Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 903
Filter
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.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731900

ABSTRACT

Psoriasis is a highly prevalent dermatological disease associated with an increased systemic inflammatory response. In addition, joint involvement is also present in around 20% of patients. Therefore, treatment modalities used in this condition should be simultaneously effective at improving skin manifestations, reducing inflammation, and addressing psoriatic arthritis when present. Twenty years ago, the introduction of biologic treatments for psoriasis was a turning point in the management of this condition, offering an effective and reasonably safe option for patients whose disease could not be adequately controlled with conventional therapies. At the moment, Janus Kinase inhibitors (JAKis) are a new class of promising molecules in the management of psoriasis. They are orally administered and can show benefits in patients who failed biologic therapy. We conducted a scoping review in order to identify randomized-controlled trials that investigated different JAKis in patients with plaque psoriasis and psoriatic arthritis, with an emphasis on molecules that have been approved by the European Medicines Agency and the Food and Drug Administration. The added value of this study is that it collected information about JAKis approved for two different indications, plaque psoriasis and psoriatic arthritis, in order to provide an integrated understanding of the range of effects that JAKis have on the whole spectrum of psoriasis manifestations.


Subject(s)
Janus Kinase Inhibitors , Janus Kinases , Psoriasis , STAT Transcription Factors , Signal Transduction , Humans , Psoriasis/drug therapy , Psoriasis/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 , Arthritis, Psoriatic/drug therapy , Arthritis, Psoriatic/metabolism
3.
Expert Rev Clin Immunol ; 20(6): 589-602, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38299575

ABSTRACT

INTRODUCTION: Juvenile dermatomyositis (JDM) is a rare autoimmune disease most commonly with proximal weakness due to inflammation and characteristic skin rashes. Most patients have a chronic or polycyclic disease course on standard therapy so better treatments are needed. An interferon signature is well-established in key tissues of JDM. Janus kinase inhibitors (jakinibs), which can decrease IFN signaling, are therefore appealing as a targeted therapy. AREAS COVERED: Herein is a review of the growing literature on JDM patients in jakinibs, including specifics of their jakinib exposure, summary of efficacy, disease features, and characteristics of patients treated, and safety parameters. EXPERT OPINION: The vast majority of refractory JDM patients respond to jakinib therapy, though they have varied features, doses, and previous/concurrent medications, and data is largely retrospective. Jakinibs are an exciting and promising treatment in JDM. Evaluation with larger prospective controlled studies is needed to answer remaining questions about jakinibs in JDM regarding dosing, which JDM patients to treat with jakinibs, potential biomarkers to use, and how best to monitor safety risks in JDM.


Subject(s)
Dermatomyositis , Janus Kinase Inhibitors , Humans , Dermatomyositis/drug therapy , Dermatomyositis/immunology , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/adverse effects , Child , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Interferons/therapeutic use , Signal Transduction/drug effects
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.
Biofactors ; 50(2): 360-370, 2024.
Article in English | MEDLINE | ID: mdl-37737462

ABSTRACT

The prevalence of diabetes mellitus (DM) is alarmingly increasing worldwide. Diabetic retinopathy (DR) is a prevailing DM microvascular complication, representing the major cause of blindness in working-age population. Inflammation is a crucial player in DR pathogenesis. JAK/STAT3 axis is a pleotropic cascade that modulates diverse inflammatory events. Nifuroxazide (Nifu) is a commonly used oral antibiotic with reported JAK/STAT3 inhibition activity. The present study investigated the potential protective effect of Nifu against diabetes-induced retinal injury. Effect of Nifu on oxidative stress, JAK/STAT3 axis and downstream inflammatory mediators has been also studied. Diabetes was induced in Sprague Dawley rats by single intraperitoneal injection of streptozotocin (50 mg/kg). Animals were assigned into four groups: normal, Nifu control, DM, and DM + Nifu. Nifu was orally administrated at 25 mg/kg/day for 8 weeks. The effects of Nifu on oxidative stress, JAK/STAT3 axis proteins, inflammatory factors, tight junction proteins, histological, and ultrastructural alterations were evaluated using spectrophotometry, gene and protein analyses, and histological studies. Nifu administration to diabetic rats attenuated histopathological and signs of retinal injury. Additionally, Nifu attenuated retinal oxidative stress, inhibited JAK and STAT3 phosphorylation, augmented the expression of STAT3 signaling inhibitor SOCS3, dampened the expression of transcription factor of inflammation NF-κB, and inflammatory cytokine TNF-α. Collectively, the current study indicated that Nifu alleviated DR progression in diabetic rats, suggesting beneficial retino-protective effect. This can be attributed to blocking JAK/STAT3 axis in retinal tissues with subsequent amelioration of oxidative stress and inflammation.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy , Hydroxybenzoates , Nitrofurans , Animals , Rats , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Inflammation/drug therapy , Inflammation/genetics , Inflammation/complications , Nitrofurans/pharmacology , Nitrofurans/therapeutic use , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Janus Kinases/antagonists & inhibitors , Janus Kinases/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/drug effects
7.
Front Biosci (Landmark Ed) ; 28(8): 176, 2023 08 23.
Article in English | MEDLINE | ID: mdl-37664943

ABSTRACT

BACKGROUND: Rheumatoid arthritis (RA) is a systemic autoimmune disease that causes progressive joint damage. The Janus kinase (JAK) inhibitors (JAK-I) represent a new therapeutic option for RA patients, blocking the intracellular JAK-STAT pathway. Today, no studies have been conducted to determine whether new biomarkers could better reflect disease activity in patients treated with JAK-I than traditional disease activity indicators. Thus, the aim of our study was to determine additional disease activity biomarkers in RA patients receiving selective JAK-1 inhibitors. METHODS: we enrolled 57 patients with RA: 34 patients were treated with Upadacitinib (UPA) and 23 patients with Filgotinib (FIL). All patients were evaluated for clinimetry with DAS28 and Crohn's Disease Activity Index (CDAI), number of tender and swollen joints, Visual Analogic Scale (VAS), Physician Global Assessment (PhGA), and Health Assessment Questionnaire (HAQ), at baseline and at the 12th week of treatment. Lymphocyte subpopulations, complete blood count, erythrocyte sedimentation rate (ESR), C-Reactive Protein (CRP), anti-cyclic citrullinated peptide antibodies (APCA), rheumatoid factor (RF) IgM, interleukin 6 (IL-6), circulating calprotectin (cCLP), tumor necrosis factor α (TNFα), soluble urokinase Plasminogen Activator Receptor (suPAR), complement functional activity were measured at baseline and after the 12th week of treatment. RESULTS: in both groups of patients, we documented a significant reduction in the clinimetric parameters DAS28, CDAI, number of tender joints, number of swollen joints, VAS, PhGA, and HAQ. Moreover, significant differences were reported for laboratory parameters of ESR, CRP, IL-6, suPAR, cCLP, and PLT/L ratio in both groups. However, no difference was demonstrated between the two groups for changes in renal, hepatic, and lipid parameters. CONCLUSIONS: the suPAR and cCLP levels may lead towards a different therapeutic choice between UPA and FIL, with the expression of two different RA pathophenotypes directing FIL towards a lymphocyte-poor form and UPA towards a myeloid form of RA.


Subject(s)
Arthritis, Rheumatoid , Janus Kinases , Receptors, Urokinase Plasminogen Activator , Humans , Arthritis, Rheumatoid/drug therapy , Biomarkers , C-Reactive Protein , Interleukin-6 , Janus Kinases/antagonists & inhibitors , Signal Transduction , STAT Transcription Factors
8.
N Engl J Med ; 388(24): 2241-2252, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37256972

ABSTRACT

BACKGROUND: Disabling pansclerotic morphea (DPM) is a rare systemic inflammatory disorder, characterized by poor wound healing, fibrosis, cytopenias, hypogammaglobulinemia, and squamous-cell carcinoma. The cause is unknown, and mortality is high. METHODS: We evaluated four patients from three unrelated families with an autosomal dominant pattern of inheritance of DPM. Genomic sequencing independently identified three heterozygous variants in a specific region of the gene that encodes signal transducer and activator of transcription 4 (STAT4). Primary skin fibroblast and cell-line assays were used to define the functional nature of the genetic defect. We also assayed gene expression using single-cell RNA sequencing of peripheral-blood mononuclear cells to identify inflammatory pathways that may be affected in DPM and that may respond to therapy. RESULTS: Genome sequencing revealed three novel heterozygous missense gain-of-function variants in STAT4. In vitro, primary skin fibroblasts showed enhanced interleukin-6 secretion, with impaired wound healing, contraction of the collagen matrix, and matrix secretion. Inhibition of Janus kinase (JAK)-STAT signaling with ruxolitinib led to improvement in the hyperinflammatory fibroblast phenotype in vitro and resolution of inflammatory markers and clinical symptoms in treated patients, without adverse effects. Single-cell RNA sequencing revealed expression patterns consistent with an immunodysregulatory phenotype that were appropriately modified through JAK inhibition. CONCLUSIONS: Gain-of-function variants in STAT4 caused DPM in the families that we studied. The JAK inhibitor ruxolitinib attenuated the dermatologic and inflammatory phenotype in vitro and in the affected family members. (Funded by the American Academy of Allergy, Asthma, and Immunology Foundation and others.).


Subject(s)
Autoimmune Diseases , Dermatologic Agents , Janus Kinases , Scleroderma, Systemic , Janus Kinases/antagonists & inhibitors , Nitriles , Pyrazoles/therapeutic use , Pyrazoles/pharmacology , Pyrimidines , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/genetics , Autoimmune Diseases/drug therapy , Autoimmune Diseases/genetics , Mutation, Missense , Gain of Function Mutation , Dermatologic Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use
9.
Bioorg Med Chem Lett ; 79: 129083, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36414177

ABSTRACT

TYK2, a member of the JAK family of proximal membrane-bound tyrosine kinases, has emerged as an attractive target for the treatment of autoimmune diseases. Herein, we report the discovery of first-in-class potent and subtype-selective TYK2 degraders. By conjugating a TYK2 ligand from a known allosteric TYK2 inhibitor with a VHL ligand as the E3 ligase ligand via alkyl linkers of various lengths, we rapidly identified TYK2 degrader 5 with moderate TYK2 degradation activity. Degrader 5 induced TYK2 degradation without affecting the protein level of subtype kinases (JAK1, JAK2, and JAK3) in Jurkat cellular assays. Furthermore, modifying the TYK2 ligand moiety of degrader 5 yielded the more potent TYK2 degrader 37 with retained selectivity for JAKs. Our subtype-selective TYK2 degraders represent valuable chemical probes for investigating the biology of TYK2 degradation.


Subject(s)
TYK2 Kinase , Humans , Autoimmune Diseases/drug therapy , Janus Kinases/antagonists & inhibitors , Ligands , Phosphorylation , Protein Processing, Post-Translational , TYK2 Kinase/antagonists & inhibitors , /pharmacology
10.
N Engl J Med ; 387(16): 1445-1455, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36260792

ABSTRACT

BACKGROUND: Vitiligo is a chronic autoimmune disease that causes skin depigmentation. A cream formulation of ruxolitinib (an inhibitor of Janus kinase 1 and 2) resulted in repigmentation in a phase 2 trial involving adults with vitiligo. METHODS: We conducted two phase 3, double-blind, vehicle-controlled trials (Topical Ruxolitinib Evaluation in Vitiligo Study 1 [TRuE-V1] and 2 [TRuE-V2]) in North America and Europe that involved patients 12 years of age or older who had nonsegmental vitiligo with depigmentation covering 10% or less of total body-surface area. Patients were randomly assigned in a 2:1 ratio to apply 1.5% ruxolitinib cream or vehicle control twice daily for 24 weeks to all vitiligo areas on the face and body, after which all patients could apply 1.5% ruxolitinib cream through week 52. The primary end point was a decrease (improvement) of at least 75% from baseline in the facial Vitiligo Area Scoring Index (F-VASI; range, 0 to 3, with higher scores indicating a greater area of facial depigmentation), or F-VASI75 response, at week 24. There were five key secondary end points, including improved responses on the Vitiligo Noticeability Scale. RESULTS: A total of 674 patients were enrolled, 330 in TRuE-V1 and 344 in TRuE-V2. In TRuE-V1, the percentage of patients with an F-VASI75 response at week 24 was 29.8% in the ruxolitinib-cream group and 7.4% in the vehicle group (relative risk, 4.0; 95% confidence interval [CI], 1.9 to 8.4; P<0.001). In TRuE-V2, the percentages were 30.9% and 11.4%, respectively (relative risk, 2.7; 95% CI, 1.5 to 4.9; P<0.001). The results for key secondary end points showed superiority of ruxolitinib cream over vehicle control. Among patients who applied ruxolitinib cream throughout 52 weeks, adverse events occurred in 54.8% in TRuE-V1 and 62.3% in TRuE-V2; the most common adverse events were application-site acne (6.3% and 6.6%, respectively), nasopharyngitis (5.4% and 6.1%), and application-site pruritus (5.4% and 5.3%). CONCLUSIONS: In two phase 3 trials, application of ruxolitinib cream resulted in greater repigmentation of vitiligo lesions than vehicle control through 52 weeks, but it was associated with acne and pruritus at the application site. Larger and longer trials are required to determine the effect and safety of ruxolitinib cream in patients with vitiligo. (Funded by Incyte; TRuE-V1 and TRuE-V2 ClinicalTrials.gov numbers, NCT04052425 and NCT04057573.).


Subject(s)
Janus Kinases , Nitriles , Pyrazoles , Pyrimidines , Vitiligo , Adult , Humans , Acne Vulgaris/chemically induced , Double-Blind Method , Pruritus/chemically induced , Treatment Outcome , Vitiligo/drug therapy , Janus Kinases/antagonists & inhibitors , Skin Cream/administration & dosage , Skin Cream/adverse effects , Skin Cream/therapeutic use , Administration, Topical , Nitriles/administration & dosage , Nitriles/adverse effects , Nitriles/therapeutic use , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Pyrazoles/therapeutic use , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/therapeutic use , Randomized Controlled Trials as Topic , Clinical Trials, Phase III as Topic
11.
Cell Mol Biol Lett ; 27(1): 60, 2022 Jul 26.
Article in English | MEDLINE | ID: mdl-35883021

ABSTRACT

The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway is involved in many immunological processes, including cell growth, proliferation, differentiation, apoptosis, and inflammatory responses. Some of these processes can contribute to cancer progression and neurodegeneration. Owing to the complexity of this pathway and its potential crosstalk with alternative pathways, monotherapy as targeted therapy has usually limited long-term efficacy. Currently, the majority of JAK-STAT-targeting drugs are still at preclinical stages. Meanwhile, a variety of plant polyphenols, especially quercetin, exert their inhibitory effects on the JAK-STAT pathway through known and unknown mechanisms. Quercetin has shown prominent inhibitory effects on the JAK-STAT pathway in terms of anti-inflammatory and antitumor activity, as well as control of neurodegenerative diseases. This review discusses the pharmacological effects of quercetin on the JAK-STAT signaling pathway in solid tumors and neurodegenerative diseases.


Subject(s)
Janus Kinase Inhibitors , Neoplasms , Neurodegenerative Diseases , Humans , Janus Kinase Inhibitors/pharmacology , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Janus Kinases/pharmacology , Neoplasms/drug therapy , Neurodegenerative Diseases/drug therapy , Quercetin/pharmacology , Quercetin/therapeutic use , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/metabolism , STAT Transcription Factors/pharmacology , Signal Transduction
12.
Brasília; CONITEC; jun. 2022.
Non-conventional in Portuguese | LILACS, Coleciona SUS | ID: biblio-1378051

ABSTRACT

CONTEXTO: Mielofibrose é uma neoplasia maligna rara que pode se desenvolver como doença primária, sendo uma doença mieloproliferativa crônica caracterizada pela falha da medula óssea e proliferação clonal de células mieloides associada com excesso de fibras de reticulina e/ou colágeno, e algum grau de atipia no megacariócito. O quadro clínico pode evoluir com esplenomegalia, anemia, sintomas constitucional (fadiga, sudorese noturna, febre), caquexia, dor óssea, infarto esplênico, prurido, trombose e sangramentos. A incidência na União Europeia e EUA é de 0,3 casos por 100.000 habitantes. Não há dados epidemiológicos robustos no Brasil. Ruxolitinibe é um inibidor seletivo das Janus Quinases associadas (JAKs) ­ JAK1 e JAK2. A desregulação da via JAK-STAT tem sido associada a vários tipos de câncer e aumento da proliferação e sobrevida de células malignas. TECNOLOGIA: Ruxolitinibe. PERGUNTA: O uso de ruxolitinibe no tratamento da mielofibrose risco intermediário-2 ou alto (classificação IPSS), em adultos, com contagem plaquetária acima de 100.000/mm3 é eficaz e seguro quando comprado ao


Subject(s)
Humans , Janus Kinases/antagonists & inhibitors , Primary Myelofibrosis/drug therapy , Unified Health System , Brazil , Cost-Benefit Analysis/economics
13.
Int J Mol Sci ; 23(9)2022 Apr 27.
Article in English | MEDLINE | ID: mdl-35563259

ABSTRACT

Canine atopic dermatitis (AD) is a common chronic inflammatory skin disorder resulting from imbalance between T lymphocytes. Current canine AD treatments use immunomodulatory drugs, but some of the dogs have limitations that do not respond to standard treatment, or relapse after a period of time. Thus, the purpose of this study was to evaluate the immunomodulatory effect of mesenchymal stem cells derived from canine adipose tissue (cASCs) and cASCs-derived extracellular vesicles (cASC-EVs) on AD. First, we isolated and characterized cASCs and cASCs-EVs to use for the improvement of canine atopic dermatitis. Here, we investigated the effect of cASCs or cASC-EVs on DNCB-induced AD in mice, before using for canine AD. Interestingly, we found that cASCs and cASC-EVs improved AD-like dermatitis, and markedly decreased levels of serum IgE, (49.6%, p = 0.002 and 32.1%, p = 0.016 respectively) epidermal inflammatory cytokines and chemokines, such as IL-4 (32%, p = 0.197 and 44%, p = 0.094 respectively), IL-13 (47.4%, p = 0.163, and 50.0%, p = 0.039 respectively), IL-31 (64.3%, p = 0.030 and 76.2%, p = 0.016 respectively), RANTES (66.7%, p = 0.002 and 55.6%, p = 0.007) and TARC (64%, p = 0.016 and 86%, p = 0.010 respectively). In addition, cASCs or cASC-EVs promoted skin barrier repair by restoring transepidermal water loss, enhancing stratum corneum hydration and upregulating the expression levels of epidermal differentiation proteins. Moreover, cASCs or cASC-EVs reduced IL-31/TRPA1-mediated pruritus and activation of JAK/STAT signaling pathway. Taken together, these results suggest the potential of cASCs or cASC-EVs for the treatment of chronic inflammation and damaged skin barrier in AD or canine AD.


Subject(s)
Cell- and Tissue-Based Therapy , Dermatitis, Atopic , Extracellular Vesicles , Inflammation , Mesenchymal Stem Cells , Pruritus , Adipose Tissue/metabolism , Animals , Cell- and Tissue-Based Therapy/methods , Cytokines/metabolism , Dermatitis, Atopic/therapy , Dogs , Extracellular Vesicles/metabolism , Inflammation/metabolism , Inflammation/therapy , Janus Kinases/antagonists & inhibitors , Janus Kinases/therapeutic use , Mesenchymal Stem Cells/metabolism , Mice , Pruritus/metabolism , Pruritus/therapy , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/therapeutic use , Signal Transduction , Skin/metabolism
14.
Brasília; CONITEC; mar. 2022.
Non-conventional in Portuguese | LILACS, BRISA/RedTESA | ID: biblio-1368853

ABSTRACT

INTRODUÇÃO: O baricitinbe, um imunomodulador que atua sobre a atividade da IL-6 (citocina pró-inflamatória), pode representar uma estratégia para o tratamento de pacientes com COVID-19 que tiveram comprometimento pulmonar devido a resposta hiperinflamátoria desencadeada pela tempestade de citocinas característica na infecção causada pelo vírus SARS-COV2. TECNOLOGIA: Baricitinibe (Olumiant®). EVIDÊNCIAS CLÍNICAS: Para seleção das evidências clínicas foi conduzida uma revisão sistemática da literatura em busca de ensaios clínicos randomizados (ECR), estudos observacionais (mundo real) e revisões sistemáticas que avaliassem os efeitos do baricitinibe como monoterapia ou associado aos cuidados usuais - definidos aqui como 'terapia padrão' (corticoesteróides sistêmicos, anticoagulantes, antimicrobianos/antivirais) no tratamento de pacientes adultos com COVID-19, hospitalizados e que necessitam de suplementação de oxigênio (máscara ou cateter nasal, alto fluxo de oxigênio ou ventilação não invasiva). As buscas eletrônicas foram realizadas nas bases de dados: the Cochrane Library, MEDLINE via Pubmed, Literatura Latino-Americana e do Caribe em Ciências da Saúde (LILACS), EMBASE e Centre for Reviews and Dissemination (CRD). O risco de viés dos estudos primários incluídos foi avaliado pelas ferramentas Risk of Bias versão 2 da Cochrane (para ECR) ou ROBINS-I (para estudos observacionais), e a qualidade metodológica das revisões sistemáticas foi avaliada pela ferramenta AMSTAR-2. A qualidade da evidência foi avaliada pelo sistema GRADE. Seis artigos foram incluídos na presente revisão, sendo dois deles referentes a um ensaio clínico randomizado (ECR), um estudo observacional e três revisões sistemáticas com meta-análise (RSMA), sendo uma


Subject(s)
Humans , Oxygen Inhalation Therapy/instrumentation , Janus Kinases/antagonists & inhibitors , Noninvasive Ventilation/instrumentation , SARS-CoV-2/drug effects , COVID-19/drug therapy , Immunosuppressive Agents/antagonists & inhibitors , Unified Health System , Brazil , Cost-Benefit Analysis/economics , Inpatients
15.
Cell Mol Biol Lett ; 27(1): 10, 2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35109786

ABSTRACT

The novel coronavirus disease 2019 (COVID-19) pandemic has spread worldwide, and finding a safe therapeutic strategy and effective vaccine is critical to overcoming severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, elucidation of pathogenesis mechanisms, especially entry routes of SARS-CoV-2 may help propose antiviral drugs and novel vaccines. Several receptors have been demonstrated for the interaction of spike (S) protein of SARS-CoV-2 with host cells, including angiotensin-converting enzyme (ACE2), ephrin ligands and Eph receptors, neuropilin 1 (NRP-1), P2X7, and CD147. The expression of these entry receptors in the central nervous system (CNS) may make the CNS prone to SARS-CoV-2 invasion, leading to neurodegenerative diseases. The present review provides potential pathological mechanisms of SARS-CoV-2 infection in the CNS, including entry receptors and cytokines involved in neuroinflammatory conditions. Moreover, it explains several neurodegenerative disorders associated with COVID-19. Finally, we suggest inflammasome and JaK inhibitors as potential therapeutic strategies for neurodegenerative diseases.


Subject(s)
COVID-19 Drug Treatment , Central Nervous System/drug effects , Inflammasomes/drug effects , Neurodegenerative Diseases/drug therapy , Receptors, Virus/genetics , SARS-CoV-2/drug effects , Virus Internalization/drug effects , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/therapeutic use , Basigin/genetics , Basigin/metabolism , COVID-19/genetics , COVID-19/metabolism , COVID-19/virology , Central Nervous System/metabolism , Central Nervous System/virology , Ephrins/genetics , Ephrins/metabolism , Gene Expression Regulation , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Immunologic Factors/therapeutic use , Inflammasomes/genetics , Inflammasomes/metabolism , Janus Kinase Inhibitors/therapeutic use , Janus Kinases/antagonists & inhibitors , Janus Kinases/genetics , Janus Kinases/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/virology , Neuropilin-1/genetics , Neuropilin-1/metabolism , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Signal Transduction
16.
Nat Commun ; 13(1): 925, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177622

ABSTRACT

Despite recent advances in cancer immunotherapy, certain tumor types, such as Glioblastomas, are highly resistant due to their tumor microenvironment disabling the anti-tumor immune response. Here we show, by applying an in-silico multidimensional model integrating spatially resolved and single-cell gene expression data of 45,615 immune cells from 12 tumor samples, that a subset of Interleukin-10-releasing HMOX1+ myeloid cells, spatially localizing to mesenchymal-like tumor regions, drive T-cell exhaustion and thus contribute to the immunosuppressive tumor microenvironment. These findings are validated using a human ex-vivo neocortical glioblastoma model inoculated with patient derived peripheral T-cells to simulate the immune compartment. This model recapitulates the dysfunctional transformation of tumor infiltrating T-cells. Inhibition of the JAK/STAT pathway rescues T-cell functionality both in our model and in-vivo, providing further evidence of IL-10 release being an important driving force of tumor immune escape. Our results thus show that integrative modelling of single cell and spatial transcriptomics data is a valuable tool to interrogate the tumor immune microenvironment and might contribute to the development of successful immunotherapies.


Subject(s)
Brain Neoplasms/immunology , Glioblastoma/immunology , Interleukin-10/metabolism , Myeloid Cells/metabolism , T-Lymphocytes/immunology , Adult , Aged , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Communication/immunology , Cell Line, Tumor , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Healthy Volunteers , Heme Oxygenase-1/metabolism , Humans , Immunotherapy/methods , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Male , Middle Aged , Neocortex/cytology , Neocortex/immunology , Neocortex/pathology , Primary Cell Culture , RNA-Seq , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Single-Cell Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tissue Culture Techniques , Tumor Escape , Tumor Microenvironment/immunology
17.
Toxicol Sci ; 186(2): 323-337, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35134999

ABSTRACT

Izencitinib (TD-1473), an oral, gut-selective pan-Janus kinase (JAK) inhibitor under investigation for treatment of inflammatory bowel diseases, was designed for optimal efficacy in the gastrointestinal tract while minimizing systemic exposures and JAK-related safety findings. The nonclinical safety of izencitinib was evaluated in rat and dog repeat-dose and rat and rabbit reproductive and developmental toxicity studies. Systemic exposures were compared with JAK inhibitory potency to determine effects at or above pharmacologic plasma concentrations (≥1× plasma average plasma concentration [Cave]:JAK 50% inhibitory concentration [IC50] ratio). In rats and dogs, 1000 and 30 mg/kg/day izencitinib, respectively, produced minimal systemic findings (ie, red/white cell changes) and low systemic concentrations (approximately 1× plasma Cave:JAK IC50 ratio) with an 8× nonclinical:clinical systemic area under the curve (AUC) margin compared with exposures at the highest clinically tested dose (300 mg, quaque die, once daily, phase 1 study in healthy volunteers). In dogs, it was possible to attain sufficient systemic exposures to result in immunosuppression characteristic of systemic JAK inhibition, but at high AUC margins (43×) compared with systemic exposures observed at the highest tested dose in humans. No adverse findings were observed in the gastrointestinal tract or systemic tissues. Izencitinib did not affect male or female fertility. Izencitinib did not affect embryonic development in rats and rabbits as commonly reported with systemic JAK inhibition, consistent with low maternal systemic concentrations (2-6× plasma Cave:JAK IC50 ratio, 10-33× nonclinical:clinical AUC margin) and negligible fetal exposures. In conclusion, the izencitinib gut-selective approach resulted in minimal systemic findings in nonclinical species at pharmacologic, clinically relevant systemic exposures, highlighting the impact of organ-selectivity in reducing systemic safety findings.


Subject(s)
Janus Kinases , Naphthyridines , Nitriles , Administration, Oral , Animals , Dogs , Embryonic Development/drug effects , Female , Healthy Volunteers , Humans , Inflammatory Bowel Diseases , Janus Kinases/antagonists & inhibitors , Male , Naphthyridines/pharmacology , Naphthyridines/toxicity , Nitriles/pharmacology , Nitriles/toxicity , Pregnancy , Rabbits , Rats , Reproduction/drug effects , Toxicity Tests
18.
Biochem J ; 479(3): 385-399, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35084016

ABSTRACT

The apelin receptor (APLNR) regulates many biological processes including metabolism, angiogenesis, circulating blood volume and cardiovascular function. Additionally, APLNR is overexpressed in various types of cancer and influences cancer progression. APLNR is reported to regulate tumor recognition during immune surveillance by modulating the IFN-γ response. However, the mechanism of APLNR cross-talk with intratumoral IFN-γ signaling remains unknown. Here, we show that activation of APLNR up-regulates IFN-γ signaling in melanoma cells through APLNR mediated ß-arrestin 1 but not ß-arrestin 2 recruitment. Our data suggests that ß-arrestin 1 directly interacts with STAT1 to inhibit STAT1 phosphorylation to attenuate IFN-γ signaling. The APLNR mutant receptor, I109A, which is deficient in ß-arrestins recruitment, is unable to enhance intratumoral IFN-γ signaling. While APLNR N112G, a constitutively active mutant receptor, increases intratumoral sensitivity to IFN-γ signaling by enhancing STAT1 phosphorylation upon IFN-γ exposure. We also demonstrate in a co-culture system that APLNR regulates tumor survival rate. Taken together, our findings reveal that APLNR modulates IFN-γ signaling in melanoma cells and suggest that APLNR may be a potential target to enhance the efficacy of immunotherapy.


Subject(s)
Apelin Receptors/physiology , Interferon-gamma/physiology , Janus Kinases/physiology , Melanoma/metabolism , Neoplasm Proteins/physiology , STAT1 Transcription Factor/physiology , Signal Transduction/physiology , beta-Arrestin 1/physiology , Apelin Receptors/antagonists & inhibitors , Apelin Receptors/chemistry , Apelin Receptors/genetics , Cell Line, Tumor , Cytotoxicity, Immunologic , HEK293 Cells , Humans , Janus Kinases/antagonists & inhibitors , Melanoma/immunology , Models, Molecular , Protein Interaction Mapping , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , T-Lymphocytes/immunology , beta-Arrestin 2/analysis
19.
Biochem Pharmacol ; 197: 114929, 2022 03.
Article in English | MEDLINE | ID: mdl-35065024

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disorder and the treatment involves the use of traditional and biological disease modifying anti-rheumatic drugs (DMARDs). Recent studies have shown JAK/STAT signaling pathway as potential target for the treatment of RA. Novel JAK/STAT inhibitors viz tofacitinib and baricitinib have been recently approved by FDA for RA treatment and have attained substantial importance. However, the discernible risks of thromboembolism, gastrointestinal (GIT) perforations, hepatotoxicity and serious infections including tuberculosis, herpes zoster associated with their administration cannot be overlooked. Furthermore, these are highly expensive which limits their application for a broader use. These limitations provide the basis of exploring novel JAK/STAT inhibitors of natural origin with increased tolerability, safety and cost-effectiveness. In this review we confer an account of various natural compounds/phytochemicals that have proved to be beneficial in attenuating inflammation in RA via modulation of JAK/STAT signaling pathway. Some of these natural compounds including resveratrol have clearly indicated biochemical and clinically significant therapeutic effects in ameliorating RA both in vivo and in clinical settings. We further discuss the physicochemical challenges of poor solubility and absorption coupled with the use of natural JAK/STAT inhibitors. We thereafter discuss and summarize various drug delivery systems (DDS) to confront the physicochemical limitations of natural JAK/STAT inhibitors with the aim to enhance the therapeutic efficacy. Overall the review unveils the potential of natural JAK/STAT inhibitors as a cost-effective approach in ameliorating RA without incorporating the risks of adverse repercussions, thus setting the stage for clinical exploration of these compounds that may possibly complement the present RA therapy.


Subject(s)
Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/drug therapy , Janus Kinase Inhibitors/administration & dosage , Janus Kinases/antagonists & inhibitors , Phytochemicals/administration & dosage , STAT Transcription Factors/antagonists & inhibitors , Arthritis, Rheumatoid/metabolism , Forecasting , Humans , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Treatment Outcome
20.
Ann Hematol ; 101(1): 131-137, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34625831

ABSTRACT

The Janus kinase inhibitor ruxolitinib is approved for the treatment of myelofibrosis (MF) and improved overall survival (OS) versus control therapy in the phase 3 COMFORT trials. The aim of this retrospective analysis was to examine the real-world impact of ruxolitinib on OS in patients with MF. The US Medicare Fee-for-Service claims database (parts A/B/D) was used to identify patients with ≥ 1 inpatient or ≥ 2 outpatient claims with an MF diagnosis (January 2010-December 2017). Eligible patients with MF were ≥ 65 years old (intermediate-1 or higher risk based on age). Patients were divided into 3 groups based on ruxolitinib approval status at diagnosis and ruxolitinib exposure: (1) preapproval, ruxolitinib-unexposed; (2) post-approval, ruxolitinib-unexposed; and (3) post-approval, ruxolitinib-exposed. In total, 1677 patients with MF were included (preapproval [all ruxolitinib-unexposed], n = 278; post-approval, n = 1399 [ruxolitinib-unexposed, n = 1127; ruxolitinib-exposed, n = 272]). Overall, median age was 78 years, and 39.8% were male. Among patients with valid death dates (preapproval, n = 119 [42.8%]; post-approval, ruxolitinib-unexposed, n = 382 [33.9%]; post-approval ruxolitinib-exposed, n = 54 [19.9%]), 1-year survival rates were 55.6%, 72.5%, and 82.3%, and median OS was 13.2 months, 44.4 months, and not reached, respectively. Risk of mortality was significantly lower post- versus preapproval regardless of exposure to ruxolitinib (ruxolitinib-unexposed: adjusted hazard ratio [HR], 0.67; ruxolitinib-exposed: adjusted HR, 0.36; P < 0.001 for both); post-approval, mortality risk was significantly lower in ruxolitinib-exposed versus ruxolitinib-unexposed patients (adjusted HR, 0.61; P = 0.002). Findings from this study complement clinical data of ruxolitinib in MF by demonstrating a survival benefit in a real-world setting.


Subject(s)
Janus Kinases/antagonists & inhibitors , Nitriles/therapeutic use , Primary Myelofibrosis/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Aged , Aged, 80 and over , Female , Humans , Male , Primary Myelofibrosis/epidemiology , Retrospective Studies , Survival Analysis , United States/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...