martin JG. IL-13 may mediate allergen-induced hyperresponsiveness independently of IL-5 or eotaxin by effects on airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2005;288: L576–84. [DOI] [PubMed] [Google Scholar]
- 54.Amrani Y, Syed F, Huang C, Li K, Liu V, Jain D, et al. Expression and activation of the oxytocin receptor in airway smooth muscle cells: regulation by TNFalpha and IL-13. Respir Res 2010;11:104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Manson ML, Safholm J, James A, Johnsson AK, Bergman P, Al-Ameri M, et al. IL-13 and IL-4, but not IL-5 nor IL-17A, induce hyperresponsiveness in isolated human small airways. J Allergy Clin Immunol 2020;145:808–17.e2. [DOI] [PubMed] [Google Scholar]
- 56.Farne HA, Wilson A, Powell C, Bax L, Milan SJ. Anti-IL5 therapies for asthma. Cochrane Database Syst Rev 2017;9:CD010834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.King B, Lee AI, Choi J. Treatment of hypereosinophilic syndrome with cutaneous involvement with the JAK inhibitors tofacitinib and ruxolitinib. J Invest Dermatol 2017;137:951–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Del Bel KL, Ragotte RJ, Saferali A, Lee S, Vercauteren SM, Mostafavi SA, et al. JAK1 gain-of-function causes an autosomal dominant immune dysregulatory and hypereosinophilic syndrome. J Allergy Clin Immunol 2017;139:2016–20.e5. [DOI] [PubMed] [Google Scholar]
- 59.Stenova E, Tarabcakova L, Babal P, Kasperova S. Hypereosinophilic syndrome-a rare adverse event of anti-cytokine treatment in rheumatoid arthritis resolved after Janus kinase inhibitor therapy. Clin Rheumatol 2020;39:3507–10. [DOI] [PubMed] [Google Scholar]
- 60.Peters MC, McGrath KW, Hawkins GA, Hastie AT, Levy BD, Israel E, et al. Plasma interleukin-6 concentrations, metabolic dysfunction, and asthma severity: a cross-sectional analysis of two cohorts. Lancet Respir Med 2016;4: 574–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Li X, Hastie AT, Peters MC, Hawkins GA, Phipatanakul W, Li H, et al. Investigation of the relationship between IL-6 and type 2 biomarkers in patients with severe asthma. J Allergy Clin Immunol 2020;145:430–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Permaul P, Peters MC, Petty CR, Cardet JC, Ly NP, Ramratnam SK, et al. The association of plasma IL-6 with measures of asthma morbidity in a moderate-severe pediatric cohort aged 6–18 years. J Allergy Clin Immunol Pract 2021;9: 2916–9.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Jevnikar Z, Ostling J, Ax E, Calven J, Thorn K, Israelsson E, et al. Epithelial IL-6 trans-signaling defines a new asthma phenotype with increased airway inflammation. J Allergy Clin Immunol 2019;143:577–90. [DOI] [PubMed] [Google Scholar]
- 64.Israel E, Denlinger LC, Bacharier LB, LaVange LM, Moore WC, Peters MC, et al. PrecISE: Precision Medicine in Severe Asthma: an adaptive platform trial with biomarker ascertainment. J Allergy Clin Immunol 2021;147:1594–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Raundhal M, Morse C, Khare A, Oriss TB, Milosevic J, Trudeau J, et al. High IFN-gamma and low SLPI mark severe asthma in mice and humans. J Clin Invest 2015;125:3037–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ray A, Raundhal M, Oriss TB, Ray P, Wenzel SE. Current concepts of severe asthma. J Clin Invest 2016;126:2394–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Camiolo MJ, Zhou X, Oriss TB, Yan Q, Gorry M, Horne W, et al. High-dimensional profiling clusters asthma severity by lymphoid and non-lymphoid status. Cell Rep 2021;35:108974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Menson KE, Mank MM, Reed LF, Walton CJ, Van Der Vliet KE, Ather JL, et al. Therapeutic efficacy of IL-17A neutralization with corticosteroid treatment in a model of antigen-driven mixed-granulocytic asthma. Am J Physiol Lung Cell Mol Physiol 2020;319:L693–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Busse WW, Holgate S, Kerwin E, Chon Y, Feng J, Lin J, et al. Randomized, double-blind, placebo-controlled study of brodalumab, a human anti-IL-17 receptor monoclonal antibody, in moderate to severe asthma. Am J Respir Crit Care Med 2013;188:1294–302. [DOI] [PubMed] [Google Scholar]
- 70.Ghoreschi K, Jesson MI, Li X, Lee JL, Ghosh S, Alsup JW, et al. Modulation of innate and adaptive immune responses by tofacitinib (CP-690,550). J Immunol 2011;186:4234–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Ahern PP, Schiering C, Buonocore S, McGeachy MJ, Cua DJ, Maloy KJ, et al. Interleukin-23 drives intestinal inflammation through direct activity on T cells. Immunity 2010;33:279–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ghoreschi K, Laurence A, Yang XP, Tato CM, McGeachy MJ, Konkel JE, et al. Generation of pathogenic T(H)17 cells in the absence of TGF-beta signalling. Nature 2010;467:967–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Zielinski CE, Mele F, Aschenbrenner D, Jarrossay D, Ronchi F, Gattorno M, et al. Pathogen-induced human TH17 cells produce IFN-gamma or IL-10 and are regulated by IL-1beta. Nature 2012;484:514–8. [DOI] [PubMed] [Google Scholar]
- 74.Cahill KN, Katz HR, Cui J, Lai J, Kazani S, Crosby-Thompson A, et al. KIT Inhibition by imatinib in patients with severe refractory asthma. N Engl J Med 2017;376:1911–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Linnekin D Early signaling pathways activated by c-Kit in hematopoietic cells. Int J Biochem Cell Biol 1999;31:1053–74. [DOI] [PubMed] [Google Scholar]
- 76.Pullen NA, Falanga YT, Morales JK, Ryan JJ. The Fyn-STAT5 pathway: a new frontier in IgE- and IgG-mediated mast cell signaling. Front Immunol 2012;3: 117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Babina M, Wang Z, Franke K, Zuberbier T. Thymic stromal lymphopoietin promotes MRGPRX2-triggered degranulation of skin mast cells in a STAT5-dependent manner with further support from JNK. Cells 2021;10:102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Hermans MAW, Schrijver B, van Holten-Neelen C, Gerth van Wijk R, van Hagen PM, van Daele PLA, et al. The JAK1/JAK2- inhibitor ruxolitinib inhibits mast cell degranulation and cytokine release. Clin Exp Allergy 2018;48:1412–20. [DOI] [PubMed] [Google Scholar]
- 79.Aguilar-Pimentel A, Graessel A, Alessandrini F, Fuchs H, Gailus-Durner V, Hrabe de Angelis M, et al. Improved efficacy of allergen-specific immunotherapy by JAK inhibition in a murine model of allergic asthma. PLoS One 2017;12: e0178563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Vian L, Le MT, Gazaniga N, Kieltyka J, Liu C, Pietropaolo G, et al. JAK inhibition differentially affects NK cell and ILC1 homeostasis. Front Immunol 2019;10: 2972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Calama E, Ramis I, Domenech A, Carreno C, De Alba J, Prats N, et al. Tofacitinib ameliorates inflammation in a rat model of airway neutrophilia induced by inhaled LPS. Pulm Pharmacol Ther 2017;43:60–7. [DOI] [PubMed] [Google Scholar]
- 82.Winthrop KL. The emerging safety profile of JAK inhibitors in rheumatic disease. Nat Rev Rheumatol 2017;13:234–43. [DOI] [PubMed] [Google Scholar]
- 83.Alvaro-Gracia JM, Garcia-Llorente JF, Valderrama M, Gomez S, Montoro M. Update on the safety profile of tofacitinib in rheumatoid arthritis from clinical trials to real-world studies: a narrative review. Rheumatol Ther 2021;8:17–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Cohen SB, Tanaka Y, Mariette X, Curtis JR, Lee EB, Nash P, et al. Long-term safety of tofacitinib up to 9.5 years: a comprehensive integrated analysis of the rheumatoid arthritis clinical development programme. RMD Open 2020;6: 3001395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Keystone EC, Taylor PC, Drescher E, Schlichting DE, Beattie SD, Berclaz PY, et al. Safety and efficacy of baricitinib at 24 weeks in patients with rheumatoid arthritis who have had an inadequate response to methotrexate. Ann Rheum Dis 2015;74:333–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bloechliger M, Reinau D, Spoendlin J, Chang SC, Kuhlbusch K, Heaney LG, et al. Adverse events profile of oral corticosteroids among asthma patients in the UK: cohort study with a nested case-control analysis. Respir Res 2018;19:75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Chen CX, Wang JJ, Li H, Yuan LT, Gale RP, Liang Y. JAK-inhibitors for corona-virus disease-2019 (COVID-19): a meta-analysis [e-pub ahead of print]. Leukemia 2021. May 14:1–5. 10.1038/s41375-021-01266-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Winthrop KL, Curtis JR, Lindsey S, Tanaka Y, Yamaoka K, Valdez H, et al. Herpes zoster and tofacitinib: clinical outcomes and the risk of concomitant therapy. Arthritis Rheumatol 2017;69:1960–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Zand MS. Tofacitinab in renal transplantation. Transplant Rev (Orlando) 2013;27: 85–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Maneiro JR, Souto A, Gomez-Reino JJ. Risks of malignancies related to tofacitinib and biological drugs in rheumatoid arthritis: systematic review, meta-analysis, and network meta-analysis. Semin Arthritis Rheum 2017;47:149–56. [DOI] [PubMed] [Google Scholar]
- 91.Woo A, Lee SW, Koh HY, Kim MA, Han MY, Yon DK. Incidence of cancer after asthma development: 2 independent population-based cohort studies. J Allergy Clin Immunol 2021;147:135–43. [DOI] [PubMed] [Google Scholar]
- 92.Arana Yi C, Tam CS, Verstovsek S. Efficacy and safety of ruxolitinib in the treatment of patients with myelofibrosis. Future Oncol 2015;11:719–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kay J, Harigai M, Rancourt J, Dickson C, Melby T, Issa M, et al. Changes in selected haematological parameters associated with JAK1/JAK2 inhibition observed in patients with rheumatoid arthritis treated with baricitinib. RMD Open 2020;6:e001370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Wolk R, Armstrong EJ, Hansen PR, Thiers B, Lan S, Tallman AM, et al. Effect of tofacitinib on lipid levels and lipid-related parameters in patients with moderate to severe psoriasis. J Clin Lipidol 2017;11:1243–56. [DOI] [PubMed] [Google Scholar]
- 95.Gladman DD, Charles-Schoeman C, McInnes IB, Veale DJ, Thiers B, Nurmohamed M, et al. Changes in lipid levels and incidence of cardiovascular events following tofacitinib treatment in patients with psoriatic arthritis: a pooled analysis across phase III and long-term extension studies. Arthritis Care Res (Hoboken) 2019;71:1387–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Conaghan PG, Mysler E, Tanaka Y, Da Silva-Tillmann B, Shaw T, Liu J, et al. Upadacitinib in rheumatoid arthritis: a benefit-risk assessment across a phase III program. Drug Saf 2021;44:515–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Al-Salama ZT, Scott LJ. Baricitinib: a review in rheumatoid arthritis. Drugs 2018; 78:761–72. [DOI] [PubMed] [Google Scholar]
- 98.Yates M, Mootoo A, Adas M, Bechman K, Rampes S, Patel V, et al. Venous thromboembolism risk with JAK inhibitors: a meta-analysis. Arthritis Rheumatol 2021;73:779–88. [DOI] [PubMed] [Google Scholar]
- 99.Waljee AK, Rogers MA, Lin P, Singal AG, Stein JD, Marks RM, et al. Short term use of oral corticosteroids and related harms among adults in the United States: population based cohort study. BMJ 2017;357:j1415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ekstrom M, Nwaru BI, Hasvold P, Wiklund F, Telg G, Janson C. Oral corticosteroid use, morbidity and mortality in asthma: a nationwide prospective cohort study in Sweden. Allergy 2019;74:2181–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Matsunaga K, Adachi M, Nagase H, Okoba T, Hayashi N, Tohda Y. Association of low-dosage systemic corticosteroid use with disease burden in asthma. NPJ Prim Care Respir Med 2020;30:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Bleecker ER, Menzies-Gow AN, Price DB, Bourdin A, Sweet S, Martin AL, et al. Systematic literature review of systemic corticosteroid use for asthma management. Am J Respir Crit Care Med 2020;201:276–93. [DOI] [PMC free article] [PubMed] [Google Scholar]