The largest database of trusted experimental protocols

Ruxolitinib

Ruxolitinib: A potent and selective Janus kinase (JAK) inhibitor used in the treatment of myelofibrosis, polycythemia vera, and other myeloproliferative neoplasms.
It works by blocking the activity of JAK1 and JAK2, key enzymes involved in the signaling pathways that drive the proliferation and survival of malignant blood cells.
Ruxolitinib has been shown to improve symptoms, reduce spleen size, and prolong overall survival in patients with these rare and serious blood disorders.
It is an important therapeutic option for managing the complicaitons of myeloproliferative neoplasms and improving quality of life for affected individuals.

Most cited protocols related to «Ruxolitinib»

This randomized, double-blind, placebo-controlled phase 3 trial was conducted at 89 sites in the United States, Australia, and Canada. Patients were randomized 1:1 to receive oral ruxolitinib phosphate tablets or matched placebo. The starting dose of ruxolitinib was 15 mg or 20 mg twice daily, depending on baseline platelet count (100 to 200×109/l or >200×109/l, respectively). The dose was adjusted for lack of efficacy or excess toxicity per protocol (Appendix). Unblinding of therapy and crossover from placebo to ruxolitinib was permitted for protocol-defined worsening splenomegaly (Appendix). The prospectively defined data cutoff occurred when half the patients remaining in the study completed the week 36 visit, and all completed the week 24 evaluation or discontinued treatment. Data for placebo-treated patients after crossover are not included in these analyses, except for the intent-to-treat (ITT) analysis of overall survival.
The primary endpoint was the proportion of patients achieving a ≥35% reduction in spleen volume from baseline to week 24, measured by magnetic resonance imaging (MRI) or computed tomography. Secondary endpoints included duration of maintenance of spleen volume reduction, proportion of patients with ≥50% reduction in Total Symptom Score (TSS) from baseline to week 24 using the modified Myelofibrosis Symptom Assessment Form (MFSAF) v2.0 diary (Appendix),10 ,11 change in TSS from baseline to week 24, and overall survival. The overall survival analysis was updated at the time of a planned data cutoff 4 months after the primary analysis. Patients completed the MFSAF every night; this electronic diary evaluated, on a scale of 0 (absent) to 10 (worst imaginable), night sweats, itching, abdominal discomfort, pain under the ribs on the left side, feeling of fullness (early satiety), muscle/bone pain, and inactivity. TSS was the sum of individual symptom scores, excluding inactivity. Exploratory endpoints included changes in body weight and JAK2V617F allele burden, achievement of transfusion independence,12 (link) and additional patient-reported outcomes (Appendix).
The study was designed to enroll 240 patients, providing 97% power to detect a treatment difference in spleen volume response at a 2-sided alpha level of 0.05 assuming ≥30% response rate for ruxolitinib and ≤10% response rate for placebo. Analyses were conducted in accordance with intent-to-treat (ITT) principles. For all applicable variables, however, patients with missing baseline values were excluded from analyses of change and percent change from baseline. In analyses of change from baseline to week 24, patients who discontinued or crossed over before week 24 were counted as nonresponders (for response measures of spleen volume reduction and symptom improvement). Comparative secondary efficacy variables were tested in a fixed-sequence-testing procedure at an alpha level of 0.05. Durability of spleen response and survival were analyzed using the Kaplan-Meier method. The statistical analysis plan is posted on NEJM.org.
This study was funded by Incyte Corporation. The first author (S.V.) and a coauthor (V.S.) wrote the initial draft of the manuscript. Medical writing assistance with an early draft was provided by Daniel Hutta, Ph.D., of Articulate Science, LLC, and funded by Incyte Corporation. All coauthors contributed to subsequent drafts and decided to submit for publication. Data were analyzed at Incyte Corporation (W.S.). All authors vouch for the accuracy and completeness of reported data and for fidelity of this report to the protocol.
Publication 2012
Abdomen Alleles BAD protein, human Blood Transfusion Bones Crossing Over, Genetic Human Body Myalgia Pain Patients Placebos Platelet Counts, Blood Primary Myelofibrosis Ribs ruxolitinib ruxolitinib phosphate Satiation Spleen Sweat Symptom Assessment X-Ray Computed Tomography

Protocol full text hidden due to copyright restrictions

Open the protocol to access the free full text link

Publication 2020
A549 Cells ACE2 protein, human Amino Acids, Essential Cells Glucose Glutamine HEPES Homo sapiens Infection Interferon, beta Proteins Pyruvate ruxolitinib SARS-CoV-2 Sodium Sulfoxide, Dimethyl Vero Cells
Experimental procedures (A21013, A37715, and A16315) were approved by the IACUC at Mayo Clinic. Twenty two-month-old C57BL/6 male mice were obtained from the National Institute on Aging (NIA). INK-ATTAC+/- transgenic mice were generated and genotyped as previously described (Baker et al., 2011 (link)). Briefly, JLK and TT conceived the idea of clearing senescent cells to test if this improves healthspan and devised the experimental strategy of making transgenic mice with a senescence-activated promoter driving the ATTAC drug-inducible suicide gene and GFP to selectively eliminate and identify senescent cells at any time postnatally. The INK-ATTAC mice were produced and phenotyped at Mayo Clinic through a collaboration among the Kirkland, J. van Deursen, and N. LeBrasseur labs. They were bred onto a C57BL/6 background in the JVD lab. KOJ in the Kirkland lab then bred them onto a 50:50 BALB/c:C57BL/6 background, genotyped mice to select INK-ATTAC heterozygotes, and aged them to 18 months. Controls for the INK-ATTAC experiments were INK-ATTAC-null; 50:50 BALB/c:C57BL/6 background mice raised in parallel. Mice were maintained under a 12 hour light and 12 hour dark cycle at 24°C with free access to food (standard mouse diet, Lab Diet 5053, St. Louis, MO, USA) and water in a pathogen-free facility. For drug treatment, ruxolitinib was dissolved in DMSO and then mixed with food. In addition to regular food, each mouse was fed a small amount of food (0.5g) containing ruxolitinib 60 mg/kg (drug/body weight) or DMSO daily. During the treatment, all mice consumed the drug-containing food completely every day. For AP20187 (10mg/kg) treatment, drug was administered by i.p. injection for three consecutive days, with 14 days between treatments. Intermittent clearance of senescent cells with AP20187 was used based on our recent finding that senolytics are effective when administered intermittently (Zhu et al., 2015 (link)).
Full text: Click here
Publication 2015
AP20187 Body Weight Cellular Senescence Diet Food Genes Heterozygote Institutional Animal Care and Use Committees Light Males Mice, Inbred C57BL Mice, Laboratory Mice, Transgenic pathogenesis Pharmaceutical Preparations ruxolitinib Senolytics Sulfoxide, Dimethyl

Protocol full text hidden due to copyright restrictions

Open the protocol to access the free full text link

Publication 2016
Animals Animals, Laboratory Bioluminescent Measurements BLOOD Bone Marrow Cells Cloning Vectors Complete Blood Count Females Flow Cytometry Homo sapiens Hypersensitivity INCB-018424 inhibitors Institutional Animal Care and Use Committees Internal Ribosome Entry Sites Janus Kinase 1 Leukemia Luciferases Mice, Inbred C57BL Mus Pharmaceutical Preparations Phosphates Pre-B Lymphocytes Proteins Retroviridae Rodent ruxolitinib Spleen Tail Veins
Sodium valproate was from Sigma (St. Louis, MO). Neratinib was supplied by Puma Biotechnology Inc. (Los Angeles, CA). Sorafenib tosylate, dasatinib, ruxolitinib, dabrafenib, trametinib and sildenafil were from Selleckchem (Houston TX). Trypsin-EDTA, DMEM, RPMI, penicillin-streptomycin were purchased from GIBCOBRL (GIBCOBRL Life Technologies, Grand Island, NY). All “H” series NSCLC lines were purchased from the ATCC and were not further validated beyond that claimed by ATCC. Cells were re-purchased every ~6 months. ADOR cells were a gift to the Dent lab from a female NSCLC patient. Spiky ovarian cancer cells were kindly provided by Dr. Karen Paz (Champions Oncology, NJ). Commercially available validated short hairpin RNA molecules to knock down RNA / protein levels were from Qiagen (Valencia, CA) (Supplementary Figure 24). Control IgG, anti-PD-1 and anti-CTLA4 endotoxin-free antibodies were purchased from Bio-X cell (West Lebanon, NH). Reagents and performance of experimental procedures were described in refs: 1, 24-28, 45, 46.
Full text: Click here
Publication 2017
anti-endotoxin antibodies Cells Cytotoxic T-Lymphocyte Antigen 4 dabrafenib Dasatinib Edetic Acid Neoplasms neratinib Non-Small Cell Lung Carcinoma Ovarian Cancer Patients Penicillins Proteins Puma ruxolitinib Short Hairpin RNA Sildenafil Sodium Valproate Sorafenib Streptomycin trametinib Trypsin Woman

Most recents protocols related to «Ruxolitinib»

Protocol full text hidden due to copyright restrictions

Open the protocol to access the free full text link

Publication 2024
Ruxolitinib (R-6688, LC Laboratories) was formulated into Nutra-gel diet (F5769-Kit, Bio-Serv) at a concentration of 1 g/kg. Mice were acclimated to untreated Nutra-gel diet (replaced daily) alongside normal rodent chow for 7 days. Mice were provided with Ruxolitinib diet or Nutra-gel diet ad libitum (replaced daily) for 2 days prior to infection and continued throughout infection study period. In experiments where mice were treated with Amphotericin B during infection, Ruxolitinib diet was added on day 18 post-infection and continued throughout the study period.
Full text: Click here
Publication Preprint 2024
Ruxolitinib (MedChem Express, cat#HY-50856) was dissolved in DMSO for a stock concentration of 60 mg/ml. Prior to injection, 10 µl of this stock solution was dissolved in 200 µl saline. 10 µl DMSO dissolved in 200 µl saline was used in controls. 200 µl of the solution was delivered daily through oral gavage followed by dmPGE2 by intradermal injection.
Full text: Click here
Publication 2024
Ruxolitinib (INCB18424, MedChemExpress) was dissolved in PEG300/dextrose 5% in a ratio 1:3 (PEG/dex) and given by oral gavage at a dosage of 30 mg/kg two times daily starting from day -1 to day 13 after allo-HCT. The ROCK1/2-inhibitor Y-27632 was diluted and given as previously described from day 4 to day 13. The control group received PEG/dex and PBS alone.
Full text: Click here
Publication 2024
Clinical response was evaluated at 1 month, 3 months, 6 months, and 12 months after the start of ruxolitinib therapy. If a new immunosuppressive medication (ISM) was given after the start of ruxolitinib treatment, the response assessment was discontinued. Complete remission (CR) was defined as the resolution of all symptoms of cGvHD without initiation of new or additional ISM during treatment with ruxolitinib. Partial remission (PR) was defined as an improvement of at least one organ grade without progression of cGvHD in other organs, whereas mixed response (MR) was defined as an improvement in one organ, while progression occurred elsewhere. Progressive disease (PD) was defined as progression of at least one organ site without any improvements in other sites. Stable organ involvement without any changes in grading was classified as stable disease (SD). For evaluation of predictive markers, patients were divided into "responder" (CR, PR) and "nonresponder" (MR, SD, PD, and additional ISM) categories.
Failure-free survival (FFS) was defined as absence of relapse of the underlying disease or NRM, and no addition of further ISM. Calculation of overall response rates (ORRs) was based on an intention-to-treat analysis. If a patient did not complete the entire follow-up period of 12 months, the respective patient was excluded from ORR and FFS calculations from the first follow-up time point that had not been completed (i.e., 1 month, 3 months, 6 months, or 12 months) onward. To assess infectious adverse events (AEs) and hematological toxicities, the Common Terminology Criteria for Adverse Events version 5.0 (CTCAE 5.0) was used.
Publication 2024

Top products related to «Ruxolitinib»

Sourced in United States, Germany, United Kingdom, China
Ruxolitinib is a selective and potent inhibitor of Janus-associated kinases (JAK) 1 and 2. It is used as a research tool in laboratory settings to study the role of JAK signaling in various biological processes.
Sourced in United States, China
Ruxolitinib is a chemical compound used in laboratory research. It is a selective and potent inhibitor of Janus kinase (JAK) enzymes, particularly JAK1 and JAK2. Ruxolitinib is commonly used as a research tool to study the role of JAK signaling pathways in various cellular processes and disease models.
Sourced in United States, France
Ruxolitinib is a Janus kinase (JAK) inhibitor. It inhibits the activity of the JAK enzymes, which are involved in the signaling pathways of various cytokines and growth factors.
Sourced in United States
Ruxolitinib is a lab equipment product manufactured by Cayman Chemical. It is a Janus kinase (JAK) inhibitor that targets the JAK1 and JAK2 enzymes.
Sourced in United States, Germany, United Kingdom, China, Italy, Sao Tome and Principe, France, Macao, India, Canada, Switzerland, Japan, Australia, Spain, Poland, Belgium, Brazil, Czechia, Portugal, Austria, Denmark, Israel, Sweden, Ireland, Hungary, Mexico, Netherlands, Singapore, Indonesia, Slovakia, Cameroon, Norway, Thailand, Chile, Finland, Malaysia, Latvia, New Zealand, Hong Kong, Pakistan, Uruguay, Bangladesh
DMSO is a versatile organic solvent commonly used in laboratory settings. It has a high boiling point, low viscosity, and the ability to dissolve a wide range of polar and non-polar compounds. DMSO's core function is as a solvent, allowing for the effective dissolution and handling of various chemical substances during research and experimentation.
Sourced in United States
Ruxolitinib is a chemical compound used in laboratory research. It functions as a selective and potent inhibitor of the Janus kinase (JAK) enzymes, particularly JAK1 and JAK2. Ruxolitinib is commonly used in experiments investigating cellular signaling pathways and their modulation.
Sourced in United States, China, United Kingdom, Germany, Australia, Japan, Canada, Italy, France, Switzerland, New Zealand, Brazil, Belgium, India, Spain, Israel, Austria, Poland, Ireland, Sweden, Macao, Netherlands, Denmark, Cameroon, Singapore, Portugal, Argentina, Holy See (Vatican City State), Morocco, Uruguay, Mexico, Thailand, Sao Tome and Principe, Hungary, Panama, Hong Kong, Norway, United Arab Emirates, Czechia, Russian Federation, Chile, Moldova, Republic of, Gabon, Palestine, State of, Saudi Arabia, Senegal
Fetal Bovine Serum (FBS) is a cell culture supplement derived from the blood of bovine fetuses. FBS provides a source of proteins, growth factors, and other components that support the growth and maintenance of various cell types in in vitro cell culture applications.
Sourced in Switzerland, United States
Ruxolitinib is a pharmaceutical product developed by Novartis. It is a lab equipment designed for specific research applications. The core function of Ruxolitinib is to serve as a tool for scientific investigations, without any interpretation or extrapolation on its intended use.
Sourced in United States
Ruxolitinib is a laboratory product manufactured by Merck Group. It is a tyrosine kinase inhibitor that targets the JAK-STAT signaling pathway. Ruxolitinib is used in research applications to study cellular signaling processes.
Sourced in United States, Germany
Tofacitinib is a chemical compound used in laboratory research. It is a Janus kinase (JAK) inhibitor, a class of drugs that block the activity of one or more of the Janus kinase enzymes. Tofacitinib is commonly used in cell-based assays and in vivo studies to investigate the role of JAK signaling in various biological processes.

More about "Ruxolitinib"

Ruxolitinib is a potent and selective Janus kinase (JAK) inhibitor that has been widely used in the treatment of myelofibrosis, polycythemia vera, and other myeloproliferative neoplasms (MPNs).
These rare and serious blood disorders are characterized by the uncontrolled proliferation and survival of malignant blood cells.
Ruxolitinib works by blocking the activity of JAK1 and JAK2, which are key enzymes involved in the signaling pathways that drive the growth and survival of these malignant cells.
By inhibiting these enzymes, Ruxolitinib has been shown to effectively improve symptoms, reduce spleen size, and prolong overall survival in patients with MPNs.
In addition to Ruxolitinib, other compounds like DMSO and FBS have been utilized in MPN research and treatment.
DMSO, or dimethyl sulfoxide, is a commonly used solvent that can enhance the solubility and delivery of Ruxolitinib and other drugs.
FBS, or fetal bovine serum, is a nutrient-rich medium that is often used to support the growth and survival of cell cultures in laboratory studies.
Tofacitinib, another JAK inhibitor, has also been investigated for the treatment of MPNs and other inflammatory conditions.
While Ruxolitinib and Tofacitinib share a similar mechanism of action, they may have different pharmacokinetic profiles and selectivity for different JAK isoforms, which can impact their efficacy and safety in different clinical settings.
By leveraging the insights gained from the MeSH term description and the Metadescription, researchers can optimize their Ruxolitinib research using AI-driven protocol comparisons and identify the most effective products and procedures.
This can help enhance the reproducibility and accuracy of their studies, ultimately leading to improved patient outcomes and a better understanding of these complex blood disorders.