Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.143
Filtrer
1.
Clin Transl Sci ; 17(9): e70010, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39222377

RÉSUMÉ

Tepotinib is approved for the treatment of patients with non-small-cell lung cancer harboring MET exon 14 skipping alterations. While edema is the most prevalent adverse event (AE) and a known class effect of MET inhibitors including tepotinib, there is still limited understanding about the factors contributing to its occurrence. Herein, we apply machine learning (ML)-based approaches to predict the likelihood of occurrence of edema in patients undergoing tepotinib treatment, and to identify factors influencing its development over time. Data from 612 patients receiving tepotinib in five Phase I/II studies were modeled with two ML algorithms, Random Forest, and Gradient Boosting Trees, to predict edema AE incidence and severity. Probability calibration was applied to give a realistic estimation of the likelihood of edema AE. Best model was tested on follow-up data and on data from clinical studies unused while training. Results showed high performances across all the tested settings, with F1 scores up to 0.961 when retraining the model with the most relevant covariates. The use of ML explainability methods identified serum albumin as the most informative longitudinal covariate, and higher age as associated with higher probabilities of more severe edema. The developed methodological framework enables the use of ML algorithms for analyzing clinical safety data and exploiting longitudinal information through various covariate engineering approaches. Probability calibration ensures the accurate estimation of the likelihood of the AE occurrence, while explainability tools can identify factors contributing to model predictions, hence supporting population and individual patient-level interpretation.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Oedème , Apprentissage machine , Humains , Oedème/induit chimiquement , Femelle , Mâle , Adulte d'âge moyen , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Sujet âgé , Tumeurs du poumon/traitement médicamenteux , Essais cliniques de phase II comme sujet , Pyrimidines/effets indésirables , Pyrimidines/administration et posologie , Essais cliniques de phase I comme sujet , Adulte , Antinéoplasiques/effets indésirables , Inhibiteurs de protéines kinases/effets indésirables , Pipéridines , Pyridazines
2.
Vet Med Sci ; 10(5): e70039, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39239737

RÉSUMÉ

Trixacarus caviae is a sarcoptic mange mite infesting guinea pigs. Infestation in immunosuppressed animals produces severe dermatological problems, including alopecia, intense pruritus, hyperkeratosis and non-dermatological issues (e.g., seizures). Treatment options are limited and include topical application of macrocyclic lactones or amitraz or injectable administration of ivermectin or doramectin. Considering the severity of the disease and the challenging treatment, the present paper aimed to determine the efficacy of oral afoxolaner in a severe case of infestation with T. caviae in a pet guinea pig. One female guinea pig was referred to the New Companion Animal Clinic due to severe dermatological problems. A clinical evaluation was done, and skin scrapings were collected and examined under the microscope. Small mites were detected and morphologically identified as T. caviae. The animal was treated with a single oral dose of 2.50 mg/kg afoxolaner, and the lesions, presence/absence of mites and intensity of pruritus were evaluated periodically until 2 months post-treatment. A week after the medication, the lesions were milder, but pruritus was still present and was attributed to the healing process. Further examinations showed significant improvement with the complete remission of clinical signs and no mites at the microscopic examination after 4 weeks. Afoxolaner was safe and effective in this guinea pig for the treatment of T. caviae mange with no repetition needed.


Sujet(s)
Naphtalènes , Animaux , Cochons d'Inde , Femelle , Naphtalènes/administration et posologie , Naphtalènes/usage thérapeutique , Acarioses/médecine vétérinaire , Acarioses/traitement médicamenteux , Acarioses/parasitologie , Acaricides/usage thérapeutique , Acaricides/administration et posologie , Animaux de compagnie , Pyrimidines/administration et posologie , Pyrimidines/usage thérapeutique , Maladies des rongeurs/traitement médicamenteux , Maladies des rongeurs/parasitologie , Isoxazoles
3.
BMC Gastroenterol ; 24(1): 273, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39160459

RÉSUMÉ

BACKGROUND: Tofacitinib is an oral Janus kinase inhibitor for the treatment of ulcerative colitis (UC). We aimed to identify the safety and effectiveness of tofacitinib in patients with UC in routine clinical settings in Korea. METHODS: This open-label, observational, prospective, post-marketing surveillance study was conducted at 22 hospitals in the Republic of Korea. Patients with moderate to severe active UC who received tofacitinib were included and followed up for up to 52 weeks. Tofacitinib was administered at a dosage of 10 mg twice daily for at least 8 weeks, followed by 5 or 10 mg twice daily at the investigator's discretion based on clinical evaluation according to the approved Korean label. Safety including adverse events (AEs) and effectiveness including clinical remission, clinical response, and endoscopic mucosal healing were evaluated. Safety analysis set was defined as all patients registered for this study who received at least one dose of tofacitinib according to the approved Korean label and followed up for safety data. Effectiveness analysis set included patients in the safety analysis set who were evaluated for overall effectiveness assessment and excluded patients who had received tofacitinib less than 8 weeks. RESULTS: A total of 110 patients were enrolled, of whom 106 patients were included in the safety population. The median duration of treatment was 370 days and the treatment duration ranged from 16 to 684 days for the safety population. AEs occurred in 42 patients (39.6%). Serious AEs (SAEs) occurred in 7 patients (6.6%) and of them, there were 2 cases of serious infections. These serious infections were reported as Adverse Event of Special Interest (AESI) in this study and no other AESI were reported. There were no cases of death during the study period. Clinical remission rates were 40.0%, 46.7%, 57.6%, and 55.1% at 8, 16, 24, and 52 weeks, and clinical response rates were 77.8%, 87.9%, 56.6%, and 81.4% at each visit, respectively. Endoscopic mucosal healing rates were 58.7% at 16 weeks and 46.2% at 52 weeks. CONCLUSION: Tofacitinib was effective in Korean patients with moderate to severe active UC and the safety findings were consistent with the known safety profile of tofacitinib. This study confirmed the safety and effectiveness of tofacitinib in Korean patients with moderate to severe active UC in routine clinical settings. TRIAL REGISTRATION: This study is registered in the ClinicalTrials.gov under the identifier NCT04071405, registered on 28 August 2019.


Sujet(s)
Rectocolite hémorragique , Pipéridines , Surveillance post-commercialisation des produits de santé , Pyrimidines , Humains , Pyrimidines/effets indésirables , Pyrimidines/usage thérapeutique , Pyrimidines/administration et posologie , Pipéridines/effets indésirables , Pipéridines/usage thérapeutique , Pipéridines/administration et posologie , Rectocolite hémorragique/traitement médicamenteux , Mâle , Femelle , Adulte , Études prospectives , Adulte d'âge moyen , République de Corée , Pyrroles/effets indésirables , Pyrroles/usage thérapeutique , Pyrroles/administration et posologie , Résultat thérapeutique , Inhibiteurs des Janus kinases/effets indésirables , Inhibiteurs des Janus kinases/usage thérapeutique , Inhibiteurs des Janus kinases/administration et posologie , Sujet âgé , Inhibiteurs de protéines kinases/effets indésirables , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/administration et posologie , Jeune adulte , Induction de rémission
4.
Lancet Oncol ; 25(8): 989-1002, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39089305

RÉSUMÉ

BACKGROUND: Patients with EGFR-mutated non-small-cell lung cancer (NSCLC) and MET amplification as a mechanism of resistance to first-line osimertinib have few treatment options. Here, we report the primary analysis of the phase 2 INSIGHT 2 study evaluating tepotinib, a highly selective MET inhibitor, combined with osimertinib in this population. METHODS: This open-label, phase 2 study was conducted at 179 academic centres and community clinics in 17 countries. Eligible patients were aged 18 years or older with an Eastern Cooperative Oncology Group performance status of 0 or 1 and advanced or metastatic EGFR-mutated NSCLC of any histology, with MET amplification by tissue biopsy fluorescence in-situ hybridisation (FISH; MET gene copy number of ≥5 or MET-to-CEP7 ratio of ≥2) or liquid biopsy next-generation sequencing (MET plasma gene copy number of ≥2·3), following progression on first-line osimertinib. Patients received oral tepotinib 500 mg plus oral osimertinib 80 mg once daily. The primary endpoint was independently assessed objective response in patients with MET amplification by central FISH treated with tepotinib plus osimertinib with at least 9 months of follow-up. Safety was analysed in patients who received at least one study drug dose. This study is registered with ClinicalTrials.gov, NCT03940703 (enrolment complete). FINDINGS: Between Feb 13, 2020, and Nov 4, 2022, 128 patients (74 [58%] female, 54 [42%] male) were enrolled and initiated tepotinib plus osimertinib. The primary activity analysis population included 98 patients with MET amplification confirmed by central FISH, previous first-line osimertinib and at least 9 months of follow-up (median 12·7 months [IQR 9·9-20·3]). The confirmed objective response rate was 50·0% (95% CI 39·7-60·3; 49 of 98 patients). The most common treatment-related grade 3 or worse adverse events were peripheral oedema (six [5%] of 128 patients), decreased appetite (five [4%]), prolonged electrocardiogram QT interval (five [4%]), and pneumonitis (four [3%]). Serious treatment-related adverse events were reported in 16 (13%) patients. Deaths of four (3%) patients were assessed as potentially related to either trial drug by the investigator due to pneumonitis (two [2%] patients), decreased platelet count (one [1%]), respiratory failure (one [1%]), and dyspnoea (one [1%]); one death was attributed to both pneumonitis and dyspnoea. INTERPRETATION: Tepotinib plus osimertinib showed promising activity and acceptable safety in patients with EGFR-mutated NSCLC and MET amplification as a mechanism of resistance to first-line osimertinib, suggesting a potential chemotherapy-sparing oral targeted therapy option that should be further investigated. FUNDING: Merck (CrossRef Funder ID: 10.13039/100009945).


Sujet(s)
Acrylamides , Dérivés de l'aniline , Protocoles de polychimiothérapie antinéoplasique , Carcinome pulmonaire non à petites cellules , Récepteurs ErbB , Amplification de gène , Tumeurs du poumon , Mutation , Protéines proto-oncogènes c-met , Humains , Acrylamides/usage thérapeutique , Femelle , Mâle , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Protéines proto-oncogènes c-met/génétique , Adulte d'âge moyen , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Sujet âgé , Récepteurs ErbB/génétique , Récepteurs ErbB/antagonistes et inhibiteurs , Dérivés de l'aniline/usage thérapeutique , Dérivés de l'aniline/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Adulte , Pyrimidines/effets indésirables , Pyrimidines/usage thérapeutique , Pyrimidines/administration et posologie , Évolution de la maladie , Sujet âgé de 80 ans ou plus , Indoles , Pipéridines , Pyridazines
5.
Dermatol Online J ; 30(3)2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-39090037

RÉSUMÉ

Lichen planus is a chronic auto-inflammatory disease that primarily affects mucocutaneous regions. There are many variants of lichen planus including cutaneous, oral, nail, follicular, and erosive forms. Without any disease-specific treatment options, multi-variant lichen planus can be a challenging disease to manage. We present a 61-year-old woman with multivariant lichen planus that was refractory to numerous systemic and topical therapies. Subsequently, her cutaneous and vulvovaginal lesions improved with the use of oral baricitinib and the erosive oral lesions improved with topical ruxolitinib.


Sujet(s)
Azétidines , Lichen plan , Nitriles , Purines , Pyrazoles , Pyrimidines , Sulfonamides , Humains , Femelle , Pyrazoles/usage thérapeutique , Pyrazoles/administration et posologie , Nitriles/usage thérapeutique , Adulte d'âge moyen , Purines/usage thérapeutique , Purines/administration et posologie , Azétidines/usage thérapeutique , Azétidines/administration et posologie , Sulfonamides/usage thérapeutique , Sulfonamides/administration et posologie , Pyrimidines/usage thérapeutique , Pyrimidines/administration et posologie , Lichen plan/traitement médicamenteux , Lichen plan/anatomopathologie , Administration par voie orale
6.
Arch Dermatol Res ; 316(8): 566, 2024 Aug 24.
Article de Anglais | MEDLINE | ID: mdl-39180702

RÉSUMÉ

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.


Sujet(s)
Pipéridines , Pyrimidines , Rosacée , Transduction du signal , Adulte , Animaux , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , Administration par voie orale , Administration par voie topique , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/immunologie , Modèles animaux de maladie humaine , Inhibiteurs des Janus kinases/pharmacologie , Inhibiteurs des Janus kinases/usage thérapeutique , Inhibiteurs des Janus kinases/administration et posologie , Janus kinases/métabolisme , Janus kinases/antagonistes et inhibiteurs , Mastocytes/effets des médicaments et des substances chimiques , Mastocytes/immunologie , Mastocytes/métabolisme , Pipéridines/pharmacologie , Pipéridines/usage thérapeutique , Pipéridines/administration et posologie , Pyrimidines/pharmacologie , Pyrimidines/administration et posologie , Pyrimidines/usage thérapeutique , Pyrroles/pharmacologie , Pyrroles/administration et posologie , Rosacée/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Peau/effets des médicaments et des substances chimiques , Peau/métabolisme , Facteurs de transcription STAT/métabolisme , Facteurs de transcription STAT/antagonistes et inhibiteurs , Facteur de transcription STAT-1/métabolisme
7.
Eur J Cancer ; 209: 114241, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39096851

RÉSUMÉ

PURPOSE: The objective of this study was to determine the recommended Phase 2 dose (RP2D) of pevonedistat, a first in class inhibitor of NEDD8 activating enzyme, in combination with irinotecan (IRN) and temozolomide (TMZ) in children with cancer. METHODS: This Phase 1 study used a rolling 6 design to evaluate escalating doses of pevonedistat in combination with standard doses of IRN and TMZ in pediatric patients with recurrent/refractory solid or CNS tumors. During cycle 1, pevonedistat was administered intravenously on days 1, 8, 10, and 12, with IRN (IV, 50 mg/m2) and TMZ (orally, 100 mg/m2), on days 8-12 of a 28-day cycle. In subsequent cycles, pevonedistat was administered on days 1, 3, and 5, with IRN/TMZ on days 1-5 of a 21-day cycle. RESULTS: Thirty patients enrolled; all were eligible and evaluable for toxicity. Six patients each enrolled on pevonedistat dose levels (DL) 1 (15 mg/m2), 2 (20 mg/m2), 3 (25 mg/m2) and 4 (35 mg/m2) as well as an expanded pharmacokinetic (PK) cohort at DL4. The maximum tolerated dose (MTD) was not exceeded. 2/12 (17 %) patients treated at the RP2D (35 mg/m2) experienced a cycle 1 dose limiting toxicity (DLT). IRN is unlikely to affect the pharmacokinetics of pevonedistat. Two patients had a partial response and 6 patients had prolonged stable disease (> 6 cycles). CONCLUSIONS: Pevonedistat in combination with IRN/TMZ is well tolerated in children with solid or CNS tumors. The RP2D of pevonedistat is 35 mg/m2 on days 1, 3, 5 in combination with IRN/TMZ.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Cyclopentanes , Tumeurs , Pyrimidines , Humains , Femelle , Mâle , Enfant , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Adolescent , Tumeurs/traitement médicamenteux , Enfant d'âge préscolaire , Pyrimidines/administration et posologie , Pyrimidines/usage thérapeutique , Pyrimidines/pharmacocinétique , Pyrimidines/effets indésirables , Cyclopentanes/administration et posologie , Cyclopentanes/usage thérapeutique , Cyclopentanes/pharmacologie , Récidive tumorale locale/traitement médicamenteux , Irinotécan/administration et posologie , Irinotécan/usage thérapeutique , Témozolomide/administration et posologie , Dose maximale tolérée , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Jeune adulte , Protéine NEDD8
9.
AAPS PharmSciTech ; 25(7): 192, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39164485

RÉSUMÉ

Lipid-based formulations (LbFs) have demonstrated success in pharmaceutical applications; however, challenges persist in dissolving entire doses of the drug into defined liquid volumes. In this study, the temperature-induced supersaturation method was employed in LbF to address drug loading and pill burden issues. Supersaturated LbFs (super-LbF) were prepared using the temperature-induced supersaturation method, where the drug load is above its equilibrium solubility. Further, the influence of the drug's physicochemical and thermal characteristics on drug loading and their relevance with an apparent degree of supersaturation (aDS) was studied using two model drugs, ibrutinib and enzalutamide. All the prepared LbFs were evaluated in terms of physical stability, dispersion, and solubilization capacity, as well as pharmacokinetic assessments. Drug re-crystallization was observed in the lipid solution on long-term storage at higher aDS values of 2-2.5. Furthermore, high-throughput lipolysis studies demonstrated a significant decrease in drug concentration across all LbFs (regardless of drug loading) due to a decline in the formulation solvation capacity and subsequent generation of in-situ supersaturation. Further, the in vivo results demonstrated comparable pharmacokinetic parameters between conventional LbF and super-LbF. The short duration of the thermodynamic metastable state limits the potential absorption benefits. However, super-LbFs of Ibr and Enz showed superior profiles, with 1.7-fold and 5.2-fold increased drug exposure compared to their respective crystalline suspensions. In summary, this study emphasizes the potential of temperature-induced supersaturation in LbF for enhancing drug loading and highlights the intricate interplay between drug properties, formulation characteristics, and in vivo performance.


Sujet(s)
Adénine , Benzamides , Chimie pharmaceutique , Lipides , Nitriles , 3-Phényl-2-thiohydantoïne , Pipéridines , Solubilité , Température , Nitriles/composition chimique , Nitriles/administration et posologie , Pipéridines/composition chimique , Pipéridines/administration et posologie , Pipéridines/pharmacocinétique , Benzamides/composition chimique , Benzamides/pharmacocinétique , Adénine/analogues et dérivés , Adénine/composition chimique , Adénine/administration et posologie , 3-Phényl-2-thiohydantoïne/pharmacocinétique , 3-Phényl-2-thiohydantoïne/administration et posologie , Lipides/composition chimique , Animaux , Chimie pharmaceutique/méthodes , Préparation de médicament/méthodes , Mâle , Pyrimidines/pharmacocinétique , Pyrimidines/composition chimique , Pyrimidines/administration et posologie , Stabilité de médicament , Cristallisation/méthodes , Pyrazoles/composition chimique , Pyrazoles/pharmacocinétique , Pyrazoles/administration et posologie , Lipolyse/effets des médicaments et des substances chimiques , Rats
10.
Clin Transl Sci ; 17(8): e70018, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39189872

RÉSUMÉ

Myelofibrosis is a chronic myeloproliferative disorder characterized by bone marrow fibrosis, splenomegaly, anemia, and constitutional symptoms, with a median survival of ≈6 years from diagnosis. While currently approved Janus kinase (JAK) inhibitors (ruxolitinib, fedratinib) improve splenomegaly and symptoms, most can exacerbate myelofibrosis-related anemia, a negative prognostic factor for survival. Momelotinib is a novel JAK1/JAK2/activin A receptor type 1 (ACVR1) inhibitor approved in the US, European Union, and the UK and is the first JAK inhibitor indicated specifically for patients with myelofibrosis with anemia. Momelotinib not only addresses the splenomegaly and symptoms associated with myelofibrosis by suppressing the hyperactive JAK-STAT (signal transducer and activator of transcription) pathway but also improves anemia and reduces transfusion dependency through ACVR1 inhibition. The recommended dose of momelotinib is 200 mg orally once daily, which was established after review of safety, efficacy, pharmacokinetic, and pharmacodynamic data. Momelotinib is metabolized primarily by CYP3A4 and excreted as metabolites in feces and urine. Steady-state maximum concentration is 479 ng/mL (CV%, 61%), with a mean AUCtau of 3288 ng.h/mL (CV%, 60%); its major metabolite, M21, is active (≈40% of pharmacological activity of parent), with a metabolite-to-parent AUC ratio of 1.4-2.1. This review describes momelotinib's mechanism of action, detailing how the JAK-STAT pathway is involved in myelofibrosis pathogenesis and ACVR1 inhibition decreases hepcidin, leading to improved erythropoiesis. Additionally, it summarizes the pivotal studies and data that informed the recommended dosage and risk/benefit assessment.


Sujet(s)
, Humains , Myélofibrose primitive/traitement médicamenteux , Myélofibrose primitive/métabolisme , Benzamides/pharmacologie , Benzamides/pharmacocinétique , Benzamides/effets indésirables , Pyrimidines/pharmacocinétique , Pyrimidines/pharmacologie , Pyrimidines/usage thérapeutique , Pyrimidines/administration et posologie , Pyrimidines/effets indésirables , Animaux , Janus kinase 1/antagonistes et inhibiteurs , Janus kinase 1/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Kinase Janus-2/antagonistes et inhibiteurs , Kinase Janus-2/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/effets indésirables , Inhibiteurs de protéines kinases/pharmacocinétique , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/administration et posologie , Composés pontés
11.
Int J Rheum Dis ; 27(9): e15311, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39198040

RÉSUMÉ

INTRODUCTION: Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial inflammation, joint swelling, and pain involving multiple joints. While biologic disease-modifying antirheumatic drugs (bDMARDs) and targeted synthetic DMARDs (tsDMARDs) are popular treatments for RA, there is limited research on their combined use. This study examined a cohort of RA patients who demonstrated inadequate response to bDMARDs and subsequently initiated combination therapy with tofacitinib and bDMARDs, assessing both the efficacy and safety profile of this therapeutic approach. METHODS: In this study, we retrospectively collected the electronic medical records (EMR) of 62 adult patients with RA who were admitted to the Fourth Affiliated Hospital Zhejiang University School of Medicine between August 2018 and December 2022. All patients had received at least one bDMARD treatment for more than 3 months and still exhibited moderate-to-high disease activity. Tofacitinib 5 mg bid was added to their original biological treatment in 28 cases, and other 34 cases switched to another bDMARD or tsDMARD as control group. Treatment was continued for 24 weeks following the initiation of combination therapy. Changes in DAS28-ESR and ACR20, 50, 70 response rates at week 24 were collected and analyzed from baseline, while changes in C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) at weeks 4, 8, 12, 24 were also collected and analyzed. RESULTS: After 24 weeks of treatment, the DAS28-ESR score in combined treatment group decreased significantly from a baseline of 5.26 ± 0.90 (3.87-8.31) to 2.67 ± 0.86 (1.41-5.11), with remission achieved by 19 patients (67.9%) and low disease activity achieved by five patients (17.9%). The DAS28-ESR in the control group exhibited a decrease from 5.20 ± 0.77 (3.87-7.23) at baseline to 3.25 ± 1.29 (1.54-5.69). In all, 13 patients (38.2%) achieved remission, while another 11 patients (32.4%) achieved low disease activity. The ACR20, 50, 70 response rates were 85.71%, 75%, and 39.29% in the combined treatment group, whereas it were 75.0%, 53.57%, 21.43% in the control group. Additionally, both ESR and CRP levels decreased significantly during the course of treatment without any reported adverse events leading to discontinuation. CONCLUSION: Our findings offer some evidence, supporting the effectiveness and safety of combining bDMARD with JAKi tofacitinib in RA patients who have an inadequate response to bDMARD monotherapy. This combination effectively manages disease activity while maintaining a relatively low and manageable incidence of adverse events. Further prospective randomized controlled trials with large sample sizes are anticipated to provide evidence-based medical support.


Sujet(s)
Antirhumatismaux , Polyarthrite rhumatoïde , Association de médicaments , Pipéridines , Pyrimidines , Humains , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/diagnostic , Pyrimidines/effets indésirables , Pyrimidines/usage thérapeutique , Pyrimidines/administration et posologie , Mâle , Études rétrospectives , Femelle , Adulte d'âge moyen , Pipéridines/effets indésirables , Pipéridines/administration et posologie , Pipéridines/usage thérapeutique , Antirhumatismaux/effets indésirables , Antirhumatismaux/usage thérapeutique , Antirhumatismaux/administration et posologie , Résultat thérapeutique , Adulte , Sujet âgé , Inhibiteurs de protéines kinases/effets indésirables , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/administration et posologie , Facteurs temps , Produits biologiques/effets indésirables , Produits biologiques/usage thérapeutique , Produits biologiques/administration et posologie , Induction de rémission
13.
Lancet Oncol ; 25(9): 1231-1244, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39214106

RÉSUMÉ

BACKGROUND: CAPItello-291 is an ongoing phase 3 trial in which capivasertib-fulvestrant significantly improved progression-free survival versus placebo-fulvestrant in patients with hormone receptor-positive, HER2-negative advanced breast cancer who had relapse or disease progression during or after aromatase inhibitor treatment, in both the overall population and in patients with PIK3CA, AKT1, or PTEN-altered tumours. This study further explored patient-reported health-related quality of life (HRQOL), functioning, symptoms, and symptom tolerability in CAPItello-291. METHODS: This phase 3, randomised, double-blind, placebo-controlled trial, which was conducted across 193 hospitals and cancer centres in 19 countries, enrolled women with any menopausal status or men, aged ≥18 years (≥20 years in Japan), with hormone receptor-positive, HER2-negative locally advanced or metastatic breast cancer who had relapse or disease progression during or after treatment with an aromatase inhibitor, with or without previous cyclin-dependent kinase (CDK) 4 or 6 inhibitor therapy. Patients had an Eastern Cooperative Oncology Group/WHO performance score of 0 or 1 and could have received up to two previous lines of endocrine therapy and up to one previous line of chemotherapy for advanced disease. Patients were randomly assigned (1:1) using block randomisation (stratified according to the presence or absence of liver metastases, previous use of a CDK4/6 inhibitor [yes vs no], and geographical region) to receive oral capivasertib 400 mg (twice daily for 4 days, followed by 3 days off) plus intramuscular fulvestrant 500 mg (every 14 days for the first three injections, then every 28 days) or placebo with matching fulvestrant dosing. The dual primary endpoint of the trial was investigator-assessed progression-free survival assessed both in the overall population and among patients with PIK3CA, AKT1, or PTEN-altered tumours. The EORTC Quality of Life Questionnaire 30-item core module (QLQ-C30) and breast module (QLQ-BR23), Patient-Reported Outcomes version of the Common Terminology Criteria for Adverse Events (PRO-CTCAE), and Patient Global Impression of Treatment Tolerability (PGI-TT) questionnaires were used to assess patient-reported outcomes. Evaluation of EORTC QLQ-C30 and EORTC QLQ-BR23 were secondary endpoints and evaluation of PRO-CTCAE and PGI-TT were pre-defined exploratory endpoints, and these endpoints are the subject of analysis in this Article. Data were collected at baseline and prespecified timepoints. Patient-reported outcomes were analysed in all randomly assigned patients with an evaluable baseline assessment and at least one evaluable post-baseline assessment. Change from baseline was assessed using mixed model with repeated measures for EORTC QLQ-C30 and summarised for QLQ-BR23. Time to deterioration was described using the Kaplan-Meier method. PGI-TT and PRO-CTCAE responses were summarised at each treatment cycle. Patient-reported outcomes were not prospectively powered for statistical comparison. The trial is registered with ClinicalTrials.gov, NCT04305496. FINDINGS: Between June 2, 2020, and Oct 13, 2021, 901 patients were enrolled, of whom 708 patients were randomly assigned to receive capivasertib-fulvestrant (n=355) or placebo-fulvestrant (n=353). The median age of the patients was 59 years (IQR 51-67) in the capivasertib-fulvestrant group and 58 years (IQR 49-66) in the placebo-fulvestrant group. At data cutoff (Aug 15, 2022), the median duration of follow-up for progression-free survival in censored patients was 13·0 months (IQR 9·1-16·7) for capivasertib-fulvestrant and 12·7 months (IQR 2·0-16·4) for placebo-fulvestrant in the overall population. EORTC QLQ-C30 global health status/quality of life (GHS/QOL) scores were maintained from baseline and were similar between treatment groups throughout the study period (difference in mean change from baseline of -2·5 [95% CI -4·5 to -0·6] with capivasertib-fulvestrant vs -5·6 [-7·9 to -3·4] with placebo-fulvestrant; treatment difference 3·1 [95% CI 0·2 to 6·0]). Median time to deterioration in EORTC QLQ-C30 GHS/QOL was 24·9 months (95% CI 13·8 to not reached) in the capivasertib-fulvestrant group and 12·0 months (10·2 to 15·7) in the placebo-fulvestrant group (hazard ratio [HR] 0·70, 95% CI 0·53 to 0·92). Time to deterioration HRs for all EORTC QLQ-C30 and QLQ-BR23 subscale scores showed little difference between the treatment groups, except for diarrhoea, which was worse in the capivasertib-fulvestrant group than in the placebo-fulvestrant group (HR 2·75, 95% CI 2·01-3·81). In PRO-CTCAE symptom assessment, the proportion of patients reporting loose and watery stools "frequently" or "almost constantly" was 29% higher at cycle 1, day 15 in the capivasertib-fulvestrant group than in the placebo-fulvestrant group, decreasing at subsequent cycles. Other PRO-CTCAE-reported symptoms (rash, mouth or throat sores, itchy skin, and numbness or tingling in hands or feet) were absent or mild in most patients in both groups throughout treatment. According to the PGI-TT, most patients in both groups reported "not at all" or "a little bit" of bother from treatment side-effects. INTERPRETATION: Patient-reported outcomes from CAPItello-291 demonstrated that capivasertib-fulvestrant delayed time to deterioration of GHS/QOL and maintained other dimensions of HRQOL (except symptoms of diarrhoea) similarly to fulvestrant. With the clinical efficacy and manageable safety profile, these exploratory results further support the positive benefit-risk profile of capivasertib-fulvestrant in this population. FUNDING: AstraZeneca.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs du sein , Fulvestrant , Mesures des résultats rapportés par les patients , Pyrimidines , Qualité de vie , Récepteur ErbB-2 , Récepteurs des oestrogènes , Récepteurs à la progestérone , Humains , Femelle , Fulvestrant/usage thérapeutique , Fulvestrant/administration et posologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Méthode en double aveugle , Récepteur ErbB-2/métabolisme , Adulte d'âge moyen , Récepteurs des oestrogènes/métabolisme , Sujet âgé , Récepteurs à la progestérone/métabolisme , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Pyrimidines/administration et posologie , Pyrimidines/usage thérapeutique , Survie sans progression , Adulte , Pyrrolidines/administration et posologie , Pyrrolidines/usage thérapeutique , Pyrroles
14.
Clin Pharmacokinet ; 63(8): 1191-1204, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39127854

RÉSUMÉ

BACKGROUND AND OBJECTIVE: Overactivation of the PI3K/AKT pathway can occur in many cancers. Capivasertib is a potent, selective pan-AKT inhibitor. The objectives of this analysis were to develop a population pharmacokinetic model for capivasertib and to quantitatively assess the impact of intrinsic and extrinsic factors on the pharmacokinetics of capivasertib. METHODS: Pharmacokinetic data from four phase I and II studies were combined. Capivasertib was administered orally at a dose range of 80-800 mg twice daily over 28-day and 21-day cycles as monotherapy or in combination with paclitaxel or fulvestrant, using continuous dosing or one of two intermittent dosing schedules: either 4 days on, 3 days off (4/3) or 2 days on, 5 days off (2/5). Several models and approaches were tested for their ability to describe capivasertib disposition. The covariates assessed included dose, schedule, age, body weight, race, sex, creatinine clearance, hepatic function, renal function, smoking status, food effect, formulation, and concomitant use with paclitaxel, fulvestrant, cytochrome P450, family 3, subfamily A (CYP3A) inducers, CYP3A inhibitors and acid-reducing agents. RESULTS: A total of 3963 capivasertib plasma concentrations from 441 patients were included. Capivasertib pharmacokinetics was adequately described by a three-compartment model where the apparent clearance (CL/F) presented a moderate time-dependent and dose-dependent clearance. Following oral administration of multiple doses of capivasertib (400 mg twice daily; [4/3]), the initial CL/F was 62.2 L/h (between-subject variability 39.3%), and after approximately 120 hours, CL/F decreased by 18%. The effective half-life was 8.34 h. Steady state was predicted to be reached on every third and fourth dosing day each week from the second week with exposure levels that produced robust inhibition of AKT but not of other related kinases. The area under the plasma concentration-time curve and maximum plasma concentration of capivasertib were proportional between the dose levels of 80-480 mg after multiple doses but more than proportional beyond 480 mg. Schedule, age, race, sex, creatinine clearance, hepatic function, renal function, smoking status and concomitant use with fulvestrant, CYP3A inducers, CYP3A inhibitors or acid-reducing agents were not significant covariates for capivasertib pharmacokinetics. Concomitant use of paclitaxel, food effect and formulation statistically significantly affected capivasertib pharmacokinetics, but the effect was low. Body weight was statistically significantly related to capivasertib CL/F, with a 12% reduction in CL/F at steady state and a 14% increase in the area under the curve for 12 hours at steady state and maximum concentration at steady state at a lower body weight (47 kg vs 67 kg reference). CONCLUSIONS: Capivasertib pharmacokinetics showed moderate between-subject variability, and most covariates assessed had no significant impact. Body weight, dose, concomitant use of paclitaxel, food effect and formulation showed statistically significant effects. However, these were predicted to impact exposure to capivasertib by <20% and were not expected to be clinically relevant. Based on the population pharmacokinetics, no a priori dose adjustment is needed for intrinsic and extrinsic factors.


Sujet(s)
Modèles biologiques , Tumeurs , Humains , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Adulte , Pyrroles/pharmacocinétique , Pyrroles/administration et posologie , Paclitaxel/pharmacocinétique , Paclitaxel/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/pharmacocinétique , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Pyrimidines/pharmacocinétique , Pyrimidines/administration et posologie , Fulvestrant/pharmacocinétique , Fulvestrant/administration et posologie , Relation dose-effet des médicaments , Inhibiteurs de protéines kinases/pharmacocinétique , Inhibiteurs de protéines kinases/administration et posologie , Sujet âgé de 80 ans ou plus , Administration par voie orale , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/administration et posologie
15.
Curr Allergy Asthma Rep ; 24(9): 485-496, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39105881

RÉSUMÉ

PURPOSE OF REVIEW: To analyze the efficacy and safety of Janus kinase inhibitors (JAKi) in the treatment of pediatric AD. RECENT FINDINGS: Adolescents with moderate and severe atopic dermatitis (AD) need systemic therapies, as stated several recent practice guidelines. (JAKi) have shown their efficacy in the treatment of adult AD, however, there is a lack of information concerning efficacy and safety of their use in pediatric AD. We found that the JAKi's abrocitinib (ABRO), baricitinib (BARI), and upadacitinib (UPA), are all an effective treatment option with a very fast onset of action for adolescents with moderate-to-severe AD. BARI was not effective in children between 2 and 10 years with moderate-to-severe AD. Fortunately, major safety issues with JAKi in adolescents with AD have not been documented in the trials, so far, contrasting with the reports in adults with AD, where these events have very rarely occurred. There are some reports of herpes zoster (HZ) infection in adolescents on JAKi, but it is not a major safety concern. Acne is a relatively common AE with UPA in adolescents; however, it is responsive to standard treatment. This review will help the clinician to choose among the JAKi according to the needs and clinical features of patients with moderate and severe AD. In the following years, with the advent of new biologicals and JAKi, these therapies will fall into place in each phase of the evolution of patients with AD.


Sujet(s)
Eczéma atopique , Inhibiteurs des Janus kinases , Humains , Eczéma atopique/traitement médicamenteux , Inhibiteurs des Janus kinases/usage thérapeutique , Inhibiteurs des Janus kinases/effets indésirables , Inhibiteurs des Janus kinases/administration et posologie , Enfant , Adolescent , Purines/usage thérapeutique , Administration par voie orale , Azétidines/usage thérapeutique , Azétidines/administration et posologie , Composés hétérocycliques 3 noyaux/usage thérapeutique , Composés hétérocycliques 3 noyaux/effets indésirables , Pyrazoles/usage thérapeutique , Pyrazoles/administration et posologie , Pyrimidines/usage thérapeutique , Pyrimidines/administration et posologie , Pyrimidines/effets indésirables , Sulfonamides/usage thérapeutique , Sulfonamides/administration et posologie , Résultat thérapeutique
16.
Int. j. morphol ; 42(4): 984-990, ago. 2024. ilus, tab
Article de Anglais | LILACS | ID: biblio-1569276

RÉSUMÉ

SUMMARY: In this study we aimed to examine the effect of novel vasodilatory drug Riociguat co-administration along resveratrol to recover neurodegeneration in experimental stroke injury. For that purpose, thirty-five adult female rats were divided into five groups (Control, MCAO, MCAO + R, MCAO + BAY, MCAO + C) of seven animals in each. Animals in Control group did not expose to any application during the experiment and sacrificed at the end of the study. Rats in the rest groups exposed to middle cerebral artery occlusion (MCAO) induced ischemic stroke. MCAO + R group received 30 mg/kg resveratrol, and MCAO + BAY group received 10 mg/kg Riociguat. The MCAO + C group received both drugs simultaneously. The drugs were administered just before the reperfusion, and the additional doses were administered 24h, and 48h hours of reperfusion. All animals in this study were sacrificed at the 72nd hour of experiment. Total brains were received for analysis. Results of this experiment indicated that MCAO led to severe injury in cerebral structure. Bax, IL-6 and IL-1ß tissue levels were up-regulated, but anti-apoptotic Bcl-2 immunoexpression was suppressed (p<0.05). In resveratrol and Riociguat treated animals, the neurodegenerations and apoptosis and inflammation associated protein expressions were improved compared to MCAO group, but the most success was obtained in combined treatment exposed animals in MCAO + C group. This study indicated that the novel soluble guanylate stimulator Riociguat is not only a potent neuroprotective drug in MCAO induced stroke, but also synergistic administration of Riociguat along with resveratrol have potential to increase the neuroprotective effect of resveratrol in experimental cerebral stroke exposed rats.


En este estudio, nuestro objetivo fue examinar el efecto de la coadministración del nuevo fármaco vasodilatador Riociguat junto con resveratrol para recuperar la neurodegeneración en lesiones por ataques cerebrovasculares experimentales. Para ello, se dividieron 35 ratas hembras adultas en cinco grupos (Control, MCAO, MCAO + R, MCAO + BAY, MCAO + C) de siete animales en cada uno. Los animales del grupo control no fueron sometidos a ninguna aplicación durante el experimento y se sacrificaron al final del estudio. Las ratas de los grupos expuestas a la oclusión de la arteria cerebral media (MCAO) indujeron un ataque cerebrovascular isquémico. El grupo MCAO + R recibió 30 mg/kg de resveratrol y el grupo MCAO + BAY recibió 10 mg/kg de Riociguat. El grupo MCAO + C recibió ambos fármacos simultáneamente. Los fármacos se administraron antes de la reperfusión y las dosis adicionales se administraron a las 24 y 48 horas de la reperfusión. Todos los animales en este estudio fueron sacrificados a las 72 horas del experimento. Se recibieron cerebros totales para su análisis. Los resultados indicaron que la MCAO provocaba lesiones graves en la estructura cerebral. Los niveles tisulares de Bax, IL-6 e IL- 1ß estaban regulados positivamente, pero se suprimió la inmunoexpresión antiapoptótica de Bcl-2 (p <0,05). En los animales tratados con resveratrol y Riociguat, las neurodegeneraciones y las expresiones de proteínas asociadas a la apoptosis y la inflamación mejoraron en comparación con el grupo MCAO, sin embargo el mayor éxito se obtuvo en el tratamiento combinado de animales expuestos en el grupo MCAO + C. Este estudio indicó que el nuevo estimulador de guanilato ciclasa soluble Riociguat no solo es un fármaco neuroprotector potente en el ataque cerebrovascular inducido por MCAO, sino que también la administración sinérgica de Riociguat junto con resveratrol tiene el potencial para aumentar el efecto neuroprotector del resveratrol en ratas experimentales expuestas a un ataque cerebrovascular.


Sujet(s)
Animaux , Femelle , Rats , Pyrazoles/administration et posologie , Pyrimidines/administration et posologie , Accident vasculaire cérébral/traitement médicamenteux , Resvératrol/administration et posologie , Artériopathies oblitérantes , Test ELISA , Immunohistochimie , Interleukine-6/analyse , Apoptose/effets des médicaments et des substances chimiques , Neuroprotecteurs , Artère cérébrale moyenne , Accident vasculaire cérébral/anatomopathologie , Activateurs d'enzymes/administration et posologie , Modèles animaux , Association de médicaments , Interleukine-1 bêta/analyse , Guanylate cyclase/effets des médicaments et des substances chimiques , Inflammation
17.
Drug Dev Res ; 85(5): e22247, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39138857

RÉSUMÉ

Inflammatory diseases including rheumatoid arthritis are major health problems. Although different techniques and drugs are clinically available for the diagnosis and therapy of the disease, novel approaches regarding radiolabeled drug delivery systems are researched. Hence, in the present study, it was aimed to design, prepare, and characterize 99mTc-radiolabeled and tofacitinib citrate-encapsulated microsphere loaded poloxamer in situ gel formulations for the intra-articular treatment. Among nine different microsphere formulations, MS/TOFA-9 was chosen as the most proper one due to particle size, high encapsulation efficiency, and in vitro drug release behavior. Poloxamer 338 at a concentration of 15% was used to prepare in situ gel formulations. For intra-articular administration, microspheres were dispersed in an in situ gel containing 15% Poloxamer 338 and characterized in terms of gelation temperature, viscosity, rheological, mechanical, and spreadability properties. After the determination of the safe dose for MS/TOFA-9 and PLX-MS/TOFA-9 as 40 µL/mL in the cell culture study performed on healthy cells, the high anti-inflammatory effects were due to significant cellular inhibition of fibroblasts. In the radiolabeling studies with 99mTc, the optimum radiolabeling condition was determined as 200 ppm SnCl2 and 0.5 mg ascorbic acid, and both 99mTc-MS/TOFA-9 and 99mTc-PLX-MS/TOFA-9 exhibited high cellular binding capacity. In conclusion, although further in vivo experiments are required, PLX-MS/TOFA-9 was found to be a promising agent for intra-articular injection in rheumatoid arthritis.


Sujet(s)
Polyarthrite rhumatoïde , Chitosane , Gels , Microsphères , Pipéridines , Pyrimidines , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/imagerie diagnostique , Pyrimidines/composition chimique , Pyrimidines/administration et posologie , Pyrimidines/pharmacocinétique , Pipéridines/composition chimique , Pipéridines/administration et posologie , Pipéridines/pharmacocinétique , Chitosane/composition chimique , Humains , Technétium/composition chimique , Injections articulaires , Pyrroles/composition chimique , Pyrroles/administration et posologie , Animaux , Poloxamère/composition chimique , Taille de particule , Libération de médicament
19.
Arch Dermatol Res ; 316(8): 518, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39134884

RÉSUMÉ

Vitiligo, an autoimmune condition characterized by depigmented skin patches due to the loss of functional melanocytes, has been linked to dysregulation in the JAK-STAT signaling pathway, particularly in IFN-g signaling. The use of JAK inhibitors, such as ruxolitinib cream, a JAK1 and JAK2 inhibitor, presents a promising approach for vitiligo treatment. This study aims to systematically assess the effectiveness and safety of ruxolitinib cream in patients with vitiligo. We conducted a systematic review and meta-analysis following PRISMA guidelines to evaluate the efficacy and safety of ruxolitinib cream for the treatment of vitiligo. A comprehensive search of PubMed, Google Scholar, and Cochrane Library databases for randomized controlled trials (RCTs). Data selection, screening, extraction, and risk of bias assessment were meticulously performed. Statistical analysis was conducted using Review Manager Software, version 5.4, with significant heterogeneity addressed through appropriate methods. Our meta-analysis included 3 studies with 830 vitiligo patients. Significant improvements were observed in F-VASI, T-VASI, F-BSA, and T-BSA scores, with greater efficacy at 24 weeks compared to 12 weeks [MD -24.17, 95% CI (-31.78 to -16.56), P < 0.00001], [MD -14.12, 95% CI (-20.54 to -7.70); P < 0.0000], [MD -16.25, 95% CI (-22.20 to -10.31), P < 0.00001], [MD -9.19, 95% CI (-13.47 to -4.92); P < 0.00001]. Ruxolitinib showed increased risk ratios for F-VASI75, F-VASI90, and F-VASI50, indicating better outcomes with longer treatment durations [MD 2.9, 95% CI 1.88-4.49; P < 0.00001], [MD 4.66, 95% CI 2.09-10.39; P = 0.0002], [MD 2.53, 95% CI 1.84-3.46; P < 0.00001]. No significant differences were found in mild and moderate adverse events, while severe cases favored ruxolitinib. Placebo had a significant advantage in any adverse events, with no significant difference in drug-related adverse events. Serious adverse events did not significantly differ between groups. The findings strongly support the efficacy of ruxolitinib therapy in improving various parameters over time for treating vitiligo. However, thorough consideration of its safety profile, particularly concerning adverse events and potential side effects, is warranted. Further studies with larger sample sizes are needed to confirm these conclusions.


Sujet(s)
Nitriles , Pyrazoles , Pyrimidines , Vitiligo , Vitiligo/traitement médicamenteux , Humains , Pyrimidines/administration et posologie , Pyrimidines/usage thérapeutique , Pyrazoles/administration et posologie , Pyrazoles/effets indésirables , Pyrazoles/usage thérapeutique , Résultat thérapeutique , Crème pour la peau/administration et posologie , Inhibiteurs des Janus kinases/usage thérapeutique , Inhibiteurs des Janus kinases/administration et posologie , Inhibiteurs des Janus kinases/effets indésirables , Essais contrôlés randomisés comme sujet
20.
Arch Dermatol Res ; 316(7): 458, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38967866

RÉSUMÉ

BACKGROUND: Alopecia areata (AA) is an autoimmune pathology manifested by loss of hair. OBJECTIVE: To evaluate and compare the efficacy and safety of tofacitinib and azathioprine in patients with AA and variants. METHODS: In this double-blind randomized controlled trail (RCT) carried out at the Department of Dermatology, Medical Teaching Institute-Lady Reading Hospital (MTI-LRH), Peshawar, Pakistan, patients aged ≥ 12 years diagnosed with AA, alopecia totalis (AT) or alopecia universalis (AU) with minimum 50% scalp hair loss for a period ≥ 06 years were included. Patients were randomly assigned to receive oral tofacitinib 5 mg twice daily (Group I) or oral azathioprine 2 mg/kg body weight once daily (Group II). The primary endpoint was Severity of Alopecia Tool (SALT) score, evaluated at baseline and 06 months follow-up. Safety was consistently assessed during the study. RESULTS: A total of 104 patients underwent random allocation into either the tofacitinib group (n = 52) or the azathioprine group (n = 52). The mean (SD) age of patients was 20.23 (7.14) years and 22.26 (8.07) years, while the mean (SD) disease duration was 6.59 (4.01) years and 7.98 (4.40) years in in Group I and II, respectively. Overall, 40 (38.5%) patients were adolescents while 70 (67.3%) were male. 52 (50%) had AA, 37 (35.5%) had AT and 15 (14.5%) had AU. Mean baseline SALT score in tofacitinib group was 91.02 ± 10.21 and azathioprine group was 91.02 ± 10.63, which at 06 months follow-up improved to 14.1 ± 24.6 and 63.9 ± 33.9, respectively (difference, 11.5 points; 95% confidence interval, 38.3-61.3, p < 0.0001). Overall, no major adverse effects and no difference among the minor adverse effects in the two groups (04 adverse events for tofacitinib group and 08 for azathioprine group: p = 0.23) was observed. CONCLUSIONS: Efficacy of tofacitinib was significantly higher than azathioprine, whilst both drugs were well-tolerated in patients with AA and variants.


Sujet(s)
Pelade , Alopécie , Azathioprine , Pipéridines , Pyrimidines , Humains , Pyrimidines/administration et posologie , Pyrimidines/effets indésirables , Pyrimidines/usage thérapeutique , Pipéridines/administration et posologie , Pipéridines/effets indésirables , Pipéridines/usage thérapeutique , Mâle , Pelade/traitement médicamenteux , Pelade/diagnostic , Méthode en double aveugle , Femelle , Azathioprine/administration et posologie , Azathioprine/effets indésirables , Azathioprine/usage thérapeutique , Adolescent , Adulte , Jeune adulte , Alopécie/traitement médicamenteux , Résultat thérapeutique , Inhibiteurs de protéines kinases/effets indésirables , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/usage thérapeutique , Administration par voie orale , Enfant , Pyrroles/administration et posologie , Pyrroles/effets indésirables , Indice de gravité de la maladie , Immunosuppresseurs/effets indésirables , Immunosuppresseurs/administration et posologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE