Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 440
Filter
1.
J Enzyme Inhib Med Chem ; 39(1): 2409771, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39377432

ABSTRACT

A series of triazolopyridine-based dual JAK/HDAC inhibitors were rationally designed and synthesised by merging different pharmacophores into one molecule. All triazolopyridine derivatives exhibited potent inhibitory activities against both targets and the best compound 4-(((5-(benzo[d][1, 3]dioxol-5-yl)-[1, 2, 4]triazolo[1, 5-a]pyridin-2-yl)amino)methyl)-N-hydroxybenzamide (19) was dug out. 19 was proved to be a pan-HDAC and JAK1/2 dual inhibitor and displayed high cytotoxicity against two cancer cell lines MDA-MB-231 and RPMI-8226 with IC50 values in submicromolar range. Docking simulation revealed that 19 fitted well into the active sites of HDAC and JAK proteins. Moreover, 19 exhibited better metabolic stability in vitro than SAHA. Our study demonstrated that compound 19 was a promising candidate for further preclinical studies.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Histone Deacetylase Inhibitors , Histone Deacetylases , Pyridines , Triazoles , Humans , Cell Proliferation/drug effects , Structure-Activity Relationship , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Pyridines/pharmacology , Pyridines/chemistry , Pyridines/chemical synthesis , Molecular Structure , Triazoles/pharmacology , Triazoles/chemistry , Triazoles/chemical synthesis , Cell Line, Tumor , Histone Deacetylases/metabolism , Molecular Docking Simulation , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/chemical synthesis , Janus Kinase Inhibitors/chemistry , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism
2.
BMC Immunol ; 25(1): 63, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39354368

ABSTRACT

OBJECTIVES: Carcinoembryonic-antigen-related cell-adhesion molecule 1 (CEACAM1) is an adhesion molecule that acts as a coinhibitory receptor in the immune system. We previously demonstrated that CEACAM1 is predominantly expressed on peripheral blood neutrophils in patients with RA. The aim of the present study was to investigate the effects of Janus kinase inhibitors (JAKi) on cytokine-activated human neutrophils and CEACAM1 expression. METHODS: Peripheral blood neutrophils were obtained from healthy subjects. Isolated neutrophils were stimulated with tumor necrosis factor-alpha (TNF-α) or granulocyte-macrophage colony-stimulating factor (GM-CSF) in the presence or absence of JAKi. The expression of CEACAM1 in peripheral blood neutrophils was analyzed by flow cytometry. Protein phosphorylation of signal transducer and activator of transcription (STAT)1, STAT3, and STAT5 was assessed by western blot using phospho-specific antibodies. RESULTS: We found that TNF-α-induced CEACAM1 expression was marginally suppressed after pretreatment with pan-JAK inhibitor, tofacitinib. Moreover, TNF-α induced STAT1 and STAT3 phosphorylation at the late stimulation phase (4 to 16 h). The expressions of CEACAM1 on neutrophils were markedly up-regulated by GM-CSF not by interleukin (IL)-6 stimulation. All JAKi inhibited GM-CSF-induced CEACAM1 expressions on neutrophils, however, the inhibitory effects of baricitinib were larger compared to those of tofacitinib or filgotinib. Moreover, CEACAM1 was marginally upregulated in interferon (IFN)-γ stimulated neutrophils. Similarly, JAKi inhibited IFN-γ-induced CEACAM1 expressions on neutrophils. CONCLUSIONS: We demonstrated that JAKi prevent GM-CSF-induced CEACAM1 expression in neutrophils, and JAKi-induced inhibition depends on their selectivity against JAK isoforms. These findings suggest that JAKi can modulate the expression of CEACAM1 in cytokine-activated neutrophils, thereby limiting their activation.


Subject(s)
Antigens, CD , Cell Adhesion Molecules , Granulocyte-Macrophage Colony-Stimulating Factor , Janus Kinase Inhibitors , Neutrophils , Pyrimidines , Tumor Necrosis Factor-alpha , Humans , Neutrophils/metabolism , Neutrophils/immunology , Neutrophils/drug effects , Cell Adhesion Molecules/metabolism , Antigens, CD/metabolism , Pyrimidines/pharmacology , Janus Kinase Inhibitors/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Phosphorylation/drug effects , Piperidines/pharmacology , Pyrroles/pharmacology , Neutrophil Activation/drug effects , Cytokines/metabolism , Signal Transduction/drug effects
3.
Rinsho Ketsueki ; 65(8): 798-809, 2024.
Article in Japanese | MEDLINE | ID: mdl-39231711

ABSTRACT

Myelofibrosis should be diagnosed according to the WHO classification (2022, 5th Ed.) and International Consensus Conference 2022 criteria. Testing for driver mutations in the three genes JAK2, CALR, and MPL is recommended to ensure a definitive diagnosis. Ruxolitinib is the only JAK inhibitor currently approved in Japan, but momelotinib is under regulatory review. The MOMENTUM study showed similar spleen volume reduction at 24 weeks and MFSAF-TSS reduction as the COMFORT study of ruxolitinib. Momelotinib acts on ACVR1 and, therefore, improves anemia through suppression of hepcidin. Anemia and/or transfusion dependency are known to be associated with overall survival duration. Consequently, supportive care measures such as ESA and danazol in lieu of transfusion should be considered in addition to JAK inhibitor selection. Mean survival after discontinuation of JAK inhibitors is 11 to 14 months. Pacritinib (not approved in Japan) is suitable for MF patients with thrombocytopenia. JAK inhibitor selection and supportive care by ESA or danazol in lieu of transfusion should be considered. Many classes of drugs other than JAK inhibitors for myelofibrosis are under investigation.


Subject(s)
Benzamides , Janus Kinase Inhibitors , Nitriles , Primary Myelofibrosis , Pyrazoles , Pyrimidines , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/diagnosis , Humans , Pyrimidines/therapeutic use , Pyrazoles/therapeutic use , Pyrazoles/pharmacology , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Bridged-Ring Compounds
4.
Cell ; 187(18): 4814-4818, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39241741

ABSTRACT

In defying conventional views that dismissed itch as trivial, I persisted in studying basophils and ILC2s in human skin and atopic dermatitis. My research on JAK inhibitors for itch ultimately led to FDA-approved drugs. This is my story of disregarding categories and definitions-a story about an unconventional path in science that emphasizes innovation over conformity.


Subject(s)
Dermatitis, Atopic , Disease Models, Animal , Pruritus , Humans , Animals , Mice , Dermatitis, Atopic/pathology , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , History, 20th Century , History, 21st Century , Basophils/metabolism , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Skin/pathology , Skin/metabolism
5.
Inflammopharmacology ; 32(5): 3229-3246, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39217589

ABSTRACT

BACKGROUND: Rheumatoid arthritis (RA) is a chronic systemic autoimmune disorder characterized by persistent inflammation leading to progressively worse disability. Janus kinase (JAK) inhibitors and tumor necrosis factor (TNF) inhibitors are pivotal in RA treatment, yet their comparative efficacy remains underexplored. AIM: This study aimed to compare the efficacy and safety of JAK inhibitors and TNF inhibitors in treating RA using data from randomized controlled trials (RCTs). METHODS: A meta-analysis and outcomes analysis were based on results of the Health Assessment Questionnaire Disability Index (HAQ-DI), Clinical Disease Activity Index (CDAI), Visual Analogue Scale (VAS), and Patient Global Assessment Scale (PtGA) and other indices as incidences of venous thromboembolism (VTE) and malignancy. RESULTS: The JAK inhibitors caused a statistically significant improvement in the HAQ-DI score [MD = 0.08, 95% CI (0.03, 0.12), p = 0.0008] compared with the TNF inhibitors. However, no significant difference was observed between the two drug classes in the CDAI score [MD = - 2.03, 95% CI (- 9.27, 5.22), p = 0.58]. JAK inhibitors were associated with an increase in the VAS score [MD = 3.62, 95% CI (0.86, 6.38), p = 0.01], but there was no significant difference in the PtGA score [MD = 1.91, 95% CI (- 3.25, 7.08), p = 0.47]. CONCLUSION: JAK inhibitors demonstrated superior efficacy in improving the functional status and reducing the disease activity in RA patients compared with TNF inhibitors. Both drug classes exhibited comparable safety profiles for VTE and malignancies, though JAK inhibitors had a higher risk for thromboembolism.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Janus Kinase Inhibitors , Tumor Necrosis Factor Inhibitors , Humans , Antirheumatic Agents/adverse effects , Antirheumatic Agents/pharmacology , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Janus Kinase Inhibitors/adverse effects , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Randomized Controlled Trials as Topic , Treatment Outcome , Tumor Necrosis Factor Inhibitors/therapeutic use , Tumor Necrosis Factor Inhibitors/adverse effects , Tumor Necrosis Factor Inhibitors/pharmacology , Venous Thromboembolism/drug therapy
6.
Nat Rev Rheumatol ; 20(10): 649-665, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39251770

ABSTRACT

Cytokines function as communication tools of the immune system, serving critical functions in many biological responses and shaping the immune response. When cytokine production or their biological activity goes awry, the homeostatic balance of the immune response is altered, leading to the development of several pathologies such as autoimmune and inflammatory disorders. Cytokines bind to specific receptors on cells, triggering the activation of intracellular enzymes known as Janus kinases (JAKs). The JAK family comprises four members, JAK1, JAK2, JAK3 and tyrosine kinase 2, which are critical for intracellular cytokine signalling. Since the mid-2010s multiple JAK inhibitors have been approved for inflammatory and haematological indications. Currently, approved JAK inhibitors have demonstrated clinical efficacy; however, improved selectivity for specific JAKs is likely to enhance safety profiles, and different strategies have been used to accomplish enhanced JAK selectivity. In this update, we discuss the background of JAK inhibitors, current approved indications and adverse effects, along with new developments in this field. We address the issue of JAK selectivity and its relevance in terms of efficacy, and describe new modalities of JAK targeting, as well as new aspects of JAK inhibitor action.


Subject(s)
Janus Kinase Inhibitors , Humans , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Signal Transduction/drug effects , Cytokines/metabolism
7.
Medicina (Kaunas) ; 60(8)2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39202657

ABSTRACT

Background and Objectives: Atopic dermatitis is a chronic inflammatory skin disorder with a significant burden on patients' quality of life. This systematic review aims to evaluate the restoration of skin barrier abnormalities with interleukin-4/interleukin-13 (IL-4/IL-13) inhibitors and Janus kinase (JAK) inhibitors in atopic dermatitis. Materials and Methods: A comprehensive review of the literature was conducted, focusing on studies that assess the use of IL-4/IL-13 inhibitors and JAK inhibitors for atopic dermatitis. We identified eligible studies by searching Medline via PubMed with a special focus on their effect on the restoration of the epidermal barrier. Included studies evaluated the transepidermal water loss (TEWL), the reduction in epidermal thickness (ET), the improvement in ceramide synthesis, and the increase in stratum corneum hydration (SCH) with IL-4/IL-13 inhibitors and JAK inhibitors. The quality of included studies was assessed using the ROBINS-I and the RoB 2.0 tool for assessing the risk of bias. Results: Ten of the included studies concern dupilumab, while two concern JAK inhibitors. Ten were observational studies and two were randomized controlled trials (RCTs). The total number of included participants was 378 concerning dupilumab and 38 concerning JAK inhibitors. Five studies did not include any comparison group, three included healthy volunteers, two were conducted versus placebo, and two compared dupilumab with other treatments. The follow-up period ranged between 29 days and 32 weeks. The results demonstrated a significant decrease in transepidermal water loss (TEWL) and an increase in SCH on eczematous lesions for patients with sustained response to dupilumab treatment and observed improvements in ET and filaggrin (FLG) staining, which further support the efficacy of JAK inhibitors in enhancing skin barrier function. Conclusions: This review underscores the efficacy of IL-4/IL-13 inhibitors in improving skin barrier function. However, the limited number of studies focusing on JAK inhibitors and the overall lack of RCTs highlight the need for further research to establish the definitive role of IL-4/IL-13 inhibitors and JAK inhibitors in the restoration of the skin barrier.


Subject(s)
Dermatitis, Atopic , Interleukin-13 , Interleukin-4 , Janus Kinase Inhibitors , Dermatitis, Atopic/drug therapy , Humans , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Interleukin-4/analysis , Antibodies, Monoclonal, Humanized/therapeutic use , Water Loss, Insensible/drug effects , Filaggrin Proteins
8.
Arch Dermatol Res ; 316(8): 566, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39180702

ABSTRACT

Rosacea is a chronic inflammatory skin disease characterized by facial erythema and telangiectasia. Despite ongoing research, the pathogenesis of rosacea remains incompletely understood, and current therapies are not entirely satisfactory. The JAK/STAT signaling pathway plays an essential role in immunoregulation, inflammation, and neurovascular regulation. Inhibition of the JAK/STAT pathway appears to hold promise as a potential therapy for rosacea. This study aimed to investigate the effects of the JAK inhibitor tofacitinib on rosacea and to preliminarily explore its therapeutic mechanism. To this end, a rosacea-like mouse model was induced using LL37 and treated with a 2% tofacitinib emulsion. The results demonstrated that topical application of tofacitinib significantly ameliorated rosacea-like phenotype, reduced the infiltration of CD4+ T cells and mast cells, and suppressed dermal angiogenesis. RT-qPCR analysis revealed a reduction in mRNA expression levels of STAT1, STAT4, and STAT5a in skin lesions following topical tofacitinib treatment. Additionally, three patients diagnosed with erythematotelangiectatic rosacea (ETR) were included in the study and treated with oral tofacitinib, leading to a significant improvement in erythema and flushing symptoms. These findings collectively suggest that tofacitinib alleviates LL37-induced rosacea-like skin inflammation in mice and rosacea skin lesions by inhibiting the JAK/STAT signaling pathway.


Subject(s)
Piperidines , Pyrimidines , Rosacea , Signal Transduction , Adult , Animals , Female , Humans , Male , Mice , Middle Aged , Administration, Oral , Administration, Topical , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/administration & dosage , Janus Kinases/metabolism , Janus Kinases/antagonists & inhibitors , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Piperidines/pharmacology , Piperidines/therapeutic use , Piperidines/administration & dosage , Pyrimidines/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Pyrroles/pharmacology , Pyrroles/administration & dosage , Rosacea/drug therapy , Signal Transduction/drug effects , Skin/pathology , Skin/drug effects , Skin/metabolism , STAT Transcription Factors/metabolism , STAT Transcription Factors/antagonists & inhibitors , STAT1 Transcription Factor/metabolism
9.
Bioorg Chem ; 152: 107696, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39167870

ABSTRACT

The JAK-STAT signalling pathway is primarily involved in cytokine signalling and induces various factors namely, erythropoietin, thrombopoietin, interferons, interleukins, and granulocyte colony-stimulating factors. These factors tremendously influenced understanding human health and illness, specifically cancer. Inhibiting the JAK/STAT pathway offers enormous therapeutic promises against cancer. Many JAK inhibitors are now being studied due to their efficacy in various cancer treatments. Further, the Nitrogen-heterocyclic (N-heterocyclic) scaffold has always shown to be a powerful tool for designing and discovering synthetic compounds with diverse pharmacological characteristics. The review focuses on several FDA-approved JAK inhibitors and their systematic categorization. The medicinal chemistry perspective is highlighted and classified review on the basis of N-heterocyclic molecules. Several examples of designing strategies of N-heterocyclic rings including pyrrolo-azepine, purine, 1H-pyrazolo[3,4-d]pyrimidine, 1H-pyrrolo[2,3-b]pyridine, pyrazole, thieno[3,2-d] pyrimidine, and, pyrimidine-based derivatives and their structure-activity relationships (SAR) are discussed. Among the various N-heterocyclic-based JAK inhibitors pyrimidine-containing compound 1 exhibited excellent inhibition activity against JAK2WT and mutated-JAK2V617F with IC50 of 2.01 and 18.84 nM respectively. Amino pyrimidine-containing compound 6 and thiopheno[3,2-d]pyrimidine-containing compound 13 expressed admirable JAK3 inhibition activity with IC50 of 1.7 nM and 1.38 nM respectively. Our review will support the medicinal chemists in refining and directing the development of novel N-heterocyclic-based JAK inhibitors.


Subject(s)
Antineoplastic Agents , Heterocyclic Compounds , Janus Kinase Inhibitors , Animals , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/chemical synthesis , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Molecular Structure , Neoplasms/drug therapy , Nitrogen/chemistry , Structure-Activity Relationship , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology
10.
Cytokine Growth Factor Rev ; 79: 1-15, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39179485

ABSTRACT

Inflammatory bowel disease (IBD) encompasses a group of non-specific chronic intestinal inflammatory conditions of unclear etiology. The current treatment and long-term management primarily involve biologics. Nevertheless, some patients experience treatment failure or intolerance to biologics [1], making these patients a primary focus of IBD research. The Janus kinase (JAK)-Signal Transducers and Activator of Transcription (STAT) signal transduction pathway is crucial to the regulation of immune and inflammatory responses [2], and plays an important role in the pathogenesis of IBD. JAK inhibitors alleviate IBD by suppressing the transmission of JAK-STAT signaling pathway. As the first small-molecule oral inhibitor for IBD, JAK inhibitors greatly improved the treatment of IBD and have demonstrated significant efficacy, with tofacitinib and upadacitinib being approved for the treatment of ulcerative colitis (UC) [3]. JAK inhibitors can effectively alleviate intestinal inflammation in IBD patients who have failed to receive biologics, which may bring new treatment opportunities for refractory IBD patients. This review aims to elucidate the crucial roles of JAK-STAT signal transduction pathway in IBD pathogenesis, examine its role in various cell types within IBD, and explore the research progress of JAK inhibitors as therapeutic agents, paving the road for new IBD treatment strategies.


Subject(s)
Inflammatory Bowel Diseases , Janus Kinase Inhibitors , Janus Kinases , STAT Transcription Factors , Signal Transduction , Humans , Signal Transduction/drug effects , STAT Transcription Factors/metabolism , STAT Transcription Factors/antagonists & inhibitors , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Inflammatory Bowel Diseases/drug therapy , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Animals , Piperidines/therapeutic use , Piperidines/pharmacology , Pyrimidines/therapeutic use , Pyrimidines/pharmacology , Colitis, Ulcerative/drug therapy , Heterocyclic Compounds, 3-Ring
11.
Life Sci Alliance ; 7(11)2024 Nov.
Article in English | MEDLINE | ID: mdl-39122555

ABSTRACT

Reduction in muscle contractile force associated with many clinical conditions incurs serious morbidity and increased mortality. Here, we report the first evidence that JAK inhibition impacts contractile force in normal human muscle. Muscle biopsies were taken from patients who were randomized to receive tofacitinib (n = 16) or placebo (n = 17) for 48 h. Single-fiber contractile force and molecular studies were carried out. The contractile force of individual diaphragm myofibers pooled from the tofacitinib group (n = 248 fibers) was significantly higher than those from the placebo group (n = 238 fibers), with a 15.7% greater mean maximum specific force (P = 0.0016). Tofacitinib treatment similarly increased fiber force in the serratus anterior muscle. The increased force was associated with reduced muscle protein oxidation and FoxO-ubiquitination-proteasome signaling, and increased levels of smooth muscle MYLK. Inhibition of MYLK attenuated the tofacitinib-dependent increase in fiber force. These data demonstrate that tofacitinib increases the contractile force of skeletal muscle and offers several underlying mechanisms. Inhibition of the JAK-STAT pathway is thus a potential new therapy for the muscle dysfunction that occurs in many clinical conditions.


Subject(s)
Janus Kinase Inhibitors , Muscle Contraction , Muscle, Skeletal , Piperidines , Pyrimidines , Humans , Piperidines/pharmacology , Pyrimidines/pharmacology , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Janus Kinase Inhibitors/pharmacology , Male , Pyrroles/pharmacology , Female , Adult , Signal Transduction/drug effects , Middle Aged , Janus Kinases/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism
12.
Drug Res (Stuttg) ; 74(8): 394-404, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39134030

ABSTRACT

Sepsis, a life-threatening condition triggered by an uncontrolled response to infection, results in a systemic inflammatory response syndrome (SIRS) and the failure of multiple organs leading to multiple organ dysfunction (MODS). In the present study, we investigated the therapeutic potential of tofacitinib (TOFA), an FDA-approved inhibitor of JAK1 and JAK3 against sepsis, using a mouse model induced by cecal ligation puncture (CLP). Swiss albino mice were employed to replicate the CLP-induced sepsis model and were randomly divided into four groups: control, CLP, 150 mg/kg TOFA, and 300 mg/kg TOFA. Six hours after the last TOFA dose, we collected blood and tissue samples from the liver, lungs, kidneys, and spleen for histological analysis. Blood samples were used to assess granulocyte and lymphocyte percentages. Throughout the experiment, we monitored body weight and short-term survival. Our comparative histological analysis revealed that 150 mg/kg TOFA had a protective effect against multiple organ damage. Conversely, the study highlighted the harmful effects of 300 mg/kg TOFA, primarily due to liver and renal toxicity within this group. In summary, our findings demonstrate that tofacitinib at an optimal dose of 150 mg/kg showed promise as a potential therapeutic intervention for sepsis-induced multiple organ failure. However, caution is warranted when considering higher dosages.


Subject(s)
Disease Models, Animal , Janus Kinase Inhibitors , Multiple Organ Failure , Piperidines , Pyrimidines , Sepsis , Piperidines/pharmacology , Piperidines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Animals , Multiple Organ Failure/drug therapy , Multiple Organ Failure/etiology , Multiple Organ Failure/prevention & control , Sepsis/drug therapy , Sepsis/complications , Mice , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Male , Pyrroles/pharmacology , Pyrroles/therapeutic use , Kidney/drug effects , Kidney/pathology , Dose-Response Relationship, Drug , Liver/drug effects , Liver/pathology
13.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125897

ABSTRACT

Rheumatoid arthritis (RA) is a highly prevalent autoimmune disorder. The pathogenesis of the disease is complex and involves various cellular populations, including fibroblast-like synoviocytes, macrophages, and T cells, among others. Identification of signalling pathways and molecules that actively contribute to the development of the disease is crucial to understanding the mechanisms involved in the chronic inflammatory environment present in affected joints. Recent studies have demonstrated that the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway regulates the behaviour of immune cells and contributes to the progression of RA. Several JAK inhibitors, such as tofacitinib, baricitinib, upadacitinib, and filgocitinib, have been developed, and their efficacy and safety in patients with RA have been comprehensively investigated in a number of clinical trials. Consequently, JAK inhibitors have been approved and registered as a treatment for patients with RA. In this review, we discuss the involvement of JAK/STAT signalling in the pathogenesis of RA and summarise the potential beneficial effects of JAK inhibitors in cells implicated in the pathogenesis of the disease. Moreover, we present the most important phase 3 clinical trials that evaluated the use of these agents in patients.


Subject(s)
Arthritis, Rheumatoid , Janus Kinase Inhibitors , Janus Kinases , Humans , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Janus Kinases/metabolism , Janus Kinases/antagonists & inhibitors , Signal Transduction/drug effects , STAT Transcription Factors/metabolism , Animals , Antirheumatic Agents/therapeutic use , Antirheumatic Agents/pharmacology
14.
Expert Opin Pharmacother ; 25(12): 1625-1645, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39136388

ABSTRACT

INTRODUCTION: Adult dermatomyositis (DM) and juvenile dermatomyositis (JDM) are rare autoimmune diseases with characteristic skin rashes, weakness, and other systemic features. Upregulated interferon signaling has been consistently described in both adult and juvenile DM which makes janus kinase inhibitors (jakinibs) an attractive therapeutic agent that has a targeted mechanism of action. AREAS COVERED: Herein is a review of the growing literature of jakinib use in adult and juvenile DM, including reports on specific disease features and safety of jakinibs in this population and a comparison between adult and juvenile DM. We performed a literature review using PubMed including all English-language publications before 1 February 2024 and abstracts from key recent rheumatology conferences. EXPERT OPINION: Jakinibs are an exciting and promising treatment in both adult and juvenile DM. Current Phase 2 and 3 randomized placebo-controlled trials of jakinibs in both adult and JDM will provide significant insights into the efficacy of this class of medication as a potentially more mechanistically targeted treatment of both skin and muscle disease. In fact, these results will likely inform the treatment paradigm of dermatomyositis in that it may even be considered as first or second line. The next five years in the therapeutic landscape of both juvenile and adult DM is an exciting time for both patients and medical providers.


Subject(s)
Dermatomyositis , Janus Kinase Inhibitors , Humans , Dermatomyositis/drug therapy , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/adverse effects , Child , Adult , Randomized Controlled Trials as Topic , Age Factors
15.
Front Immunol ; 15: 1443704, 2024.
Article in English | MEDLINE | ID: mdl-39188724

ABSTRACT

Introduction: The Janus kinase (JAK) family includes four cytoplasmic tyrosine kinases (JAK1, JAK2, JAK3, and TYK2) constitutively bound to several cytokine receptors. JAKs phosphorylate downstream signal transducers and activators of transcription (STAT). JAK-STAT5 pathways play a critical role in basophil and mast cell activation. Previous studies have demonstrated that inhibitors of JAK-STAT pathway blocked the activation of mast cells and basophils. Methods: In this study, we investigated the in vitro effects of ruxolitinib, a JAK1/2 inhibitor, on IgE- and IL-3-mediated release of mediators from human basophils, as well as substance P-induced mediator release from skin mast cells (HSMCs). Results: Ruxolitinib concentration-dependently inhibited IgE-mediated release of preformed (histamine) and de novo synthesized mediators (leukotriene C4) from human basophils. Ruxolitinib also inhibited anti-IgE- and IL-3-mediated cytokine (IL-4 and IL-13) release from basophils, as well as the secretion of preformed mediators (histamine, tryptase, and chymase) from substance P-activated HSMCs. Discussion: These results indicate that ruxolitinib, inhibiting the release of several mediators from human basophils and mast cells, is a potential candidate for the treatment of inflammatory disorders.


Subject(s)
Basophils , Janus Kinase 1 , Janus Kinase 2 , Mast Cells , Nitriles , Pyrazoles , Pyrimidines , Humans , Basophils/drug effects , Basophils/immunology , Basophils/metabolism , Pyrimidines/pharmacology , Nitriles/pharmacology , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Pyrazoles/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Janus Kinase 2/antagonists & inhibitors , Cells, Cultured , Janus Kinase Inhibitors/pharmacology , Cytokines/metabolism , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Protein Kinase Inhibitors/pharmacology
16.
Dermatologie (Heidelb) ; 75(10): 781-790, 2024 Oct.
Article in German | MEDLINE | ID: mdl-39212722

ABSTRACT

Immune factors such as interferon­É£ and interleukin 4 belong to the group of cytokines that are dependent on type I/II receptors for their signal transmission. Upon activation, these receptors transmit their signal to the cell nucleus and, thus, modulate gene transcription via a signaling cascade consisting of Janus kinases (JAK). This family of four kinases (JAK 1, JAK 2, JAK 3, and tyrosine kinase 2 (TYK2)) subsequently activate members of the signal transducer and activator of transcription (STAT). This finding turned the JAK/STAT signaling pathway into a pharmacological target for the treatment of inflammatory diseases in which cytokines using type I/II receptors play a pathogenic role. In 2018, the European Medicines Agency (EMA) approved tofacitinib for the treatment of psoriatic arthritis. This was the first approval of a JAK/STAT pathway inhibitor for patients treated by dermatologists and rheumatologists. Since then, several new JAK inhibitors have been approved for dermatologic diseases such as atopic dermatitis, alopecia areata, vitiligo, and plaque-type psoriasis. In addition, JAK inhibitors are being investigated for the treatment of many other skin diseases. Thus, systemic JAK inhibitors complete the spectrum of immunotherapeutics with a broader immunological approach compared to monoclonal antibodies. The low molecular weight of JAK inhibitors enables the preparation of these drugs for both systemic and topical administration. Their utilization could represent a valuable alternative to topical steroids. The safety profile of JAK inhibitors must be taken into account. Possible long-term effects may become apparent in the next few years. This article describes both approved JAK inhibitors and relevant new JAK inhibitors that are promising candidates for approval as therapeutics in dermatology.


Subject(s)
Janus Kinase Inhibitors , Skin Diseases , Humans , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Skin Diseases/drug therapy , Pyrimidines/therapeutic use , Pyrimidines/pharmacology , Piperidines/therapeutic use , Piperidines/pharmacology , Pyrroles/therapeutic use , Pyrroles/pharmacology , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism
17.
Korean J Gastroenterol ; 84(2): 51-64, 2024 Aug 25.
Article in Korean | MEDLINE | ID: mdl-39176461

ABSTRACT

Small molecules, including Janus kinase (JAK) inhibitors and sphingosine-1-phosphate receptor modulators (S1PRMs), are promising new treatments for inflammatory bowel disease (IBD). Small molecules exhibit more predictable pharmacokinetics than biologics, are less likely to induce immune responses, and can be administered orally. JAK inhibitors function by blocking the activity of JAK enzymes, which prevents the subsequent phosphorylation and activation of signal transducer and activator of transcription (STAT) proteins. Tofacitinib and filgotinib are approved for treating ulcerative colitis (UC), while upadacitinib is approved for UC and Crohn's disease. Nevertheless, JAK inhibitors can increase the risk of herpes zoster, cancer, major adverse cardiovascular events, and venous thromboembolism. S1PRMs bind to S1PRs, particularly S1PR1, on lymphocytes. This interaction inhibits lymphocytes from exiting the lymph nodes and migrating to the gut, thereby reducing inflammation and the immune response in the intestinal mucosa. Ozanimod and etrasimod are S1PRMs approved for the treatment of UC, but they can cause side effects such as bradycardia, conduction disorder, and macular edema. Overall, JAK inhibitors and S1PRMs offer significant benefits in managing IBD, although their potential side effects require careful monitoring.


Subject(s)
Inflammatory Bowel Diseases , Janus Kinase Inhibitors , Sphingosine 1 Phosphate Receptor Modulators , Humans , Indans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Oxadiazoles , Pyridines , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/antagonists & inhibitors , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , Sphingosine 1 Phosphate Receptor Modulators/therapeutic use , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Triazoles
18.
Drugs ; 84(8): 877-894, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38949688

ABSTRACT

Rheumatoid arthritis (RA) is a chronic immune-mediated inflammatory disease which can induce progressive disability if not properly treated early. Over the last 20 years, the improvement of knowledge on the pathogenesis of the disease has made available several drugs targeting key elements of the pathogenetic process, which now represent the preferred treatment option after the failure of first-line therapy with conventional drugs such as methotrexate (MTX). To this category of targeted drugs belong anti-cytokine or cell-targeted biological agents and more recently also Janus kinase inhibitors (JAKis). In the absence to date of specific biomarkers to guide the therapeutic choice in the context of true precision medicine, the choice of the first targeted drug after MTX failure is guided by treatment cost (especially after the marketing of biosimilar products) and by the clinical characteristics of the patient (age, sex, comorbidities and compliance) and the disease (presence or absence of autoantibodies and systemic or extra-articular manifestations), which may influence the efficacy and safety profile of the available products. This viewpoint focuses on the decision-making process underlying the personalized approach to RA therapy and will analyse the evidence in the literature supporting the choice of individual products and in particular the differential choice between biological drugs and JAKis.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Biological Products , Janus Kinase Inhibitors , Humans , Arthritis, Rheumatoid/drug therapy , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Antirheumatic Agents/therapeutic use , Antirheumatic Agents/pharmacology , Biological Products/therapeutic use , Biological Products/pharmacology , Methotrexate/therapeutic use , Methotrexate/pharmacology , Precision Medicine
20.
Hepatol Commun ; 8(8)2024 08 01.
Article in English | MEDLINE | ID: mdl-39082962

ABSTRACT

BACKGROUND: HBV infects ~257 million people and can cause hepatocellular carcinoma. Since current drugs are not curative, novel therapies are needed. HBV infects chimpanzee and human livers. However, chimpanzee studies are severely restricted and cost-prohibitive, while transgenic/chimeric mouse models that circumvent the species barrier lack natural HBV infection and disease progression. Thus, in vitro human models of HBV infection are useful in addressing the above limitations. Induced pluripotent stem cell-derived hepatocyte-like cells mitigate the supply limitations of primary human hepatocytes and the abnormal proliferation/functions of hepatoma cell lines. However, variable infection across donors, deficient drug metabolism capacity, and/or low throughput limit iHep utility for drug development. METHODS: We developed an optimal pipeline using combinations of small molecules, Janus kinase inhibitor, and 3',5'-cAMP to infect iHep-containing micropatterned co-cultures (iMPCC) with stromal fibroblasts within 96-well plates with serum-derived HBV and cell culture-derived HBV (cHBV). Polyethylene glycol was necessary for cell-derived HBV but not for serum-derived HBV infection. RESULTS: Unlike iHep monocultures, iMPCCs created from 3 iHep donors could sustain HBV infection for 2+ weeks. Infected iMPCCs maintained high levels of differentiated functions, including drug metabolism capacity. HBV antigen secretion and gene expression patterns in infected iMPCCs in pathways such as fatty acid metabolism and cholesterol biosynthesis were comparable to primary human hepatocyte-MPCCs. Furthermore, iMPCCs could help elucidate the effects of interferons and direct-acting antiviral drugs on the HBV lifecycle and any hepatotoxicity; iMPCC response to compounds was similar to primary human hepatocyte-MPCCs. CONCLUSIONS: The iMPCC platform can enable the development of safe and efficacious drugs against HBV and ultimately help elucidate genotype-phenotype relationships in HBV pathogenesis.


Subject(s)
Hepatitis B virus , Hepatocytes , Induced Pluripotent Stem Cells , Humans , Hepatocytes/virology , Induced Pluripotent Stem Cells/virology , Induced Pluripotent Stem Cells/metabolism , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Hepatitis B virus/drug effects , Hepatitis B/virology , Hepatitis B/drug therapy , Coculture Techniques , Janus Kinase Inhibitors/pharmacology , Antiviral Agents/pharmacology , Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL