- Review
- Open access
- Published:
JAK inhibitors: a new choice for diabetes mellitus?
Diabetology & Metabolic Syndrome volume 17, Article number: 33 (2025)
Abstract
Altered tyrosine kinase signaling is associated with a variety of diseases. Tyrosine kinases can be classified into two groups: receptor type and nonreceptor type. Nonreceptor-type tyrosine kinases are subdivided into Janus kinases (JAKs), focal adhesion kinases (FAKs) and tec protein tyrosine kinases (TECs). The beneficial effects of receptor-type tyrosine kinase inhibitors (TKIs) for the treatment of diabetes mellitus (DM) and the mechanisms involved have been previously described. Recently, several clinical cases involving the reversal of type 1 diabetes mellitus (T1DM) during treatment with JAK inhibitors have been reported, and clinical studies have described the improvement of type 2 diabetes mellitus (T2DM) during treatment with JAK inhibitors. In vivo and in vitro experimental studies have elucidated some of the mechanisms behind this effect, which seem to be based mainly on the reduction in β-cell disruption and the improvement of insulin resistance. In this review, we briefly describe the beneficial effects of JAK inhibitors among nonreceptor tyrosine kinase inhibitors for the treatment of DM and attempt to analyze the mechanisms involved.
Introduction
Diabetes mellitus (DM) is a common endocrine disease. Currently, the incidence of DM is rapidly increasing globally, and acute and chronic complications, especially chronic complications involving multiple organs and high rates of disability and death, seriously affect the physical and mental health of patients; thus, the increasing disease burden of DM is a major public health concern. DM can be classified into various types, but the most common types are type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). T1DM is caused by autoimmune β-cell destruction, which usually results in absolute insulin deficiency, including potential autoimmune diabetes mellitus during adulthood. T2DM is caused by a gradual decrease in β-cell insulin secretion, which is often accompanied by insulin resistance [1]. JAK-STAT(Janus kinase-signal transducer and activator of transcription) is a newly discovered intracellular signaling pathway closely related to cytokines in recent years and is involved in many important biological processes, such as cell proliferation, differentiation, apoptosis, and immunomodulation. Cytokine receptor binding transmits signals to tyrosine to phosphorylate and activate receptor-associated JAK, which then phosphorylates and activates STATs. Tyrosine-phosphorylated STATs (pSTATs) form dimers that are translocated to the nucleus, where they bind to target DNA sequences and regulate gene expression (the pSTAT canonical pathway). Unphosphorylated STATs also form dimers that enter the nucleus and regulate transcription (uSTAT non-canonical pathway). Multiple inducible negative feedback systems are used to constrain signaling circuits, including suppressor of cytokine signaling (SOCS) family proteins, inhibitor of activated STAT protein (PIAS) family proteins, tyrosine phosphatase proteins (PTPs), USP18 and ISG15 [2]. This pathway has been shown to be integral to both type I (IFN-α/β) and type II (IFNγ) interferons (collectively also known as type II cytokines) and to all cytokines whose receptors are members of the cytokine receptor superfamily, also known as type I cytokine receptors. These type I cytokines include the short-chain cytokines interleukin (IL)−2, IL-3, IL-4, IL-5, GM-CSF, IL-7, IL-9, IL-13, and IL-15 and the long-chain cytokines IL-6, IL-11, OSM, CNTF, CT-1, growth hormone, prolactin, erythropoietin, and thrombopoietin [3]. In mammals, the JAK family consists of four members: JAK1, JAK2, JAK 3, and Tyk2 [4]. Targeting JAK-associated pathways through the use of JAK inhibitors has rapidly entered the clinical arena in a wide range of disease states, including myeloproliferative neoplasms, rheumatoid arthritis, other immune-mediated joint diseases, and numerous inflammatory skin diseases and inflammatory bowel diseases [5]. At present, DM is mainly treated with medication and insulin replacement therapy to achieve good glycemic control. Conventional medications in diabetes treatment focus on insulin secretion and insulin sensitization, thus causing unwanted side effects and leading to both patient noncompliance and treatment failure [6]. With respect to insulin replacement therapy, insulin is an anabolic hormone that stimulates a large number of cellular responses. Simple insulin replacement therapy has difficulty in stably controlling the blood insulin concentration, thereby increasing the risk of obesity and cardiovascular disease [7]. In addition, neither of the two therapies above can improve the basic pathological mechanism of DM and the function of the pancreatic islets. Therefore, in addition to delaying the progression of diabetes by controlling blood sugar, improving pancreatic islet function through the preservation of β-cells is crucial in diabetes mellitus, which includes both type 1 and type 2 diabetes. β-cell dysfunction, reduced mass, and apoptosis are central to insufficient insulin secretion in both types [8]. The destruction of β-cells is generally thought to be T-cell-mediated [9]. Teplizumab is a humanized immunoglobulin G1 monoclonal antibody that binds with high affinity to the ε chain of CD3 (the signaling hexamer CD3 is a T-cell co-receptor indispensable for the activity of the T-cell receptor). Teplizumab is the only FDA-approved drug for the treatment of T1DM [10], and its mechanism of action is that teplizumab binds to CD3 to induce Tregs tolerance in autoimmune diseases [11], thereby reducing the destruction of β-cells. Therefore, the prevention or delay of β-cell destruction has an important role in the treatment of diabetes. Recently, several clinical studies have been reported the reversal of T1DM during JAK inhibitor treatment [12,13,14] (Table 1), and a small number of clinical studies have reported the benefit of JAK inhibitors in the treatment of T2DM [15, 16] (Table 1). Several basic studies to elaborate on its mechanism, mainly focusing on reducing the destruction of β cells and improving insulin resistance (Table 2). In the present study, we attempt to analyze the possible mechanisms (Fig. 1) behind such effects from basic experimental studies and discuss their potential role as a new therapeutic paradigm for diabetes.
T1DM
Data from animal models and in vitro studies
AZD1480 (Table 3) is an ATP-competitive inhibitor with specificity for JAK1 and JAK2 that targets the same active site as clinically approved drugs (ruxolitinib and baricitinib). Therefore, it can be used as a proof of principle for the use of JAK inhibitors in the treatment of diabetes. It was found that AZD1480, a JAK1/JAK2 inhibitor, blocked or delayed T-cell destruction of β-cells (reducing immune cell infiltration into pancreatic islets) by inhibiting β-cell MHC class I upregulation and reversed newly diagnosed diabetes in NOD mice (and reversed autoimmune insulinitis in NOD mice) [17]. Upadacitinib (ABT494) (Table 3) is also a JAK inhibitor that competes with adenosine triphosphate (ATP), which potently inhibits JAK1 and weakly inhibits the other isoforms JAK2, JAK3 and TYK2 [18]. Another study showed that upadacitinib (ABT494) could block β-cell MHC class 1 upregulation and inhibit the effects of common γ-chain cytokines on T-cell proliferation and effector T-cell and memory T-cell production, thereby reversing T1DM [19]. In addition, the lead compound JANEX-1 (Table 3) is a rationally designed potent and specific JAK3 inhibitor that does not affect the enzymatic activity of other protein tyrosine kinases [20, 21]. Targeting JAK3 with JANEX-1 in a NOD mouse model was able to block the development of insulin and diabetes in NOD-scid mice following splenocyte overgrafting in diabetic NOD mice. Most importantly, JANEX-1 was able to prevent the development of spontaneous autoimmune diabetes in a NOD type 1 diabetic mouse model [22]. Finally, deucravacitinib (Table 3) is an oral Tyk2 inhibitor that binds to the regulatory (JH2 pseudokinase) structural domain of Tyk2, is highly selective for Tyk2, and has little to no activity against JAK1-3 [23]. It has been shown that deucravacitinib can be used for the prevention or treatment of early T1DM by protecting β-cells from the deleterious effects of proinflammatory cytokines without affecting β-cell function or survival [24]. Thus, despite the paucity of basic experiments, the benefit of JAK inhibitors for T1DM can also be seen.
Clinical observations
In addition to basic experiments, evidence from clinical studies further elucidates the beneficial role of JAK inhibitors in T1DM. There is evidence that JAK inhibitors may have some therapeutic benefit in T1DM. First, a female patient with T1DM treated with baricitinib (Table 3) for rheumatoid arthritis (RA) experienced a rapid decrease in the daily insulin dose required (17 → 11 units), as well as a decrease in HbA1c levels (7.4% → 6.4%), and the patient's ability to secrete insulin was maintained for more than one year [13]. In addition, in a neonate with STAT3-GOF combined with IDDM, ruxolitinib (Table 3), as an off-label treatment, resulted in an immediate and significant decrease in insulin requirements (reduced from 6 to 0.6 IU/kg/day), and ruxolitinib insulin requirements remained low for more than 1 year of monotherapy (0.6 IU/kg/day); HbA1c that stabilized at 6.8% also confirmed continued diabetes control, and insulin autoantibody (IAA antibody) levels improved significantly with ruxolitinib [14]. Finally, in a 15-year-old adolescent with STAT1 gain of function combined with T1DM, 21 months after the diagnosis of T1DM and 12 months after the initiation of ruxolitinib (well beyond the "honeymoon" period of T1DM), exogenous insulin was discontinued, and the patient remained in a normoglycemic state. The patient also remained normal without insulin for more than a year [12]. Recently, Michaela Waibel et al. conducted a clinical study enrolling 91 patients with T1DM, in which patients diagnosed with T1DM within the 100 days prior to the start of the study were randomized to receive either baricitinib (4 mg once daily) or a matching placebo orally for 48 weeks, with a 2-h mixed-meal tolerance test performed at week 48. The ability of baricitinib to maintain β-cell function in patients with T1DM was evaluated by mean C-peptide levels determined from the area under the concentration‒time curve, as well as change from baseline in glycosylated hemoglobin levels, daily insulin doses, and measures of glycemic control assessed via continuous glucose monitoring. The study revealed that at 48 weeks, the median mean C-peptide level resulting from mixed dietary stimulation was greater in the baricitinib group than in the placebo group, the mean daily insulin dose was lower in the baricitinib group than in the placebo group, and the glycated hemoglobin levels were similar in the two trial groups. Estimates of mean C-peptide levels based on mixed dietary stimuli suggest that daily administration of baricitinib for more than 48 weeks appears to preserve β-cell function in patients with recent-onset T1DM [25]. The above data from different clinical observations support data from animal models of T1DM, suggesting that JAK inhibitors may all be useful in treating T1DM by preserving β-cell function and quality.
T2DM
Data from animal models and in vitro studies
Recently,Hauwa'u Yakubu Bako et al. conducted a basic study in which a rat model of T2DM was treated with different doses of tofacitinib (10, 20 mg/kg BW), aspirin (100, 200 mg/kg BW) and two doses of both drugs in combination for 9 weeks. The study revealed that T2DM resulted in a significant decrease in the serum insulin concentration and a significant reversal in all the tofacitinib-treated diabetic groups. Compared with those in the T2DM model group, the HOMA-IR values were significantly lower in all the diabetes groups treated with tofacitinib alone, the HOMA-β values were significantly greater, and the weekly blood glucose levels were lower [26]. Thus, although there are limited basic experiments, the concept that JAK inhibitors treat T2DM by improving insulin resistance emerges.
Clinical observations
In addition to basic experiments, evidence from clinical observations has further elucidated the therapeutic role of JAK inhibitors in T2DM. Claudia Di Muzio et al. conducted a clinical study enrolling 40 patients with RA with T2DM given tofacitinib (Table 3) at the appropriate dose according to the manufacturer's instructions. Glucose-lowering therapy was also maintained during the follow-up period, with the primary endpoint being the change in the 1998 update of the Homeostatic IR Model Assessment (HOMA2-IR) in patients with RA combined with T2DM, who were treated with tofacitinib for 6 months. The study revealed a gradual decrease in HOMA2-IR, i.e., a gradual reduction in the level of insulin resistance, and an increase in HOMA2-β, i.e., an improvement in insulin sensitivity, in patients treated with tofacitinib; a trend toward a decrease in glycosylated hemoglobin was also documented [15]. In addition, Cristina Martinez-Molina et al. conducted a clinical study that included 13 patients with T2DM combined with RA, six of whom were treated with tofacitinib and seven of whom were treated with baricitinib. The primary endpoint was glycated hemoglobin(HbA1c) after 6 months of treatment with JAK inhibitors. The study found a significant reduction in HbA1c in the treated baricitinib group, with 57.1% of patients treated with baricitinib having HbA1c values < 7%; in addition, 28.6% of patients treated with baricitinib required a reduction in the OAD dosee. However, there was no significant reduction in the tofacitinib group [16]. These studies support the findings of basic experiments suggesting that JAK inhibitors counteract T2DM by improving insulin resistance.
The respective possible mechanisms
JAK1
The JAK1 pathway is important for cytokines that share the common γ chain for type 1 cytokine receptors (e.g., IL-2, IL-6, and IL-21). JAK1 is also part of the canonical signaling pathway for type 1 and type 2 interferons [27]. The pathology of T1DM in NOD mice and humans is characterized by the presence of autoreactive cytotoxic CD8+T lymphocytes (CTLs) that mediate β-cell destruction. These cells acquire effector functions in pancreatic islets, a process that is dependent on the action of cytokines [28, 29]. Among these cytokines, IL-2, IL-7, and IL-15 have been shown to be three cytokines with indispensable roles in CD8+ T-cell memory generation and maintenance [30]. IL-2 and IL-21 are major cytokines required for CTL growth and differentiation [31, 32]. It has been shown that a selective inhibitor of JAK1 (ABT 317) blocks IL-2-, IL-7-, and IL-15-mediated phosphorylation of STAT5 and IL-21-mediated phosphorylation of STAT3, which inhibits the effects of common γ-chain cytokines on T-cell proliferation and effector and memory T-cell production, and thus reduces the destruction of β-cells by CTLs [19]. On the other hand, the overexpression of human leukocyte antigen class I (HLA-I) molecules on pancreatic β-cells is widely recognized as a hallmark feature of the pathogenesis of T1DM [33], this may increase the visibility of β-cells to self-reactive CD8+ T cells, thereby accelerating β-cell destruction. The presence of residual β-cells is critical for HLA-I overexpression in islet cells at all stages of the disease. The most likely drivers are interferons released by β-cells (type I or type III interferons) or produced from the influx of autoreactive immune cells (type II interferon). In both cases, the JAK/STAT pathway would be activated to induce downstream expression of interferon-stimulated genes [34]. This study also revealed that in NOD mice, a JAK1 selective inhibitor (ABT 317) reduced IFN-γ signaling in β-cells and prevented the upregulation of β-cell MHC class I, which reduced β-cell destruction [19].
JAK2
JAK2 plays an important role in signaling events that mediate innate and adaptive immunity. In the cytoplasm, the tyrosine protein kinase JAK2 plays a key role in signaling through association with type I receptors such as GHR, erythropoietin (EPOR), leptin (LEPR), prolactin (PRLR), and thrombopoietin (THPO) or type II receptors such as interferon-γ (IFN-γ), granulocyte/macrophage colony-stimulating factor (GM-CSF) and various interleukins (IL)-IL-3, IL-5, and IL-12 [35]. Basic experimental and clinical studies on the treatment of diabetes with a separate JAK2-selective inhibitor have not been reported. Sorafenib is an anticancer drug approved by the United States Food and Drug Administration (US FDA) for the treatment of unresectable hepatocellular carcinoma and advanced renal cell carcinoma. Sorafenib inhibits tumor growth and angiogenesis through targeting both the RAF/MEK/ERK pathway and receptor tyrosine kinases [36]. In vitro studies have shown that sorafenib indirectly inhibits the IL-12-induced phosphorylation of JAK2 and STAT4, thereby inhibiting Th1 cell differentiation. The Th1 cell population is a key mediator in the pathogenesis of T1DM. In the context of DM, β-cells are destroyed by cytotoxic T-lymphocytes (CTLs) and macrophages, both of which are regulated by helper T-lymphocytes (Th)1 cells [37]. In addition, since both JAK1 and JAK2 associate with IFN-γ and play key roles in IFN-γ signaling, they may have similar mechanisms. Interferon-γ-inducible protein 10 (CXCL10, also called IP-10) is considered an important chemokine in TIDM [38] and is required for the transportation of CTLs to sites of inflammation. It has been shown that AZD1480 (JAK1/JAK2 inhibitor) blocked IFN-γ-induced HLA-class I upregulation in β-cells and inhibited CXCL10 secretion in pancreatic islets; this resulted in a decrease in chemoattraction, migration, and accumulation of autoreactive CTLs in pancreatic islets, which led to attenuated interactions of autoreactive CTLs with AZD1480-treated β-cells and a reduction in the synaptic residence time, thereby reducing immune cell infiltration of pancreatic islets [17]. However, since JAK2 signaling is located downstream of EPO, GM-CSF, and TPO, JAK1-selective inhibitors have a better safety profile and lower risk of anemia than do JAK1/JAK2 inhibitors or JAK2-selective inhibitors [39].
JAK3
JAK3, a member of the JAK family, plays a crucial role in T-cell development and the homeostasis of the immune system homeostasis due to its association with the common γ chain (γc) of cytokine receptors [40] . PHA is a mitogenic agent obtained from the seed extract of cauliflower bean that can induce erythrocyte agglutination and stimulate mitosis in progressive lymphocytes in cell culture. It has been shown that JANEX-1 (JAK3-selective inhibitor) inhibits JAK3 dependent, IL-2-induced STAT5 tyrosine phosphorylation and alters the cytokine secretion pattern of T cells from diabetes-susceptible NOD mice with or without PHA stimulation. Furthermore, treatment with the JANEX-1 results in a significant increase in IL-10 secretion, a significant decrease in IL-2 secretion, and inhibition of the response of the PHA and proliferative responses to antigens (including the diabetic autoantigen GAD65) [22]. Therefore, there is a decrease in T-cell accumulation and a decrease in β-cell destruction. And it has been shown that exogenous IL-10 prevents insulitis or diabetes in NOD mice [41, 42]. Regulatory T cells are among the important factors involved in maintaining the immune tolerance of an organism, and regulate the immunity of the organism by inhibiting the activation and proliferation of potentially autoreactive T cells present in normal organisms via active regulation. It has been found that WHI-P131 (a JAK3-selective inhibitor) promotes TGF-β secretion by CD4 + T cells both in vivo and in vitro and significantly reduces IL-4, IFN-γ, and IL-2 production by CD4+ T cells. Induction of these cells may be a potential mechanism for the antidiabetic effects of JAK3-selective inhibitors in NOD mice [43]. In addition, tofacitinib is a JAK1/JAK3 inhibitor, and studies in rats with induced insulin resistance have shown that high levels of proinflammatory cytokines produced by the JAK-STAT and NF-κB signaling pathways lead to chronic inflammation and that high levels of proinflammatory cytokines increase the synthesis of cytokine inhibitory coactivators (SOCS) to inhibit insulin signaling, leading to insulin resistance. While tofacitinib can inhibit the production of high-level proinflammatory cytokines by inhibiting the JAK-STAT and NF-κB signaling pathways, it can also reduce SOCS and ultimately alleviate insulin resistance [26]; therefore, this may be a similar mechanism for inhibiting JAK or JAK3 to reduce insulin resistance.
TYK 2
Tyrosine kinase 2 (TYK2) is also an intracellular signaling tyrosine kinase that regulates signaling through a limited number of cytokines, including IL-12, IL-23, and type I IFNs (IFN-α and IFN-β), and plays a central role in the pathophysiology of IMID. Deucravacitinib is a novel orally-administered and selective TYK2 inhibitor with a unique binding mode to the regulatory (pseudokinase) structural domain rather than to the highly conserved ATP-binding site in the catalytic structural domain. TYK2 binds via a metastable mechanism that locks the regulatory structural domain in an inhibitory conformation with the catalytic structural domain, thereby trapping TYK2 in an inactive state and blocking receptor-mediated activation and downstream signaling [44, 45]. IFN-α is present in the pancreatic islets of T1DM patients [46] IFN-α plays a central role in the early stages of diabetes, and in human β-cells, IFN-α induces endoplasmic reticulum (ER) stress and prolonged the overexpression of HLA class I through the activation of the TYK2 signaling and STAT pathways. In addition, IFN-α synergizes with IL-1β to induce β-cell apoptosis [47, 48]. In mature stem cell pancreatic islets, the deletion or inhibition of TYK2 prevents IFN-α-induced antigen processing and presentation, including MHC class I and class II expression, which enhances their survival in response to CD8+ T-cell cytotoxicity [49]. Furthermore, in a study using deucravacitinib-mediated inhibition of TYK2 to analyze its effect on T1DM, it was found that deucravacitinib blocked the effect of IFN-α in a dose-dependent manner, i.e., it inhibited, among other factors, STAT1 and STAT2 activation and MHC class I overexpression, without affecting β-cell survival and function. In addition, decucravacitinib partially reduced apoptosis and inflammation in IFN-α + IL-1β or IFNγ + IL-1β-pretreated cells. However, it had no significant effect on ER stress [24].
Conclusion
In summary, case reports, clinical studies, and experimental animal and in vitro studies provide some evidence that JAK inhibitors improve the clinical performance of T1DM; in contrast, although there are two clinical studies in which JAK inhibitors improve the clinical performance of T2DM, few related experimental animal and in vitro studies exist. In this review, we found that JAK inhibitors act mainly against two major defects in diabetes, i.e., improvement of insulin resistance and reduction of β-cell dysfunction. One of the most common mechanisms is to block IFN and associated cytokine signaling by inhibiting JAK, which reduces T-cell activation, proliferation, accumulation, etc., and thus reduces β-cell destruction. In this work, we summarize previous related studies and highlight the possible mechanisms of the beneficial effects of JAK on DM; however, several limitations remain. First, the deep mechanism of action between related molecules and pathways is not fully understood. Second, the efficacy and safety of each molecule after inhibition have not been determined. Third, because the current research in this area is being explored, there are some limitations in this paper, as the number of relevant studies included is small. Therefore, owing to the potential benefits of JAK inhibitors on DM, we urgently need more basic experimental and clinical studies on JAK inhibitors and DM, including more clinical studies on JAK3 inhibitors and T1DM, experimental animal studies and then clinical studies on TYK2 inhibitors and T1DM, as well as studies on the molecular mechanism of action of JAK inhibitors on the benefits of T2DM. Currently, JAK inhibitors have been approved for use in many autoimmune diseases, and we believe that through more in-depth studies in the future, JAK inhibitors will inject newer and more powerful energy into the treatment and management of DM.
Data availability
No datasets were generated or analysed during the current study.
References
American Diabetes Association Professional Practice Committee. Glycemic targets: Standards of Medical Care in Diabetes—2022. Diabetes Care. 2022;45(Supplement_1):S83–96.
Philips RL, Wang Y, Cheon H, et al. The JAK-STAT pathway at 30: much learned, much more to do. Cell. 2022;185(21):3857–76.
Leonard WJ, O’Shea JJ. JAKS AND STATS: biological implications. Annu Rev Immunol. 1998;16(1):293–322.
Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J Cell Sci. 2004;117(8):1281–3.
McLornan DP, Pope JE, Gotlib J, et al. Current and future status of JAK inhibitors. Lancet. 2021;398(10302):803–16.
Tan SY, Mei Wong JL, Sim YJ, et al. Type 1 and 2 diabetes mellitus: a review on current treatment approach and gene therapy as potential intervention. Diabetes Metab Syndr. 2019;13(1):364–72.
Kolb H, Kempf K, Röhling M, et al. Insulin: too much of a good thing is bad. BMC Med. 2020;18(1):224.
Taneera J, Saber-Ayad MM. Preservation of β-cells as a therapeutic strategy for diabetes. Horm Metab Res. 2024;56(04):261–71.
Katsarou A, Gudbjörnsdottir S, Rawshani A, et al. Type 1 diabetes mellitus. Nat Rev Dis Primers. 2017;3(1):17016.
Keam SJ. Teplizumab: first approval. Drugs. 2023;83(5):439–45.
Mignogna C, Maddaloni E, D’Onofrio L, et al. Investigational therapies targeting CD3 for prevention and treatment of type 1 diabetes. Expert Opin Investig Drugs. 2021;30(12):1209–19.
Chaimowitz NS, Ebenezer SJ, Hanson IC, et al. STAT1 gain of function, type 1 diabetes, and reversal with JAK inhibition. N Engl J Med. 2020;383(15):1494–6.
Fujita Y, Nawata M, Nagayasu A, et al. Fifty-two-week results of clinical and imaging assessments of a patient with rheumatoid arthritis complicated by systemic sclerosis with interstitial pneumonia and type 1 diabetes despite multiple disease-modifying antirheumatic drug therapy that was successfully treated with baricitinib: a novel case report. Case Rep Rheumatol. 2019;2019:1–5.
Wegehaupt O, Muckenhaupt T, Johnson MB, et al. Ruxolitinib controls lymphoproliferation and diabetes in a STAT3-GOF patient. J Clin Immunol. 2020;40(8):1207–10.
Di Muzio C, Di Cola I, Shariat Panahi A, et al. The effects of suppressing inflammation by tofacitinib may simultaneously improve glycaemic parameters and inflammatory markers in rheumatoid arthritis patients with comorbid type 2 diabetes: a proof-of-concept, open, prospective, clinical study. Arthritis Res Ther. 2024;26(1):14.
Martinez-Molina C, Diaz-Torne C, Park HS, et al. Tofacitinib and baricitinib in type 2 diabetic patients with rheumatoid arthritis. Medicina. 2024;60(3):360.
Trivedi PM, Graham KL, Scott NA, et al. Repurposed JAK1/JAK2 inhibitor reverses established autoimmune insulitis in NOD mice. Diabetes. 2017;66:1650–60.
Mohamed M-EF, Bhatnagar S, Parmentier JM, et al. Upadacitinib: mechanism of action, clinical, and translational science. Clin Transl Sci. 2024;17(1):e13688.
Lee M-S, Wong FS, Kim S, et al. The JAK1 selective inhibitor ABT 317 blocks signaling through interferon-γ and common γ chain cytokine receptors to reverse autoimmune diabetes in NOD mice. Front Immunol. 2020;11:58843.
Uckun FM, Vassilev A, Dibirdik I, et al. Targeting JAK3 tyrosine kinase-linked signal transduction pathways with rationally-designed inhibitors. Anticancer Agents Med Chem. 2007;7(6):612–23.
Sudbeck EA, Liu X-P, Narla RK, et al. Structure-based design of specific inhibitors of janus kinase 3 as apoptosis-inducing antileukemic agents. Clin Cancer Res. 1999;5(6):1569–82.
Cetkovic-Cvrlje M, Dragt AL, Vassilev A, et al. Targeting JAK3 with JANEX-1 for prevention of autoimmune type 1 diabetes in NOD mice. Clin Immunol. 2003;106(3):213–25.
Rusiñol L, Puig L. Tyk2 targeting in immune-mediated inflammatory diseases. Int J Mol Sci. 2023;24(4):3391.
Dos Santos RS, Guzman-Llorens D, Perez-Serna AA, et al. Deucravacitinib, a tyrosine kinase 2 pseudokinase inhibitor, protects human EndoC-βH1 β-cells against proinflammatory insults. Front Immunol. 2023;14:1263926.
Waibel M, Wentworth JM, So M, et al. Baricitinib and β-cell function in patients with new-onset type 1 diabetes. N Engl J Med. 2023;389:2140–50.
Bako HY, Ibrahim MA, Isah MS, et al. Inhibition of JAK-STAT and NF-κB signalling systems could be a novel therapeutic target against insulin resistance and type 2 diabetes. Life Sci. 2019;239:117045.
Clarke B, Yates M, Adas M, et al. The safety of JAK-1 inhibitors. Rheumatology. 2021;60(Supplement_2):ii24–30.
Sutherland APR, Graham KL, Papadimitriou M, et al. IL-21 regulates SOCS1 expression in autoreactive CD8+ T cells but is not required for acquisition of CTL activity in the islets of non-obese diabetic mice. Sci Rep. 2019;9(1):15302.
Graham KL, Krishnamurthy B, Fynch S, et al. Autoreactive cytotoxic T lymphocytes acquire higher expression of cytotoxic effector markers in the islets of NOD mice after priming in pancreatic lymph nodes. Am J Pathol. 2011;178(6):2716–25.
Schluns KS, Lefrançois L. Cytokine control of memory T-cell development and survival. Nat Rev Immunol. 2003;3(4):269–79.
Sutherland APR, Joller N, Michaud M, et al. IL-21 promotes CD8+ CTL activity via the transcription factor T-bet. J Immunol. 2013;190(8):3977–84.
Erard F, Corthesy P, Nabholz M, et al. Interleukin 2 is both necessary and sufficient for the growth and differentiation of Lectin-stimulated cytolytic T lymphocyte precursors. J Immunol. 1985;134(3):1644–52.
Richardson SJ, Rodriguez-Calvo T, Gerling IC, et al. Islet cell hyperexpression of HLA class I antigens: a defining feature in type 1 diabetes. Diabetologia. 2016;59(11):2448–58.
Russell MA, Richardson SJ, Morgan NG. The role of the interferon/JAK-STAT axis in driving islet HLA-I hyperexpression in type 1 diabetes. Front Endocrinol. 2023;14:1270325.
Sopjani M, Morina R, Uka V, et al. JAK2-mediated intracellular signaling. Curr Mol Med. 2021;21(5):417–25.
Abdelgalil AA, Alkahtani HM, Al-Jenoobi FI. Sorafenib. Profiles Drug Subst Excip Relat Methodol. 2019;44:239–66.
Adorini L. Interleukin 12 and autoimmune diabetes. Nat Genet. 2001;27(2):131–2.
Ahmadi Z, Arababadi MK, Hassanshahi G. CXCL10 activities, biological structure, and source along with its significant role played in pathophysiology of type I diabetes mellitus. Inflammation. 2013;36(2):364–71.
Schwartz DM, Kanno Y, Villarino A, et al. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov. 2017;17(1):78.
Papageorgiou AC, Wikman LEK. Is JAK3 a new drug target for Immunomodulation-based therapies? Trends Pharmacol Sci. 2004;25(11):558–62.
Pennline KJ, Roque-Gaffney E, Monahan M. Recombinant human IL-10 prevents the onset of diabetes in the nonobese diabetic mouse. Clin Immunol Immunopathol. 1994;71(2):169–75.
Zheng XX, Steele AW, Hancock WW, et al. A noncytolytic IL-10/Fc fusion protein prevents diabetes, blocks autoimmunity, and promotes suppressor phenomena in NOD mice. J Immunol (Baltimore, Md: 1950). 1997;158(9):4507–13.
Cetkovic-Cvrlje M, Olson M, Ghate K. Targeting Janus tyrosine kinase 3 (JAK3) with an inhibitor induces secretion of TGF-β by CD4+ T cells. Cell Mol Immunol. 2012;9(4):350–60.
Chimalakonda A, Burke J, Cheng L, et al. Selectivity profile of the tyrosine kinase 2 inhibitor deucravacitinib compared with janus kinase 1/2/3 inhibitors. Dermatol Ther. 2021;11(5):1763–76.
Burke JR, Cheng L, Gillooly KM, et al. Autoimmune pathways in mice and humans are blocked by pharmacological stabilization of the TYK2 pseudokinase domain. Sci Transl Med. 2019;11(502):eaaw1736.
Foulis Alan K, Farquharson Maura A, Meager A. Immunoreactive α-interferon in insulin-secreting β cells in type 1 diabetes mellitus. Lancet. 1987;330(8573):1423–7.
Marroqui L, Dos Santos RS, Op De Beeck A, et al. Interferon-α mediates human beta cell HLA class I overexpression, endoplasmic reticulum stress and apoptosis, three hallmarks of early human type 1 diabetes. Diabetologia. 2017;60(4):656–67.
Colli ML, Ramos-RodrÃguez M, Nakayasu ES, et al. An integrated multi-omics approach identifies the landscape of interferon-α-mediated responses of human pancreatic beta cells. Nat Commun. 2020;11(1):2584.
Chandra V, Ibrahim H, Halliez C, et al. The type 1 diabetes gene TYK2 regulates β-cell development and its responses to interferon-α. Nat Commun. 2022;13(1):6363.
Dougados M, Van Der Heijde D, Chen Y-C. Baricitinib in patients with inadequate response or intolerance to conventional synthetic DMARDs: results from the RA-BUILD study. Ann Rheumatic Dis. 2017;76(1):88–95.
Phan K, Sebaratnam DF. JAK inhibitors for alopecia areata: a systematic review and meta-analysis. J Eur Acad Dermatol Venereol. 2019;33(5):850–6.
Kalil AC, Patterson TF, Mehta AK, et al. Baricitinib plus Remdesivir for hospitalized adults with Covid-19. N Engl J Med. 2021;384(9):795–807.
Freitas E, Guttman-Yassky E, Torres T. Baricitinib for the treatment of alopecia areata. Drugs. 2023;83(9):761–70.
Mascarenhas J, Hoffman R. Ruxolitinib: the first FDA approved therapy for the treatment of myelofibrosis. Clin Cancer Res. 2012;18(11):3008–14.
Haq M, Adnan G. Ruxolitinib. In: StatPearls [Internet]. StatPearls Publishing; 2023.
Modi B, Hernandez-Henderson M, Yang D, et al. Ruxolitinib as salvage therapy for chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2019;25(2):265–9.
Dhillon S. Tofacitinib: a review in rheumatoid arthritis. Drugs. 2017;77(18):1987–2001.
Rakieh C, Conaghan PG. Tofacitinib for treatment of rheumatoid arthritis. Adv Ther. 2013;30(8):713–26.
Berekmeri A, Mahmood F, Wittmann M, Helliwell P. Tofacitinib for the treatment of psoriasis and psoriatic arthritis. Expert Rev Clin Immunol. 2018;14(9):719–30.
Berekmeri A, Mahmood F, Wittmann M, et al. Tofacitinib for the treatment of psoriasis and psoriatic arthritis. Expert Rev Clin Immunol. 2018;14(9):719–30.
Sandborn WJ, Su C, Sands BE, et al. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2017;376(18):1723–36.
Jones NT, Keller CL, Abadie RB, et al. Safety and Effectiveness of tofacitinib in treating polyarticular course juvenile idiopathic arthritis. Cureus. 2023;15(11):e48258.
Acknowledgements
We thank B. Li for the guidance and revision of this manuscript.
Funding
This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
Author information
Authors and Affiliations
Contributions
Bo Li conceived the project, designed the structure and made critical revisions of the manuscript. Mengjun Zhou wrote the manuscript. Qi Shen prepared the figures and tables. All authors have read and approved the final version of the manuscript for submission.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
All authors approved the final manuscript and the submission to this journal.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Zhou, M., Shen, Q. & Li, B. JAK inhibitors: a new choice for diabetes mellitus?. Diabetol Metab Syndr 17, 33 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13098-025-01582-2
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13098-025-01582-2