yingweiwo

Baricitinib phosphate (LY-3009104, INCB-028050, Olumiant)

Alias: trade name Olumiant; LY3009104 phosphate; LY-3009104; LY 3009104; INCB028050 phosphate; INCB-028050; INCB 028050; Baricitinib phosphate;Baricitinib phosphate; 1187595-84-1; Baricitinib (phosphate); Baricitinib phosphate salt; INCB-28050; XIB47S8NNB;
Cat No.:V0096 Purity: ≥98%
Baricitinib phosphate, the phosphate salt of Baricitinib which is also known as LY3009104 or INCB028050 or trade name Olumiant, is apotent,selective, ATP competitive and orally bioavailable inhibitor of tyrosine-protein kinase JAK1 or JAK2.
Baricitinib phosphate (LY-3009104, INCB-028050, Olumiant)
Baricitinib phosphate (LY-3009104, INCB-028050, Olumiant) Chemical Structure CAS No.: 1187595-84-1
Product category: JAK
This product is for research use only, not for human use. We do not sell to patients.
Size Price Stock Qty
2mg
5mg
50mg
100mg
250mg
500mg
1g
Other Sizes

Other Forms of Baricitinib phosphate (LY-3009104, INCB-028050, Olumiant):

  • Baricitinib (LY-3009104, INCB-028050)
Official Supplier of:
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Top Publications Citing lnvivochem Products
Purity & Quality Control Documentation

Purity: ≥98%

Product Description

Baricitinib phosphate, the phosphate salt of Baricitinib which is also known as LY3009104 or INCB028050 or trade name Olumiant, is a potent, selective, ATP competitive and orally bioavailable inhibitor of tyrosine-protein kinase JAK1 or JAK2. It is an FDA approved drug for the treatment of rheumatoid arthritis (RA) in the United States. In vitro, it is able to inhibit JAK1 and JAK2 with IC50 values in the low nanomolar range of 5.9 and 5.7 nM, respectively, while it displays low inhibitory activity for JAK3 and moderate activity for TYK2. Baricitinib inhibits intracellular signaling of several proinflammatory cytokines such as IL-6 and IL-23 at concentrations<50 nM. JAK signaling is central to a number of fundamental processes including the generation of RBCs. On Nov 20, 2020, the U.S. Food and Drug Administration also issued an emergency use authorization (EUA) for baricitinib to be used in combination with remdesivir, for the treatment of suspected or laboratory confirmed COVID-19 in hospitalized adults and pediatric patients two years of age or older requiring supplemental oxygen, invasive mechanical ventilation, or extracorporeal membrane oxygenation (ECMO). On June 13, 2022, Olumiant (baricitinib) was approved by FDA to treat adult patients with severe alopecia areata, a disorder that often appears as patchy baldness and affects more than 300,000 people in the U.S. each year. Today’s action marks the first FDA approval of a systemic treatment (i.e. treats the entire body rather than a specific location) for alopecia areata.

Biological Activity I Assay Protocols (From Reference)
Targets
JAK2 (IC50 = 5.7 nM); JAK1 (IC50 = 5.9 nM); Tyk2 (IC50 = 53nM); JAK3 (IC50 = 560nM)
ln Vitro
Baricitinib phosphate (INCB028050 phosphate) is a strong inhibitor of JAK signaling and function in experiments conducted on cells. Baricitinib has IC50 values of 44 nM and 40 nM, respectively, which prevent IL-6-stimulated phosphorylation of the canonical substrate STAT3 (pSTAT3) and the subsequent generation of the chemokine MCP-1 in PBMCs. INCB028050 also suppresses pSTAT3 activated by IL-23 (IC50=20 nM) in isolated naive T-cells. Significantly, at an IC50 value of 50 nM, this inhibition stopped the production of two pathogenic cytokines (IL-17 and IL-22) by Th17 cells, a subset of helper T cells with observable inflammatory and pathogenic characteristics. On the other hand, when evaluated at concentrations up to 10 μM, the structurally similar but ineffective JAK1/2 inhibitors INCB027753 and INCB029843 have no discernible effect in any of these assay systems[1].
ln Vivo
Treatment with baricitinib phosphate (INCB028050 phosphate) suppresses the rise in hind paw volumes during the first two weeks of treatment by 50% at a dose of 1 mg/kg and by more than 95% at doses of 3 or 10 mg/kg when compared to the vehicle. It is feasible for animals exhibiting a noticeable improvement in swelling to have >100% inhibition because baseline paw volume measurements are obtained on treatment day 0 in animals with substantial symptoms of disease[1]. When compared to mice given with vehicle control, mice treated with baricitinib (0.7 mg/day) show significantly less inflammation as measured by H&E staining, less CD8 infiltration, and less MHC class I and class II expression. When compared to mice treated with a vehicle control, the number of CD8+NKG2D+ cells, which are important disease effectors in alopecia areata (AA) in humans and animals, is significantly reduced in mice treated with baricitinib[2].
Enzyme Assay
Enzyme assays were performed using a homogeneous time-resolved fluorescence assay with recombinant epitope tagged kinase domains (JAK1, 837-1142; JAK2, 828-1132; JAK3, 718-1124; Tyk2, 873-1187) or full-length enzyme (cMET and Chk2) and peptide substrate. Each enzyme reaction was performed with or without test compound (11-point dilution), JAK, cMET, or Chk2 enzyme, 500 nM (100 nM for Chk2) peptide, ATP (at the Km specific for each kinase or 1 mM), and 2.0% DMSO in assay buffer. The calculated IC50 value is the compound concentration required for inhibition of 50% of the fluorescent signal. Additional kinase assays were performed at Cerep using standard conditions at 200 nM. Enzymes tested included: Abl, Akt1, AurA, AurB, CDC2, CDK2, CDK4, CHK2, c-kit, EGFR, EphB4, ERK1, ERK2, FLT-1, HER2, IGF1R, IKKα, IKKβ, JNK1, Lck, MEK1, p38α, p70S6K, PKA, PKCα, Src, and ZAP70[1].
Cell Assay
Cellular assays[1]
Human PBMCs were isolated by leukapheresis followed by Ficoll-Hypaque centrifugation. For the determination of IL-6–induced MCP-1 production, PBMCs were plated at 3.3 × 105 cells per well in RPMI 1640 + 10% FCS in the presence or absence of various concentrations of INCB028050. Following preincubation with compound for 10 min at room temperature, cells were stimulated by adding 10 ng/ml human recombinant IL-6 to each well. Cells were incubated for 48 h at 37°C, 5% CO2. Supernatants were harvested and analyzed by ELISA for levels of human MCP-1. The ability of INCB028050 to inhibit IL-6–induced secretion of MCP-1 is reported as the concentration required for 50% inhibition (IC50). Proliferation of Ba/F3-TEL-JAK3 cells was performed over 3 d using Cell-Titer Glo following standard assay conditions. For the determination of IL-23–induced IL-17 and IL-22, PBMCs were maintained in RPMI 1640 medium supplemented with 10% FBS, 2 mM l-glutamine, 100 μg/ml streptomycin, and 100 U/ml penicillin. T cells were activated by culturing with anti-CD3 and anti-CD28 Abs. After 2 d, the cells were washed and recultured with IL-23 (100 ng/ml), IL-2 (10 ng/ml) and various concentrations of INCB028050. Cells were incubated for an additional 4 d at 37°C, then supernatants were collected, and secretion of IL-17 and IL-22 were measured by ELISA. The ability of INCB028050 to inhibit IL-23–induced secretion of IL-17 and IL-22 is reported as the concentration required for 50% inhibition (IC50).
Phospho-STAT3 analysis[1]
Isolated cells.[1]
For analysis of phospho-STAT3 in human PBMCs or PHA-stimulated T cells, cell extracts were prepared after 10−15 min preincubation with different concentrations of INCB028050 and stimulation of cells for 15 min with IL-6 (100 ng/ml), IL-12 (20 ng/ml), or IL-23 (100 ng/ml). The extracts were then analyzed for phosphorylated STAT3 by using a phospho-STAT3 specific ELISA.
Whole blood.[1]
Blood drawn from rats was collected into heparinized tubes and then aliquoted into microfuge tubes (0.3 ml per sample). In stimulation experiments, INCB028050 at various concentrations was added for 10 min prior to stimulation with human IL-6 (100 ng/ml) for 15 min at 37°C. RBCs were lysed using hypotonic conditions. WBCs were then quickly pelleted and lysed to make total cellular extracts. The extracts were analyzed for phosphorylated STAT3 by using a phospho-STAT3–specific ELISA. Blood from animals that were dosed with INCB028050 was drawn at various times after INCB028050 administration and processed as described above.
Animal Protocol
In vivo experiments[1]
Animals were housed in a barrier facility accredited by the Association for Assessment and Accreditation of Laboratory Animal Care International. All of the procedures were conducted in accordance with the U.S. Public Health Service Policy on Humane Care and Use of Laboratory Animals and with Incyte Animal Care and Use Committee guidelines. Animals were fed standard rodent chow and provided with water ad libitum.
Pharmacokinetics.[1]
Female rats (n = 6 per gender per group) were given a dose of 10 mg/kg INCB028050 suspended in 0.5% methylcellulose and given by oral gavage at 10 ml/kg. The first three rats were bled at 0 (predose), 2, 8, and 24 h, and the second three rats were bled 1, 4, and 12 h after dosing. EDTA was used as the anticoagulant, and samples were centrifuged to obtain plasma. An analytical method for the quantification of INCB028050 has been developed and used to analyze samples from toxicology studies. The method combines a protein precipitation extraction with 10% methanol in acetonitrile and LC/MS/MS analysis. The method has demonstrated a linear assay range 1–5000 nM using 0.1 ml of study samples. Data were processed using Analyst 1.3.1. A standard curve was determined from peak area ratio versus concentration using a weighted linear regression (1/x2).
View More

Rat adjuvant-induced arthritis.[1]
Adjuvant-induced arthritis was elicited in rats according to established methods. Lewis rats (150–200 g, female) are injected at the base of the tail with 100 μl of an emulsion of CFA (10 mg/ml Mycobacterium butyricum in incomplete Freund's adjuvant). Rats exhibited signs of inflammation within 2 wk of the injection of CFA. Each rat paw was scored following visual observation using a rating of 0–3, (0 = normal; 1 = redness and minimal swelling of digits; 2 = moderate swelling of the digits and/or paw; 3 = severe swelling of digits and/or paw). Individual animal paw scores are combined and recorded as a sum of all four paws and groups means of these totals are reported. Percent inhibition in clinical score/severity is calculated using the following formula: In addition, a plethysmometer was used to measure paw volumes taken at baseline and study termination. At the termination of the experiment, paws were removed from euthanized rats for histologic analyses. Treatment was initiated when significant signs of disease were noted, and groups of animals were sorted so that mean scores would be equivalent—usually occurring 2 wk after adjuvant injection. Graphs reflect endpoints collected only immediately prior to and after therapy was initiated (treatment day 0). Groups consisted of six animals, and statistical differences between treatment and vehicle controls were assessed using two-tailed Student t tests or ANOVA with a Dunnett’s test when appropriate.


Collagen-induced arthritis.[1]
DBA/1j mice (4–5-wk old males) were purchased from The Jackson Laboratory (Bar Harbor, ME). The model was established as described with minor modifications. Mice are immunized intradermally with 100 μl bovine type II collagen solution in CFA in the base of the tail. Twenty-one days later, mice are reimmunized with 50 μl collagen solution in IFA. Mouse paws and ankles were monitored for clinical signs of disease, scored on a scale from 0–3 (0 = normal; 1 = slight redness; 2 = moderate redness and swelling; 3 = moderate/severe redness and swelling). In the experiments performed in this study, treatment began when all animals had at least one affected paw and groups randomized to contain similar mean scores. Each group contained six animals. Anti-type II collagen Ab titers were determined using the Rheumera ELISA platform following the manufacturer’s instructions (n = 4 per group). Serum samples were diluted 1:100,000 and frozen prior to analysis. Two-tailed Student t tests were used to compare individual treatment groups to controls.
Anti-collagen Ab-induced arthritis.[1]
BALB/c mice (7–8-wk-old, female) were purchased from Charles River Laboratories. The model was initiated as described with minor modifications. Mice were injected with 200 μl arthogenic anti-collagen Ab. Two days later, mice were injected i.p. with LPS (Escherichia coli-derived, 25 μg) and treatment was initiated the following day (n = 5 per group). Scoring of mice was similar to that described above in the collagen-induced arthritis model. Differences in clinical scores at study termination (last day shown) were analyzed for significance using a Student two-sided t test. Hematalogic parameters were measured using a Bayer Advia120. Two-tailed Student t tests were used to compare individual treatment groups to controls.

Dissolved in 5% dimethyl acetamide, 0.5% methocellulose; 180 mg/kg/day; Oral gavage
JAK2V617F-driven mouse model
References
[1]. Fridman JS, et al. Selective inhibition of JAK1 and JAK2 is efficacious in rodent models of arthritis: preclinical characterization of INCB028050. J Immunol. 2010 May 1;184(9):5298-307.
[2]. Jabbari A, et al. Reversal of Alopecia Areata Following Treatment With the JAK1/2 Inhibitor Baricitinib. EBioMedicine. 2015 Feb 26;2(4):351-5
Additional Infomation
Inhibiting signal transduction induced by inflammatory cytokines offers a new approach for the treatment of autoimmune diseases such as rheumatoid arthritis. Kinase inhibitors have shown promising oral disease-modifying antirheumatic drug potential with efficacy similar to anti-TNF biologics. Direct and indirect inhibition of the JAKs, with small molecule inhibitors like CP-690,550 and INCB018424 or neutralizing Abs, such as the anti-IL6 receptor Ab tocilizumab, have demonstrated rapid and sustained improvement in clinical measures of disease, consistent with their respective preclinical experiments. Therefore, it is of interest to identify optimized JAK inhibitors with unique profiles to maximize therapeutic opportunities. INCB028050 is a selective orally bioavailable JAK1/JAK2 inhibitor with nanomolar potency against JAK1 (5.9 nM) and JAK2 (5.7 nM). INCB028050 inhibits intracellular signaling of multiple proinflammatory cytokines including IL-6 and IL-23 at concentrations <50 nM. Significant efficacy, as assessed by improvements in clinical, histologic and radiographic signs of disease, was achieved in the rat adjuvant arthritis model with doses of INCB028050 providing partial and/or periodic inhibition of JAK1/JAK2 and no inhibition of JAK3. Diminution of inflammatory Th1 and Th17 associated cytokine mRNA levels was observed in the draining lymph nodes of treated rats. INCB028050 was also effective in multiple murine models of arthritis, with no evidence of suppression of humoral immunity or adverse hematologic effects. These data suggest that fractional inhibition of JAK1 and JAK2 is sufficient for significant activity in autoimmune disease models. Clinical evaluation of INCB028050 in RA is ongoing.[1]
Background: Alopecia areata (AA) is an autoimmune disease resulting in hair loss with devastating psychosocial consequences. Despite its high prevalence, there are no FDA-approved treatments for AA. Prior studies have identified a prominent interferon signature in AA, which signals through JAK molecules.[2]
Methods: A patient with AA was enrolled in a clinical trial to examine the efficacy of baricitinib, a JAK1/2 inhibitor, to treat concomitant CANDLE syndrome. In vivo, preclinical studies were conducted using the C3H/HeJ AA mouse model to assess the mechanism of clinical improvement by baricitinib.[2]
Findings: The patient exhibited a striking improvement of his AA on baricitinib over several months. In vivo studies using the C3H/HeJ mouse model demonstrated a strong correlation between resolution of the interferon signature and clinical improvement during baricitinib treatment.[2]
Interpretation: Baricitinib may be an effective treatment for AA and warrants further investigation in clinical trials.[2]
Keywords: Alopecia areata; Autoimmune disease; Autoinflammatory; Baricitinib; CANDLE syndrome; Gene expression profiling; Interferon gamma; JAK inhibitor.[2]
These protocols are for reference only. InvivoChem does not independently validate these methods.
Physicochemical Properties
Molecular Formula
C16H20N7O6PS
Molecular Weight
469.41
Exact Mass
469.09332
Elemental Analysis
C, 40.94; H, 4.29; N, 20.89; O, 20.45; P, 6.60; S, 6.83
CAS #
1187595-84-1
Related CAS #
Baricitinib;1187594-09-7
PubChem CID
44231848
Appearance
Typically exists as white to yellow solids at room temperature
LogP
1.19
tPSA
216.51
SMILES
S(C([H])([H])C([H])([H])[H])(N1C([H])([H])C(C([H])([H])C#N)(C1([H])[H])N1C([H])=C(C2=C3C([H])=C([H])N([H])C3=NC([H])=N2)C([H])=N1)(=O)=O.P(=O)(O[H])(O[H])O[H]
InChi Key
FBPOWTFFUBBKBB-UHFFFAOYSA-N
InChi Code
InChI=1S/C16H17N7O2S.H3O4P/c1-2-26(24,25)22-9-16(10-22,4-5-17)23-8-12(7-21-23)14-13-3-6-18-15(13)20-11-19-14;1-5(2,3)4/h3,6-8,11H,2,4,9-10H2,1H3,(H,18,19,20);(H3,1,2,3,4)
Chemical Name
(1-(Ethylsulfonyl)-3-(4-(7H-pyrrolo(2,3-d)pyrimidin-4-yl)-1H-pyrazol-1-yl)azetidin-3-yl)ethanenitrile phosphate
Synonyms
trade name Olumiant; LY3009104 phosphate; LY-3009104; LY 3009104; INCB028050 phosphate; INCB-028050; INCB 028050; Baricitinib phosphate;Baricitinib phosphate; 1187595-84-1; Baricitinib (phosphate); Baricitinib phosphate salt; INCB-28050; XIB47S8NNB;
HS Tariff Code
2934.99.9001
Storage

Powder      -20°C    3 years

                     4°C     2 years

In solvent   -80°C    6 months

                  -20°C    1 month

Note: Please store this product in a sealed and protected environment, avoid exposure to moisture.
Shipping Condition
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
Solubility Data
Solubility (In Vitro)
DMSO: >30 mg/mL
Water: N/A
Ethanol:N/A
Solubility (In Vivo)
Solubility in Formulation 1: ≥ 2.08 mg/mL (4.43 mM) (saturation unknown) in 10% DMSO + 40% PEG300 + 5% Tween80 + 45% Saline (add these co-solvents sequentially from left to right, and one by one), clear solution.
For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 20.8 mg/mL clear DMSO stock solution to 400 μL PEG300 and mix evenly; then add 50 μL Tween-80 to the above solution and mix evenly; then add 450 μL normal saline to adjust the volume to 1 mL.
Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH₂ O to obtain a clear solution.

Solubility in Formulation 2: 2.08 mg/mL (4.43 mM) in 10% DMSO + 90% (20% SBE-β-CD in Saline) (add these co-solvents sequentially from left to right, and one by one), suspension solution; with ultrasonication.
For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 20.8 mg/mL clear DMSO stock solution to 900 μL of 20% SBE-β-CD physiological saline solution and mix evenly.
Preparation of 20% SBE-β-CD in Saline (4°C,1 week): Dissolve 2 g SBE-β-CD in 10 mL saline to obtain a clear solution.

View More

Solubility in Formulation 3: ≥ 2.08 mg/mL (4.43 mM) (saturation unknown) in 10% DMSO + 90% Corn Oil (add these co-solvents sequentially from left to right, and one by one), clear solution.
For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 20.8 mg/mL clear DMSO stock solution to 900 μL of corn oil and mix evenly..


Solubility in Formulation 4: 0.5% CMC+0.25% Tween 80:30mg/mL

 (Please use freshly prepared in vivo formulations for optimal results.)
Preparing Stock Solutions 1 mg 5 mg 10 mg
1 mM 2.1303 mL 10.6517 mL 21.3033 mL
5 mM 0.4261 mL 2.1303 mL 4.2607 mL
10 mM 0.2130 mL 1.0652 mL 2.1303 mL

*Note: Please select an appropriate solvent for the preparation of stock solution based on your experiment needs. For most products, DMSO can be used for preparing stock solutions (e.g. 5 mM, 10 mM, or 20 mM concentration); some products with high aqueous solubility may be dissolved in water directly. Solubility information is available at the above Solubility Data section. Once the stock solution is prepared, aliquot it to routine usage volumes and store at -20°C or -80°C. Avoid repeated freeze and thaw cycles.

Calculator

Molarity Calculator allows you to calculate the mass, volume, and/or concentration required for a solution, as detailed below:

  • Calculate the Mass of a compound required to prepare a solution of known volume and concentration
  • Calculate the Volume of solution required to dissolve a compound of known mass to a desired concentration
  • Calculate the Concentration of a solution resulting from a known mass of compound in a specific volume
An example of molarity calculation using the molarity calculator is shown below:
What is the mass of compound required to make a 10 mM stock solution in 5 ml of DMSO given that the molecular weight of the compound is 350.26 g/mol?
  • Enter 350.26 in the Molecular Weight (MW) box
  • Enter 10 in the Concentration box and choose the correct unit (mM)
  • Enter 5 in the Volume box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 17.513 mg appears in the Mass box. In a similar way, you may calculate the volume and concentration.

Dilution Calculator allows you to calculate how to dilute a stock solution of known concentrations. For example, you may Enter C1, C2 & V2 to calculate V1, as detailed below:

What volume of a given 10 mM stock solution is required to make 25 ml of a 25 μM solution?
Using the equation C1V1 = C2V2, where C1=10 mM, C2=25 μM, V2=25 ml and V1 is the unknown:
  • Enter 10 into the Concentration (Start) box and choose the correct unit (mM)
  • Enter 25 into the Concentration (End) box and select the correct unit (mM)
  • Enter 25 into the Volume (End) box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 62.5 μL (0.1 ml) appears in the Volume (Start) box
g/mol

Molecular Weight Calculator allows you to calculate the molar mass and elemental composition of a compound, as detailed below:

Note: Chemical formula is case sensitive: C12H18N3O4  c12h18n3o4
Instructions to calculate molar mass (molecular weight) of a chemical compound:
  • To calculate molar mass of a chemical compound, please enter the chemical/molecular formula and click the “Calculate’ button.
Definitions of molecular mass, molecular weight, molar mass and molar weight:
  • Molecular mass (or molecular weight) is the mass of one molecule of a substance and is expressed in the unified atomic mass units (u). (1 u is equal to 1/12 the mass of one atom of carbon-12)
  • Molar mass (molar weight) is the mass of one mole of a substance and is expressed in g/mol.
/

Reconstitution Calculator allows you to calculate the volume of solvent required to reconstitute your vial.

  • Enter the mass of the reagent and the desired reconstitution concentration as well as the correct units
  • Click the “Calculate” button
  • The answer appears in the Volume (to add to vial) box
In vivo Formulation Calculator (Clear solution)
Step 1: Enter information below (Recommended: An additional animal to make allowance for loss during the experiment)
Step 2: Enter in vivo formulation (This is only a calculator, not the exact formulation for a specific product. Please contact us first if there is no in vivo formulation in the solubility section.)
+
+
+

Calculation results

Working concentration mg/mL;

Method for preparing DMSO stock solution mg drug pre-dissolved in μL DMSO (stock solution concentration mg/mL). Please contact us first if the concentration exceeds the DMSO solubility of the batch of drug.

Method for preparing in vivo formulation:Take μL DMSO stock solution, next add μL PEG300, mix and clarify, next addμL Tween 80, mix and clarify, next add μL ddH2O,mix and clarify.

(1) Please be sure that the solution is clear before the addition of next solvent. Dissolution methods like vortex, ultrasound or warming and heat may be used to aid dissolving.
             (2) Be sure to add the solvent(s) in order.

Clinical Trial Information
NCT Number Recruitment interventions Conditions Sponsor/Collaborators Start Date Phases
NCT01398475 Completed Has Results Drug: LY3009104 Chronic Inflammatory Disorder
Arthritis, Rheumatoid
Eli Lilly and Company July 2011 Phase 1
NCT05452564 Recruiting Drug: Baricitinib 2 MG Oral Tablet Human Immunodeficiency Virus William Tyor May 18, 2023 Phase 2
NCT04640168 Completed Has Results Drug: Baricitinib Drug: Dexamethasone Recurrent Glioma
Refractory Glioma
National Institute of Allergy and
Infectious Diseases (NIAID)
December 2, 2020 Phase 3
Biological Data
  • Baricitinib phosphate

    Cellular activity of INCB028050.J Immunol.2010 May 1;184(9):5298-307.

  • Baricitinib phosphate

    Anti-inflammatory and DMARD activity of once daily INCB028050 in rats with established disease in the adjuvant arthritis model.J Immunol.2010 May 1;184(9):5298-307.

  • Baricitinib phosphate

    Suppression of delayed-type hypersensitivity by INCB028050.J Immunol.2010 May 1;184(9):5298-307.

  • Baricitinib phosphate

    INCB028050 is efficacious and well tolerated independently of effects on humoral immunity.J Immunol.2010 May 1;184(9):5298-307.

  • Baricitinib phosphate

    INCB028050 improves clinical and histologic signs of disease in the murine CIA model.

Contact Us