Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Am Chem Soc ; 146(19): 13317-13325, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38700457

ABSTRACT

We describe the synthesis and biological testing of ruthenium-bipyridine ruxolitinib (RuBiRuxo), a photoreleasable form of ruxolitinib, a JAK inhibitor used as an antitumoral agent in cutaneous T-cell lymphomas (CTCL). This novel caged compound is synthesized efficiently, is stable in aqueous solution at room temperature, and is photoreleased rapidly by visible light. Irradiation of RuBiRuxo reduces cell proliferation and induces apoptosis in a light- and time-dependent manner in a CTCL cell line. This effect is specific and is mediated by a decreased phosphorylation of STAT proteins. Our results demonstrate the potential of ruthenium-based photocompounds and light-based therapeutic approaches for the potential treatment of cutaneous lymphomas and other pathologies.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Proliferation , Nitriles , Pyrazoles , Pyrimidines , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Nitriles/chemistry , Nitriles/pharmacology , Nitriles/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/chemical synthesis , Apoptosis/drug effects , Pyrazoles/pharmacology , Pyrazoles/chemistry , Pyrazoles/chemical synthesis , Cell Line, Tumor , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/chemical synthesis , Ruthenium/chemistry , Ruthenium/pharmacology , Light , Molecular Structure , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism
2.
ACS Appl Bio Mater ; 7(5): 3179-3189, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38581305

ABSTRACT

Ruxolitinib (RXL) is a Janus kinase inhibitor used for treating intermediate- or high-risk myelofibrosis. This study presents an electrode modified with electrochemically polymerized taurine on a carbon paste electrode via cyclic voltammetry (CV). The surface characterization of the poly(taurine)-CP electrode was evaluated by using electrochemical (electrochemical impedance spectroscopy─EIS, CV), morphological (scanning electron microscope─SEM), and spectroscopic (Fourier-transform infrared spectroscopy─FT-IR) techniques. Under optimized conditions, RXL exhibited good linearity within the 0.01-1.0 µM concentration range, with a limit of detection (LOD) of 0.005 µM. The proposed electrochemical sensor demonstrated excellent selectivity, accuracy, precision, and repeatability. Furthermore, it effectively detected RXL in human urine and pharmaceutical samples.


Subject(s)
Carbon , Electrochemical Techniques , Electrodes , Materials Testing , Nitriles , Pyrazoles , Pyrimidines , Taurine , Nitriles/chemistry , Nitriles/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Carbon/chemistry , Humans , Pyrazoles/chemistry , Pyrazoles/pharmacology , Taurine/chemistry , Taurine/analogs & derivatives , Taurine/pharmacology , Particle Size , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/pharmacology , Polymerization , Molecular Structure
3.
Phys Chem Chem Phys ; 26(17): 13420-13431, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38647171

ABSTRACT

Autoimmune inflammatory diseases, such as rheumatoid arthritis (RA) and ulcerative colitis, are associated with an uncontrolled production of cytokines leading to the pronounced inflammatory response of these disorders. Their therapy is currently focused on the inhibition of cytokine receptors, such as the Janus kinase (JAK) protein family. Tofacitinib and peficitinib are JAK inhibitors that have been recently approved to treat rheumatoid arthritis. In this study, an in-depth analysis was carried out through quantum biochemistry to understand the interactions involved in the complexes formed by JAK1 and tofacitinib or peficitinib. Computational analyses provided new insights into the binding mechanisms between tofacitinib or peficitinib and JAK1. The essential amino acid residues that support the complex are also identified and reported. Additionally, we report new interactions, such as van der Waals; hydrogen bonds; and alkyl, pi-alkyl, and pi-sulfur forces, that stabilize the complexes. The computational results revealed that peficitinib presents a similar affinity to JAK1 compared to tofacitinib based on their interaction energies.


Subject(s)
Adamantane/analogs & derivatives , Janus Kinase 1 , Niacinamide , Niacinamide/analogs & derivatives , Piperidines , Pyrimidines , Pyrimidines/chemistry , Pyrimidines/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Piperidines/therapeutic use , Niacinamide/chemistry , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 1/chemistry , Humans , Quantum Theory , Autoimmune Diseases/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Hydrogen Bonding , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Adamantane/chemistry , Pyrroles/chemistry , Pyrroles/pharmacology , Molecular Docking Simulation
4.
Adv Healthc Mater ; 13(12): e2303256, 2024 May.
Article in English | MEDLINE | ID: mdl-38207170

ABSTRACT

Janus kinase (JAK) inhibitors are approved for many dermatologic disorders, but their use is limited by systemic toxicities including serious cardiovascular events and malignancy. To overcome these limitations, injectable hydrogels are engineered for the local and sustained delivery of baricitinib, a representative JAK inhibitor. Hydrogels are formed via disulfide crosslinking of thiolated hyaluronic acid macromers. Dynamic thioimidate bonds are introduced between the thiolated hyaluronic acid and nitrile-containing baricitinib for drug tethering, which is confirmed with 1H and 13C nuclear magnetic resonance (NMR). Release of baricitinib is tunable over six weeks in vitro and active in inhibiting JAK signaling in a cell line containing a luciferase reporter reflecting interferon signaling. For in vivo activity, baricitinib hydrogels or controls are injected intradermally into an imiquimod-induced mouse model of psoriasis. Imiquimod increases epidermal thickness in mice, which is unaffected when treated with baricitinib or hydrogel alone. Treatment with baricitinib hydrogels suppresses the increased epidermal thickness in mice treated with imiquimod, suggesting that the sustained and local release of baricitinib is important for a therapeutic outcome. This study is the first to utilize a thioimidate chemistry to deliver JAK inhibitors to the skin through injectable hydrogels, which has translational potential for treating inflammatory disorders.


Subject(s)
Azetidines , Hydrogels , Purines , Pyrazoles , Skin , Sulfonamides , Animals , Hydrogels/chemistry , Purines/chemistry , Purines/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Sulfonamides/administration & dosage , Mice , Pyrazoles/chemistry , Pyrazoles/pharmacology , Azetidines/chemistry , Azetidines/pharmacology , Skin/drug effects , Skin/metabolism , Skin/pathology , Humans , Psoriasis/drug therapy , Psoriasis/pathology , Psoriasis/chemically induced , Imiquimod/chemistry , Imiquimod/pharmacology , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/pharmacology , Female
5.
Bioorg Chem ; 143: 107095, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38211548

ABSTRACT

Cancer is indeed considered a hazardous and potentially life-threatening disorder. The JAK/STAT pathway is an important intracellular signaling cascade essential for many physiological functions, such as immune response, cell proliferation, and differentiation. Dysregulation of this pathway aids in the progression and development of cancer. The downstream JAK2/STAT3 signaling cascades are legitimate targets against which newer anticancer drugs can be developed to prevent and treat cancer. Understanding the mechanisms behind JAK2/STAT3 participation in cancer has paved the way for developing innovative targeted medicines with the potential to improve cancer treatment outcomes. This article provides information on the current scenario and recent advancements in the design and development of anticancer drugs targeting JAK2/STAT3, including structural analysis and SAR investigations of synthesized molecules. Numerous preclinical and clinical trials are ongoing on these inhibitors, which are highlighted to gain more insight into the broader development prospects of inhibitors of JAK2/STAT3.


Subject(s)
Antineoplastic Agents , Janus Kinase Inhibitors , Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Janus Kinase 2/antagonists & inhibitors , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/pharmacology , Janus Kinases/antagonists & inhibitors , Neoplasms/drug therapy , Signal Transduction , STAT Transcription Factors/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Structure-Activity Relationship
6.
Molecules ; 28(15)2023 Aug 06.
Article in English | MEDLINE | ID: mdl-37570884

ABSTRACT

Rheumatoid arthritis (RA) remains one of the most prevalent autoimmune diseases worldwide. Janus kinase 3 (JAK3) is an essential enzyme for treating autoimmune diseases, including RA. Molecular modeling techniques play a crucial role in the search for new drugs by reducing time delays. In this study, the 3D-QSAR approach is employed to predict new JAK3 inhibitors. Two robust models, both field-based with R2 = 0.93, R = 0.96, and Q2 = 87, and atom-based with R2 = 0.94, R = 0.97, and Q2 = 86, yielded good results by identifying groups that may readily direct their interaction. A reliable pharmacophore model, DHRRR1, was provided in this work to enable the clear characterization of chemical features, leading to the design of 13 inhibitors with their pIC50 values. The DHRRR1 model yielded a validation result with a ROC value of 0.87. Five promising inhibitors were selected for further study based on an ADMET analysis of their pharmacokinetic properties and covalent docking (CovDock). Compared to the FDA-approved drug tofacitinib, the pharmaceutical features, binding affinity and stability of the inhibitors were analyzed through CovDock, 300 ns molecular dynamics simulations, free energy binding calculations and ADMET predictions. The results show that the inhibitors have strong binding affinity, stability and favorable pharmaceutical properties. The newly predicted molecules, as JAK3 inhibitors for the treatment of RA, are promising candidates for use as drugs.


Subject(s)
2-Aminopurine , Antirheumatic Agents , Computer-Aided Design , Drug Design , Janus Kinase 3 , Janus Kinase Inhibitors , 2-Aminopurine/analogs & derivatives , 2-Aminopurine/pharmacology , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/pharmacology , Janus Kinase 3/antagonists & inhibitors , Quantitative Structure-Activity Relationship , Piperidines/chemistry , Piperidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Arthritis, Rheumatoid/drug therapy , Antirheumatic Agents/chemistry , Antirheumatic Agents/pharmacology , Pharmacophore
7.
Int J Mol Sci ; 23(4)2022 Feb 16.
Article in English | MEDLINE | ID: mdl-35216314

ABSTRACT

Hyperactivation of Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) signaling is an attractive therapeutic target for tumor therapy. Herein, forty-eight novel meridianin derivatives were designed and synthesized, and their antitumor activity was evaluated in vitro both for activity optimization and structure-activity relationship (SAR) study. The results indicated that most derivatives exhibited significantly improved antitumor activity, especially for compound 6e. The compound 6e contains an isothiouronium linked by an alkyl chain consisting of six carbon atoms with IC50 ranging from 1.11 to 2.80 µM on various cancer cell lines. Consistently, the 6e dose dependently induced the apoptosis of A549 and DU145 cells, in which STAT3 is constitutively active. Western blotting assays indicated that the phosphorylation levels of JAK1, JAK2 and STAT3 were inhibited by 6e at 5 µM without significant change in the total STAT3 level. Moreover, 6e also suppressed the expression of STAT3 downstream genes, including c-Myc, Cyclin D1 and Bcl-XL at 10 µM. An additional in vivo study revealed that 6e at the dose of 10 mg/kg could potently inhibit the DU145 xenograft tumor without obvious body weight loss. These results clearly indicate that 6e could be a potential antitumor agent by targeting the JAK/STAT3 signaling pathway.


Subject(s)
Indole Alkaloids , Janus Kinase Inhibitors , STAT3 Transcription Factor , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Indole Alkaloids/chemistry , Indole Alkaloids/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/pharmacology , Phosphorylation , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 55: 128451, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34774741

ABSTRACT

JAK inhibitors have been considered as useful targets for the treatment of related diseases. However, first-generation JAK inhibitors have side effects such as anemia, thrombocytopenia, neutropenia and headaches which have been suggested to result from high JAK2 inhibition. Second-generation JAK inhibitors with more specific JAK isozyme inhibition have been studied to eliminate these adverse effects. In this study, novel 4-(1,5- or 2,5-triazole)-pyrrolopyrimidine derivatives with aromatic moieties were synthesized as JAK1 inhibitors, and an in vitro enzyme assay was used to evaluate the JAK inhibitory effects. Among these JAK1 inhibitors, the compound 23a showed an IC50 level of 72 nM, as well as being selective against other JAKs by 12 times or more: the results of molecular docking studies suggested that the high JAK1 selectivity resulted from a key interaction between the iodine atom of compound 23a and His-885 of hJAK1.


Subject(s)
Janus Kinase 1/antagonists & inhibitors , Janus Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Triazoles/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Janus Kinase 1/metabolism , Janus Kinase Inhibitors/chemical synthesis , Janus Kinase Inhibitors/chemistry , Models, Molecular , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry
9.
Mol Pharm ; 18(8): 3063-3072, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34247482

ABSTRACT

Traditionally, cutaneous drug delivery is studied by skin accumulation or skin permeation, while alternative techniques may enable the interactions between the drug and the skin to be studied in more detail. Time-resolved skin profiling for pharmacokinetic monitoring of two Janus Kinase (JAK) inhibitors, tofacitinib and LEO 37319A, was performed using dermal open-flow microperfusion (dOFM) for sampling of perfusate in an ex vivo and in vivo setup in pig skin. Additionally, matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) was performed to investigate depth-resolved skin distributions at defined time points ex vivo in human skin. By dOFM, higher skin concentrations were observed for tofacitinib compared to LEO 37319A, which was supported by the lower molecular weight, higher solubility, lipophilicity, and degree of protein binding. Using MALDI-MSI, the two compounds were observed to show different skin distributions, which was interpreted to be caused by the difference in the ability of the two molecules to interact with the skin compartments. In conclusion, the techniques assessed time- and depth-resolved skin concentrations and were able to show differences in the pharmacokinetic profiles of two JAK inhibitors. Thus, evidence shows that the two techniques can be used as complementary methods to support decision making in drug development.


Subject(s)
Janus Kinase Inhibitors/administration & dosage , Janus Kinase Inhibitors/pharmacokinetics , Perfusion/methods , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Skin Absorption/drug effects , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Administration, Cutaneous , Animals , Drug Compounding/methods , Female , Humans , Janus Kinase Inhibitors/chemistry , Middle Aged , Molecular Weight , Piperidines/chemistry , Pyrimidines/chemistry , Skin/drug effects , Skin/metabolism , Solubility , Swine , Tissue Distribution
10.
FEBS J ; 288(22): 6528-6542, 2021 11.
Article in English | MEDLINE | ID: mdl-34009721

ABSTRACT

Calcific aortic valve disease (CAVD) is the most prevalent valvulopathy worldwide. Growing evidence supports a role for viral and cell-derived double-stranded (ds)-RNA in cardiovascular pathophysiology. Poly(I:C), a dsRNA surrogate, has been shown to induce inflammation, type I interferon (IFN) responses, and osteogenesis through Toll-like receptor 3 in aortic valve interstitial cells (VIC). Here, we aimed to determine whether IFN signaling via Janus kinase (JAK)/Signal transducers and activators of transcription (STAT) mediates dsRNA-induced responses in primary human VIC. Western blot, ELISA, qPCR, calcification, flow cytometry, and enzymatic assays were performed to evaluate the mechanisms of dsRNA-induced inflammation and calcification. Poly(I:C) triggered a type I IFN response characterized by IFN-regulatory factors gene upregulation, IFN-ß secretion, and STAT1 activation. Additionally, Poly(I:C) promoted VIC inflammation via NF-κB and subsequent adhesion molecule expression, and cytokine secretion. Pretreatment with ruxolitinib, a clinically used JAK inhibitor, abrogated these responses. Moreover, Poly(I:C) promoted a pro-osteogenic phenotype and increased VIC calcification to a higher extent in cells from males. Inhibition of JAK with ruxolitinib or a type I IFN receptor blocking antibody blunted Poly(I:C)-induced calcification. Mechanistically, Poly(I:C) promoted VIC apoptosis in calcification medium, which was inhibited by ruxolitinib. Moreover, Poly(I:C) co-operated with IFN-γ to increase VIC calcification by synergistically activating extracellular signal-regulated kinases and hypoxia-inducible factor-1α pathways. In conclusion, JAK/STAT signaling mediates dsRNA-triggered inflammation, apoptosis, and calcification and may contribute to a positive autocrine loop in human VIC in the presence of IFN-γ. Blockade of dsRNA responses with JAK inhibitors may be a promising therapeutic avenue for CAVD.


Subject(s)
Aortic Valve Stenosis/drug therapy , Aortic Valve/drug effects , Aortic Valve/pathology , Calcinosis/drug therapy , Inflammation/drug therapy , Janus Kinase Inhibitors/pharmacology , Nitriles/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Double-Stranded/antagonists & inhibitors , Adolescent , Adult , Aged , Aortic Valve/metabolism , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Calcinosis/metabolism , Calcinosis/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Janus Kinase Inhibitors/chemistry , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Male , Middle Aged , Nitriles/chemistry , Pyrazoles/chemistry , Pyrimidines/chemistry , RNA, Double-Stranded/metabolism , Young Adult
11.
Drug Des Devel Ther ; 15: 1135-1147, 2021.
Article in English | MEDLINE | ID: mdl-33731985

ABSTRACT

Atopic dermatitis (AD) is a chronic, pruritic, inflammatory skin disease that predominantly affects children. However, it can persist in adulthood and/or start at older ages. Both dysfunction of the epidermal barrier and immune dysregulation are known to play a role in the pathogenesis of AD. In the last years, numerous studies showed that Janus kinase (JAK) enzymes have a key role in AD pathogenesis. Therefore, oral and topical JAK inhibitors are new emerging treatments for AD. We report the data relating to abrocitinib, an oral JAK1 inhibitor. For this purpose, we examined articles already published concerning, in particular, concluded clinical trials. Furthermore, we also report the design of current ongoing clinical trials. The search was carried out considering the main search engines relating to medical literature and clinical trials. From all the data we collected, abrocitinib proved to be an effective drug in significantly reducing the severity of moderate-to-severe AD when compared to placebo. Furthermore, the efficacy was similar to other well-established treatment for AD, such as dupilumab. Adverse events were generally mild; indeed, the drug was definitively suspended only in few patients.


Subject(s)
Dermatitis, Atopic/drug therapy , Janus Kinase 1/antagonists & inhibitors , Janus Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Clinical Trials as Topic , Dermatitis, Atopic/metabolism , Humans , Janus Kinase 1/metabolism , Janus Kinase Inhibitors/chemistry , Pyrimidines/chemistry , Sulfonamides/chemistry
12.
Leukemia ; 35(1): 1-17, 2021 01.
Article in English | MEDLINE | ID: mdl-32647323

ABSTRACT

Myeloproliferative neoplasm (MPN)-associated myelofibrosis (MF) is characterized by cytopenias, marrow fibrosis, constitutional symptoms, extramedullary hematopoiesis, splenomegaly, and shortened survival. Constitutive activation of the janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway in MF leads to cell proliferation, inhibition of cell death, and clonal expansion of myeloproliferative malignant cells. Fedratinib is a selective oral JAK2 inhibitor recently approved in the United States for treatment of adult patients with intermediate-2 or high-risk MF. In mouse models of JAK2V617F-driven myeloproliferative disease, fedratinib blocked phosphorylation of STAT5, increased survival, and improved MF-associated disease features, including reduction of white blood cell counts, hematocrit, splenomegaly, and fibrosis. Fedratinib exerts off-target inhibitory activity against bromodomain-containing protein 4 (BRD4); combination JAK/STAT and BRD4 inhibition was shown to synergistically block NF-kB hyperactivation and inflammatory cytokine production, attenuating disease burden and reversing bone marrow fibrosis in animal models of MPNs. In patients, fedratinib is rapidly absorbed and dosed once daily (effective half-life 41 h). Fedratinib showed robust clinical activity in JAK-inhibitor-naïve patients and in patients with MF who were relapsed, refractory, or intolerant to prior ruxolitinib therapy. Fedratinib is effective regardless of JAK2 mutation status. Onset of spleen and symptom responses are typically seen within the first 1-2 months of treatment. The most common adverse events (AEs) with fedratinib are grades 1-2 gastrointestinal events, which are most frequent during early treatment and decrease over time. Treatment discontinuation due to hematologic AEs in clinical trials was uncommon (~3%). Suspected cases of Wernicke's encephalopathy were reported during fedratinib trials in ~1% of patients; thiamine levels should be monitored before and during fedratinib treatment as medically indicated. Phase III trials are ongoing to assess fedratinib effects on long-term safety, efficacy, and overall survival. The recent approval of fedratinib provides a much-needed addition to the limited therapeutic options available for patients with MF.


Subject(s)
Janus Kinase Inhibitors/therapeutic use , Myeloproliferative Disorders/complications , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/etiology , Pyrrolidines/therapeutic use , Sulfonamides/therapeutic use , Animals , Biomarkers , Clinical Trials as Topic , Cytokines/metabolism , Disease Susceptibility , Drug Monitoring , Humans , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/pharmacology , Janus Kinases/antagonists & inhibitors , Janus Kinases/chemistry , Janus Kinases/genetics , Janus Kinases/metabolism , Molecular Targeted Therapy , Myeloproliferative Disorders/diagnosis , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/metabolism , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Treatment Outcome
13.
J Mol Model ; 26(9): 235, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32808204

ABSTRACT

Rheumatoid arthritis is an autoimmune disorder causing joint deformity and work disability. Several drugs are available to deal with the disease including conventional drugs; biological drugs such as TNFα inhibitors, B cell-targeted drugs, T cell co-stimulation inhibitors, interleukin-6 inhibitors, and interleukin-1 inhibitors; and kinase inhibitory drugs. In spite of the broad spectrum of drugs available, the disease remains uncontrolled in a number of patients and there is a need for new drugs with better efficacy and universal response rate. The failure of the available drugs to control the disease can be owed to the complex pathogenesis with complementary pathways of disease progression. The blockade of one pathway cannot supersede pathogenesis through other complementary pathways. Janus kinase (JAK) and Bruton's tyrosine kinase (BTK) are the two important mediators of disease which control a number of signaling pathways involved in rheumatoid arthritis pathogenesis. In this study, using the computer-aided drug designing techniques (virtual screening, molecular docking, and molecular dynamics studies), we have designed piperidinyl dipyrrolopyridine-based dual inhibitors of Janus kinase and Bruton's tyrosine kinase. Dual JAK and BTK inhibitors seem promising to fight the complex pathogenesis of the disease at multiple fronts and can be the future drug for patients unresponsive to current remedies.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/chemistry , Janus Kinase Inhibitors/chemistry , Janus Kinases/chemistry , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Arthritis, Rheumatoid/drug therapy , Binding Sites , Drug Design , Drug Development , Humans , Janus Kinase Inhibitors/pharmacology , Janus Kinases/antagonists & inhibitors , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Structure-Activity Relationship
14.
Rheumatology (Oxford) ; 59(9): 2217-2225, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32406509

ABSTRACT

OBJECTIVES: Small molecule tyrosine kinase inhibitors [smTKI, comprising mostly of Janus kinase (JAK) and to a lesser extent, spleen tyrosine kinase (SyK) inhibitors] modulate the cytokine receptor-mediated intracellular signal cascade, and are an effective treatment for autoimmune diseases and malignancies. As smTKI are novel, long-term safety is uncertain. Due to increasing use, characterization of their true adverse event profile is critical. METHODS: We performed a systematic review and meta-analysis of all published trial data on the pulmonary and serious adverse effects of smTKIs in autoimmune disease. EMBASE, MEDLINE, CENTRAL and Pneumotox databases were searched up to April 2019 for randomized controlled trials, observational studies and post marketing surveillance, comparing any smTKI with placebo or another therapy, or as monotherapy at different doses. Primary outcomes comprised of any respiratory complications including upper and lower respiratory tract infections (URTI, LRTI), influenza, pneumonia, opportunistic respiratory infections, drug-induced interstitial lung disease, pulmonary embolism and lung neoplasm. RESULTS: We identified 4667 citations for screening, and selected 319 studies for full text review. Seventy-nine studies were analysed, including 47 randomized controlled trials, 25 observational studies and seven post-marketing surveillance studies, comprising 159 652 participants. There were significantly increased risks of URTI [risk difference (RD) 0.03; 95% CI: 0.01, 0.05; P = 0.00; 36 studies, 14 724 participants], LRTI (RD 0.01; 95% CI: 0.00, 0.02; P = 0.02; 24 studies, 12 302 participants), influenza (RD 0.01; 95% CI: 0.00, 0.01; P = 0.04; 22 studies, 10 684 participants), and pneumonia (RD 0.00; 95% CI: 0.00, 0.01; P = 0.02; 33 studies, 15 511 participants). No increased risk was found for other respiratory complications, including pulmonary embolism. CONCLUSION: SmTKI increases the risk of non-opportunistic respiratory infections compared with placebo. The risk of any serious pulmonary adverse events is low.


Subject(s)
Autoimmune Diseases/drug therapy , Janus Kinase Inhibitors/adverse effects , Lung Diseases/chemically induced , Piperidines/adverse effects , Pyrimidines/adverse effects , Small Molecule Libraries/adverse effects , Adult , Aged , Female , Humans , Janus Kinase Inhibitors/chemistry , Male , Middle Aged , Piperidines/chemistry , Product Surveillance, Postmarketing , Pyrimidines/chemistry , Randomized Controlled Trials as Topic , Small Molecule Libraries/chemistry
15.
Molecules ; 25(9)2020 May 01.
Article in English | MEDLINE | ID: mdl-32370021

ABSTRACT

The solubility and thermodynamic analysis of baricitinib (BNB) in various dimethyl sulfoxide (DMSO) + water mixtures were performed. The "mole fraction solubilities (xe)" of BNB in DMSO and water mixtures were determined at "T = 298.2-323.2 K" and "p = 0.1 MPa" using an isothermal saturation technique. "Hansen solubility parameters (HSPs)" of BNB, pure DMSO, pure water and "DMSO + water" mixtures free of BNB were also estimated. The xe data of BNB was regressed well by five different thermodynamics-based co-solvency models, which included "Apelblat, Van't Hoff, Yalkowsky-Roseman, Jouyban-Acree and Jouyban-Acree-Van't Hoff models" with overall deviations of <5.0%. The highest and lowest xe value of BNB was computed in pure DMSO (1.69 × 10-1 at T = 323.2 K) and pure water (2.23 × 10-5 at T = 298.2 K), respectively. The HSP of BNB was found to be closer to that of pure DMSO. Based on activity coefficient data, maximum solute-solvent molecular interactions were observed in BNB-DMSO compared to BNB-water. The results of "apparent thermodynamic analysis" indicated endothermic and entropy-drive dissolution of BNB in all "DMSO + water" combinations including mono-solvents (water and DMSO). "Enthalpy-entropy compensation analysis" showed enthalpy-driven to be the main mechanism of solvation of BNB.


Subject(s)
Antineoplastic Agents/chemistry , Azetidines/chemistry , Janus Kinase Inhibitors/chemistry , Models, Molecular , Purines/chemistry , Pyrazoles/chemistry , Sulfonamides/chemistry , Dimethyl Sulfoxide/chemistry , Molecular Structure , Solubility , Solvents/chemistry , Thermodynamics , Water/chemistry
16.
Molecules ; 25(7)2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32244454

ABSTRACT

Baricitinib, is a selective and reversible Janus kinase inhibitor, is commonly used to treat adult patients with moderately to severely active rheumatoid arthritis (RA). A fast, reproducible and sensitive method of liquid chromatography-tandem mass spectrometry (LC-MS/MS) for the quantification of baricitinib in rat plasma has been developed. Irbersartan was used as the internal standard (IS). Baracitinib and IS were extracted from plasma by liquid-liquid extraction using a mixture of n-hexane and dichloromethane (1:1) as extracting agent. Chromatographic separation was performed using Acquity UPLC HILIC BEH 1.7 µm 2.1 × 50 mm column with the mobile phase consisting of 0.1% formic acid in acetonitrile and 20 mM ammonium acetate (pH 3) (97:3). The electrospray ionization in the positive-mode was used for sample ionization in the multiple reaction monitoring mode. Baricitinib and the IS were quantified using precursor-to-production transitions of m/z 372.15 > 251.24 and 429.69 > 207.35 for baricitinib and IS, respectively. The method was validated according to the recent FDA and EMA guidelines for bioanalytical method validation. The lower limit of quantification was 0.2 ng/mL, whereas the intra-day and inter-day accuracies of quality control (QCs) samples were ranged between 85.31% to 89.97% and 87.50% to 88.33%, respectively. Linearity, recovery, precision, and stability parameters were found to be within the acceptable range. The method was applied successfully applied in pilot pharmacokinetic studies.


Subject(s)
Azetidines/pharmacokinetics , Chromatography, High Pressure Liquid , Janus Kinase Inhibitors/pharmacokinetics , Purines/pharmacokinetics , Pyrazoles/pharmacokinetics , Sulfonamides/pharmacokinetics , Tandem Mass Spectrometry , Animals , Azetidines/chemistry , Drug Monitoring , Humans , Janus Kinase Inhibitors/chemistry , Purines/chemistry , Pyrazoles/chemistry , Rats , Reproducibility of Results , Sensitivity and Specificity , Sulfonamides/chemistry
17.
Drugs ; 80(6): 609-615, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32166597

ABSTRACT

Delgocitinib, a janus kinase (JAK) inhibitor, is being developed by Japan Tobacco for the treatment of autoimmune disorders and hypersensitivity, including inflammatory skin conditions. The JAK family of tyrosine kinases plays an important role in mediating the biological effects of several inflammatory cytokines, e.g. IL-4, IL-13 and IL-31, which are elevated in patients with atopic dermatitis. Delgocitinib inhibits all members of the JAK family [JAK1, JAK2, JAK3 and tyrosine kinase 2]. Topical delgocitinib (Corectim®) is approved in Japan for the treatment of atopic dermatitis. This article summarizes the milestones in the development of delgocitinib leading to this first approval for the treatment of adults with atopic dermatitis. Clinical development of the topical formulation is also underway for alopecia areata, chronic hand eczema, discoid lupus erythematosus, inverse psoriasis and atopic dermatitis in several countries worldwide. Clinical development of an oral formulation of delgocitinib is also underway in Japan for the treatment of autoimmune disorders and hypersensitivity.


Subject(s)
Autoimmune Diseases/drug therapy , Drug Approval , Hypersensitivity/drug therapy , Janus Kinase Inhibitors/pharmacology , Janus Kinases/antagonists & inhibitors , Pyrroles/pharmacology , Animals , Autoimmune Diseases/metabolism , Humans , Hypersensitivity/metabolism , Janus Kinase Inhibitors/chemistry , Molecular Conformation , Pyrroles/chemistry
18.
J Med Chem ; 63(6): 2915-2929, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32134643

ABSTRACT

To identify Janus kinase (JAK) inhibitors that selectively target gastrointestinal tissues with limited systemic exposures, a class of imidazopyrrolopyridines with a range of physical properties was prepared and evaluated. We identified compounds with low intrinsic permeability and determined a correlation between permeability and physicochemical properties, clogP and tPSA, for a subset of compounds. This low intrinsic permeability translated into compounds displaying high colonic exposure and low systemic exposure after oral dosing at 25 mg/kg in mouse. In a mouse PK/PD model, oral dosing of lead compound 2 demonstrated dose-dependent inhibition of pSTAT phosphorylation in colonic explants post-oral dose but low systemic exposure and no measurable systemic pharmacodynamic activity. We thus demonstrate the utility of JAK inhibitors with low intrinsic permeability as a feasible approach to develop gut-restricted, pharmacologically active molecules with a potential advantage over systemically available compounds that are limited by systemic on-target adverse events.


Subject(s)
Inflammatory Bowel Diseases/drug therapy , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/pharmacokinetics , Pyridines/pharmacology , Pyridines/pharmacokinetics , Administration, Oral , Animals , Dogs , Drug Discovery , Female , Humans , Inflammatory Bowel Diseases/metabolism , Janus Kinase Inhibitors/administration & dosage , Janus Kinase Inhibitors/chemistry , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Permeability , Phosphorylation/drug effects , Pyridines/administration & dosage , Pyridines/chemistry
19.
Rev Med Suisse ; 16(685): 477-480, 2020 Mar 11.
Article in French | MEDLINE | ID: mdl-32167248

ABSTRACT

If treatment target is not reached with a conventional disease-modifying antirheumatic drug (csDMARD) such as methotrexate in rheumatoid arthritis, it is recommended to use a biologic (bDMARD) or a Janus kinase (JAK) inhibitor, small synthetic molecules recently developed. Oral administration and short half-life can favour the choice of JAK inhibitors, as well as the opportunity to use it in monotherapy, even though co-treatment with csDMARD is recommended. As yet long-term studies are lacking, little is known about adverse effects although risk of herpes virus infections is clearly higher with JAK inhibitors than bDMARD treatments.


Lors du contrôle insuffisant de l'activité de la polyarthrite rhumatoïde (PR) par traitement conventionnel (csDMARD) comme le méthotrexate, il est recommandé d'introduire un traitement biologique (bDMARD) ou un inhibiteur de Janus kinase (JAK). Ces derniers sont de petites molécules synthétiques récemment développées. La prise per os et une demi-vie courte peuvent favoriser le choix d'un inhibiteur de JAK. De plus, une utilisation en monothérapie est envisageable, bien qu'une association avec un csDMARD soit préférée. En raison d'un manque d'études à long terme, leurs effets indésirables sont peu connus, hormis les infections à virus herpétique qui sont plus élevées qu'avec les bDMARD.


Subject(s)
Antirheumatic Agents/chemistry , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Janus Kinase Inhibitors/therapeutic use , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/adverse effects , Drug Therapy, Combination , Humans , Janus Kinase Inhibitors/administration & dosage , Janus Kinase Inhibitors/adverse effects , Janus Kinase Inhibitors/chemistry , Methotrexate/therapeutic use , Treatment Outcome
20.
Drugs ; 80(4): 341-352, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32020553

ABSTRACT

Despite advances in the treatment of psoriasis, there is an unmet need for effective and safe oral treatments. The Janus Kinase-Signal Transducer and Activator of Transcription (JAK-STAT) pathway plays a significant role in intracellular signalling of cytokines of numerous cellular processes, important in both normal and pathological states of immune-mediated inflammatory diseases. Particularly in psoriasis, where the interleukin (IL)-23/IL-17 axis is currently considered the crucial pathogenic pathway, blocking the JAK-STAT pathway with small molecules would be expected to be clinically effective. However, relative non-specificity and low therapeutic index of the available JAK inhibitors have delayed their integration into the therapeutic armamentarium of psoriasis. Current research appears to be focused on Tyrosine kinase 2 (TYK2), the first described member of the JAK family. Data from the Phase II trial of BMS-986165-a selective TYK2 inhibitor-in psoriasis have been published and clinical results are encouraging, with a large Phase III programme ongoing. Further, the selective TYK2 inhibitor PF-06826647 is being tested in moderate-to-severe psoriasis in a Phase II clinical trial. Brepocitinib, a potent TYK2/JAK1 inhibitor, is also being evaluated, as both oral and topical treatment. Results of studies with TYK2 inhibitors will be important in assessing the clinical efficacy and safety of these drugs and their place in the therapeutic armamentarium of psoriasis. This article reviews current data on the impact of JAK inhibitors in the treatment of adult patients with moderate-to-severe psoriasis.


Subject(s)
Heterocyclic Compounds/pharmacology , Janus Kinase Inhibitors/pharmacology , Psoriasis/drug therapy , TYK2 Kinase/antagonists & inhibitors , Heterocyclic Compounds/chemistry , Humans , Janus Kinase Inhibitors/chemistry , Psoriasis/metabolism , TYK2 Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...