C16H17N7O2S
1187594-09-7
Alopecia Areata (AA), Coronavirus Disease 2019 (COVID‑19), Severe Atopic Dermatitis, Moderate Atopic dermatitis, Moderate, active Rheumatoid arthritis, Severe, active Rheumatoid arthritis
Baricitinib, marketed under the brand name Olumiant®, is an orally administered small molecule that functions as a selective and reversible inhibitor of Janus kinase (JAK) 1 and JAK2.[1] Developed by Incyte and licensed to Eli Lilly and Company, it represents a significant immunomodulatory agent in modern therapeutics.[3] The core mechanism of Baricitinib involves the targeted disruption of the JAK-Signal Transducer and Activator of Transcription (STAT) signaling pathway. This pathway is a critical conduit for numerous pro-inflammatory cytokines and growth factors implicated in the pathophysiology of various autoimmune diseases and the hyperinflammatory state observed in severe cases of Coronavirus Disease 2019 (COVID-19).[5]
The therapeutic applications of Baricitinib are diverse, with key approvals for moderately to severely active rheumatoid arthritis (RA), severe alopecia areata (AA), and for hospitalized patients with COVID-19 requiring supplemental oxygen or more advanced respiratory support.[2] Furthermore, it has received approval in Europe for atopic dermatitis and juvenile idiopathic arthritis (JIA).[5] Clinical trials have substantiated its efficacy, demonstrating significant improvements in patient-reported outcomes for RA, substantial hair regrowth in severe AA, and a noteworthy reduction in mortality for patients with severe COVID-19.[9]
Despite its demonstrated benefits, the use of Baricitinib requires careful consideration of its safety profile. The U.S. Food and Drug Administration (FDA) has mandated a boxed warning, a class-wide alert for JAK inhibitors, highlighting the risks of serious infections, all-cause mortality, malignancy, major adverse cardiovascular events (MACE), and thrombosis.[7] Consequently, Baricitinib is positioned as a highly effective but complex therapeutic agent, demanding meticulous patient selection and a comprehensive understanding of its multifaceted benefit-risk profile to ensure its appropriate and safe use.
Baricitinib is a synthetic organic compound classified chemically as a pyrrolopyrimidine, containing pyrazole, azetidine, sulfonamide, and nitrile functional groups.[1] Its formal International Union of Pure and Applied Chemistry (IUPAC) name is 2-[1-ethylsulfonyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]azetidin-3-yl]acetonitrile.[1] During its development and in research literature, it has been referred to by several identifiers, including the development codes INCB-028050 and LY-3009104.[5] Its globally recognized brand name is Olumiant®.[2]
Baricitinib is an achiral small molecule with the molecular formula C16H17N7O2S and a molecular weight of approximately 371.42 g/mol.[2] Its unique structure is definitively captured by standard chemical identifiers that are crucial for research and regulatory purposes. In its solid state, Baricitinib appears as a white to off-white powder with a melting point between 212°C and 215°C.[15] Its solubility characteristics are important for formulation and laboratory use; it is soluble in organic solvents such as dimethyl sulfoxide (DMSO) and dimethylformamide (DMF).[15]
Table 1: Drug Identification and Physicochemical Properties
Property | Value | Source(s) |
---|---|---|
Drug Name | Baricitinib | 1 |
Brand Name | Olumiant® | 2 |
DrugBank ID | DB11817 | 2 |
CAS Number | 1187594-09-7 | 2 |
Molecular Formula | C16H17N7O2S | 2 |
Molecular Weight | 371.42 g/mol | 2 |
IUPAC Name | 2-[1-ethylsulfonyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]azetidin-3-yl]acetonitrile | 1 |
SMILES String | CCS(=O)(=O)N1CC(C1)(CC#N)N2C=C(C=N2)C3=C4C=CNC4=NC=N3 | 1 |
InChIKey | XUZMWHLSFXCVMG-UHFFFAOYSA-N | 1 |
Appearance | White to off-white solid | 15 |
Melting Point | 212–215 °C | 16 |
Solubility | Soluble in DMSO (30–37.14 mg/mL), DMF (50 mg/mL) | 15 |
Baricitinib exerts its therapeutic effects by targeting the Janus kinase (JAK) family, a group of four intracellular tyrosine kinases (JAK1, JAK2, JAK3, and TYK2) that are pivotal for signal transduction from a wide array of cytokine and growth factor receptors.[1] Baricitinib is a potent, selective, and reversible inhibitor of JAK1 and JAK2. In cell-free assays, it demonstrates a half-maximal inhibitory concentration (
IC50) of 5.9 nM for JAK1 and 5.7 nM for JAK2, indicating high-affinity binding.[2]
A key pharmacological feature of Baricitinib is its selectivity profile. It has a lower affinity for TYK2 (IC50 = 53 nM) and is significantly less potent against JAK3 (IC50 > 400 nM), exhibiting over 100-fold selectivity for JAK1/2 compared to JAK3.[2] This selectivity is a foundational element of its design. JAK3 signaling is critically linked to the common gamma chain receptor subunit, which is essential for the function of cytokines vital for lymphocyte development and function (e.g., IL-2, IL-4, IL-7). By largely sparing JAK3, Baricitinib was designed to theoretically achieve its anti-inflammatory effects while potentially mitigating some of the broader immunosuppressive consequences associated with non-selective, pan-JAK inhibition. Although the clinical relevance of this selectivity is still under investigation, particularly in light of the class-wide boxed warnings, it remains a key differentiator from other JAK inhibitors.[5]
The therapeutic efficacy of Baricitinib stems from its ability to interrupt the JAK-STAT signaling pathway. This pathway is initiated when a cytokine or growth factor binds to its corresponding cell surface receptor, causing the receptors to dimerize. This brings the associated JAKs into close proximity, allowing them to auto-phosphorylate and become activated. These activated JAKs then phosphorylate specific tyrosine residues on the receptor, creating docking sites for Signal Transducers and Activators of Transcription (STATs). Once docked, STATs are themselves phosphorylated by the JAKs, leading to their dimerization and subsequent translocation into the cell nucleus. Inside the nucleus, STAT dimers act as transcription factors, modulating the expression of genes that regulate inflammation, hematopoiesis, and immune function.[5]
By inhibiting JAK1 and JAK2, Baricitinib effectively blocks this cascade. This blockade prevents the phosphorylation and activation of STATs, thereby downregulating the gene expression of numerous pro-inflammatory mediators. The cytokines whose signaling is attenuated by Baricitinib include interleukins IL-2, IL-6, IL-12, and IL-23, as well as interferons like IFN-γ, all of which are deeply implicated in the pathophysiology of autoimmune diseases.[5]
The primary pharmacodynamic effect of Baricitinib is potent immunosuppression and anti-inflammation, which underpins its use in autoimmune conditions like RA and AA.[1] In the context of RA, it is classified as a disease-modifying antirheumatic drug (DMARD) because it not only ameliorates symptoms like pain and swelling but has also been shown in animal models to slow disease progression by preserving cartilage and bone structure.[1]
The utility of Baricitinib in COVID-19 reveals a more complex mechanism. Its efficacy is not solely attributable to its established anti-inflammatory properties. While dampening the "cytokine storm"—the systemic hyperinflammatory response driven by cytokines like IL-6—is a primary benefit of JAK1/2 inhibition, a "dual-action" mechanism has been proposed. Research suggests that Baricitinib also inhibits numb-associated kinase (NAK) AAK1, an enzyme that regulates clathrin-mediated endocytosis.[2] This process is exploited by many viruses, including SARS-CoV-2, to gain entry into host cells. Therefore, Baricitinib may simultaneously mitigate the life-threatening hyperinflammation characteristic of severe COVID-19 while also directly interfering with the virus's ability to infect cells. This dual mechanism helps to explain its particular effectiveness and distinguishes its therapeutic profile from that of pure anti-inflammatory agents like corticosteroids.[2]
The pharmacokinetic profile of Baricitinib is characterized by rapid oral absorption, tissue distribution, minimal metabolism, and predominantly renal excretion, which collectively support a convenient once-daily dosing regimen.[2]
Following oral administration, Baricitinib is rapidly absorbed from the gastrointestinal tract, reaching peak plasma concentrations (Tmax) in approximately one hour (range 0.5–3 hours).[2] It exhibits good oral bioavailability, with an absolute bioavailability of approximately 79–80%.[5] The presence of food has a minimal effect on its overall exposure; a high-fat meal was found to decrease the area under the curve (AUC) by about 11% and the maximum concentration (
Cmax) by 18%, while delaying Tmax by 0.5 hours. These changes are not considered clinically significant, allowing Baricitinib to be administered with or without food.[2]
Baricitinib distributes into tissues beyond the plasma volume, as indicated by a volume of distribution (Vd) of 76 L following intravenous administration.[5] It is approximately 50% bound to plasma proteins, suggesting a moderate degree of binding that leaves a substantial fraction of the drug free to exert its pharmacological effects.[3]
Metabolism is a minor pathway for the elimination of Baricitinib. Less than 10% of an administered dose undergoes metabolism, which is primarily mediated by the cytochrome P450 enzyme CYP3A4.[2] Importantly, the resulting metabolites are not quantifiable in plasma, indicating that the parent compound, Baricitinib, is the sole contributor to its therapeutic activity.[5] This low level of metabolism suggests a lower potential for metabolic drug-drug interactions compared to extensively metabolized drugs.
The primary route of elimination for Baricitinib is renal excretion. Approximately 75% of an administered dose is eliminated in the urine, with another 20% eliminated in the feces.[2] The elimination half-life (
t1/2) in healthy subjects and patients with rheumatoid arthritis is approximately 12.5 hours, which is consistent with its once-daily dosing schedule.[2] A slightly shorter half-life of 10.8 hours has been observed in intubated COVID-19 patients receiving the drug via nasogastric tube.[5]
Table 2: Summary of Pharmacokinetic Parameters
Parameter | Value | Source(s) |
---|---|---|
Absolute Bioavailability | ~79–80% | 2 |
Time to Peak (Tmax) | ~1 hour (range: 0.5–3 hours) | 2 |
Effect of Food | Minimal; ~11% decrease in AUC, ~18% decrease in Cmax | 5 |
Plasma Protein Binding | ~50% | 3 |
Volume of Distribution (Vd) | 76 L | 5 |
Primary Metabolizing Enzyme | CYP3A4 (minor pathway) | 2 |
% Metabolized | <10% | 2 |
Primary Route of Elimination | Renal (~75% in urine) | 2 |
Elimination Half-Life | ~12.5 hours (RA); ~10.8 hours (intubated COVID-19) | 2 |
The clinical development program for Baricitinib has demonstrated its efficacy across a range of inflammatory and autoimmune conditions, leading to approvals for rheumatoid arthritis, alopecia areata, and COVID-19.
The efficacy of Baricitinib in RA was established through a comprehensive Phase 3 program, including the RA-BEAM, RA-BUILD, and RA-BEACON trials, which studied patients with inadequate responses to methotrexate (MTX), conventional synthetic DMARDs (csDMARDs), and biologic DMARDs (bDMARDs), respectively.[23]
The RA-BEAM trial (NCT01710358) was a pivotal 52-week, randomized, double-blind study involving over 1,300 patients with an inadequate response to MTX. It compared Baricitinib 4 mg once daily against both placebo and an active comparator, adalimumab 40 mg biweekly, with all patients continuing on background MTX.[9] At the primary endpoint of week 12, Baricitinib demonstrated superiority over placebo in achieving an American College of Rheumatology 20% response (ACR20), with 70% of Baricitinib-treated patients responding compared to 40% in the placebo group.[9]
A particularly compelling finding from RA-BEAM was Baricitinib's performance against the active comparator, adalimumab. While both drugs were effective, Baricitinib showed statistically significant superiority in key patient-reported outcomes (PROs). A physician may focus on objective measures like swollen joint counts, but a patient experiences the disease through symptoms like pain, fatigue, and functional limitations. In RA-BEAM, patients treated with Baricitinib reported significantly greater improvements in pain, physical function (as measured by the Health Assessment Questionnaire-Disability Index, HAQ-DI), and the severity and duration of morning joint stiffness compared to patients treated with adalimumab at week 12, with these benefits sustained through week 52.[24] Post-hoc analyses further suggested that this pain relief with Baricitinib occurred more rapidly than with adalimumab and may have been partially independent of the degree of inflammation control, hinting at a potential direct effect on pain perception pathways that warrants further investigation.[26]
Baricitinib became the first-ever FDA-approved systemic treatment for severe AA based on the results of two Phase 3 trials, BRAVE-AA1 (NCT03570749) and BRAVE-AA2 (NCT03899259).[11] These randomized, placebo-controlled trials enrolled over 1,200 adults with severe AA, defined as having a Severity of Alopecia Tool (SALT) score of ≥50 (indicating ≥50% scalp hair loss).[12]
The primary endpoint was the proportion of patients achieving a SALT score of ≤20 (representing 80% or more scalp hair coverage) at week 36. In BRAVE-AA1, this endpoint was achieved by 35% of patients on Baricitinib 4 mg and 22% on Baricitinib 2 mg, compared to just 5% of patients on placebo. The results were similar in BRAVE-AA2, with response rates of 33% (4 mg) and 17% (2 mg) versus 3% for placebo. All comparisons against placebo were highly statistically significant (p≤0.001).[11]
Long-term data from these trials indicate that hair regrowth is a gradual process and that continued therapy is necessary to achieve maximum benefit. Response rates continued to increase through week 52, at which point 40.9% of patients on the 4 mg dose in BRAVE-AA1 achieved a SALT score of ≤20.[28] Efficacy was maintained through 104 weeks of continuous treatment, with no new safety signals emerging, underscoring the importance of long-term treatment for this chronic condition.[27]
The role of Baricitinib in treating hospitalized patients with COVID-19 was evaluated in two major trials: ACTT-2 and COV-BARRIER.
The ACTT-2 trial (NCT04401579) randomized over 1,000 patients to receive either Baricitinib plus remdesivir or placebo plus remdesivir.[30] The study met its primary endpoint, showing a statistically significant one-day reduction in the median time to recovery for the combination group compared to the remdesivir-alone group (7 days vs. 8 days).[30] The benefit was most pronounced in patients requiring high-flow oxygen or non-invasive ventilation at baseline, for whom the median time to recovery was reduced from 18 days to 10 days.[31]
The COV-BARRIER trial (NCT04421027) enrolled over 1,500 hospitalized patients with elevated inflammatory markers and compared Baricitinib to placebo, both added to standard of care (which included corticosteroids for approximately 90% of patients).[10] This trial presented a critical dichotomy in its results. It failed to meet its primary endpoint, which was a composite of preventing progression to high-flow oxygen, non-invasive ventilation, invasive mechanical ventilation, or death by day 28 (27.8% in the Baricitinib group vs. 30.5% in the placebo group;
p=0.18).[10] However, Baricitinib demonstrated a significant benefit in a key secondary endpoint: all-cause mortality. Treatment with Baricitinib led to a 39% relative reduction in 28-day mortality (8.1% vs. 13.1%; Hazard Ratio 0.57).[2] The number needed to treat (NNT) to prevent one death was 20. This suggests that Baricitinib's primary value in COVID-19 is not in preventing disease progression in moderately ill patients, but rather in mitigating the lethal hyperinflammatory response in those who are already severely ill, thereby functioning as a targeted, life-saving intervention.
Table 3: Efficacy Endpoints from Pivotal RA, AA, and COVID-19 Trials
Indication | Trial Name | Endpoint | Baricitinib Result | Comparator Result | p-value / HR (95% CI) |
---|---|---|---|---|---|
RA | RA-BEAM | ACR20 Response (Week 12) | 70% | Placebo: 40% | p≤0.001 |
RA | RA-BEAM | Pain (VAS) Change (Week 12) | -34.8 | Adalimumab: -29.6 | p≤0.01 |
AA | BRAVE-AA1 | SALT Score ≤20 (Week 36) | 4 mg: 35% | Placebo: 5% | p≤0.001 |
AA | BRAVE-AA2 | SALT Score ≤20 (Week 36) | 4 mg: 33% | Placebo: 3% | p≤0.001 |
COVID-19 | ACTT-2 | Median Time to Recovery | 7 days | Placebo + RDV: 8 days | HR 1.16 (1.01-1.33) |
COVID-19 | COV-BARRIER | Progression or Death (Day 28) | 27.8% | Placebo + SOC: 30.5% | p=0.18 |
COVID-19 | COV-BARRIER | All-Cause Mortality (Day 28) | 8.1% | Placebo + SOC: 13.1% | HR 0.57 (0.41-0.78) |
The use of Baricitinib is associated with significant safety considerations, headlined by a class-wide FDA-mandated boxed warning. A thorough understanding of this risk profile is essential for appropriate patient selection and monitoring.
In 2021, the FDA required revisions to the boxed warning for Baricitinib and other JAK inhibitors to reflect risks identified primarily in a large, post-marketing safety study of another JAK inhibitor, tofacitinib.[33] The FDA applied these warnings to Baricitinib based on the shared mechanism of action, a decision made with prudence from a public health standpoint but which means the precise magnitude of these risks for Baricitinib specifically has not been defined by a similar dedicated trial. This "extrapolated risk" creates a degree of uncertainty for clinicians. The warning includes [7]:
This safety profile creates a profound clinical challenge, as the patient populations most in need of potent immunomodulation (e.g., older adults with severe RA and multiple comorbidities) are often the same ones at the highest baseline risk for MACE, malignancy, and thrombosis.[7] The decision to use Baricitinib thus becomes a complex negotiation of competing risks: the risk of progressive, uncontrolled autoimmune disease versus the iatrogenic risk from the treatment, necessitating in-depth shared decision-making with the patient.
Beyond the boxed warning, common treatment-emergent adverse events (reported in ≥1% of patients) include upper respiratory tract infections, headache, nausea, and acne (particularly in the AA population).[11] In COVID-19 trials, elevated liver enzymes (ALT/AST), thrombocytosis, and urinary tract infections were also common.[7]
Other clinically significant adverse events include gastrointestinal perforations, which have been reported in clinical studies. The risk may be higher in patients with a history of diverticulitis or those taking concomitant NSAIDs or corticosteroids.[35] Laboratory abnormalities are also a key monitoring parameter and include dose-dependent decreases in neutrophil and lymphocyte counts (neutropenia, lymphopenia), decreases in hemoglobin (anemia), and elevations in liver transaminases, lipids (LDL-C, HDL-C), and creatine phosphokinase.[7]
Baricitinib is contraindicated or not recommended in several patient populations due to its risk profile. This includes patients with active serious infections (including active TB), severe renal impairment (estimated glomerular filtration rate <30 mL/min/1.73 m2), or severe hepatic impairment.[22] Its use during pregnancy is not recommended due to evidence of teratogenicity in animal studies, and effective contraception is advised.[36] Caution is warranted in patients with a history of thromboembolism.[36]
Table 4: Summary of FDA Boxed Warning and Key Risk Factors
Warning Category | Description of Risk | Key Risk Factors / Considerations |
---|---|---|
Serious Infections | Increased risk of bacterial, fungal, viral, and opportunistic infections, including TB. | Chronic or recurrent infections, residence in endemic areas, concomitant immunosuppressants. Pre-screening for TB is essential. |
Mortality | Higher rate of all-cause mortality observed with another JAK inhibitor vs. TNF blockers. | Risk extrapolated to Baricitinib; consider individual patient benefits vs. risks. |
Malignancy | Increased risk of lymphoma and other cancers, including NMSC. | Current or past smokers, known malignancy (other than treated NMSC). |
MACE | Increased risk of heart attack, stroke, and cardiovascular death. | Age ≥50 years, at least one cardiovascular risk factor (e.g., hypertension, hyperlipidemia, diabetes), current or past smoker. |
Thrombosis | Increased incidence of DVT, PE, and arterial thrombosis. | History of thrombosis, known risk factors for thrombosis (e.g., obesity, prolonged immobilization). |
Correct dosing and awareness of potential interactions are critical for the safe and effective use of Baricitinib. Dosing varies by indication and is subject to adjustment based on patient characteristics and concomitant medications.
Baricitinib was discovered by Incyte Corporation and subsequently licensed to Eli Lilly and Company for further development and commercialization.[4] Its journey to market has been marked by distinct regulatory pathways across different jurisdictions and indications.
The European Medicines Agency (EMA) was the first major regulatory body to approve Baricitinib, granting marketing authorization for the treatment of RA in February 2017.[2] The path in the United States was more complex. In April 2017, the FDA issued a Complete Response Letter, initially declining approval due to concerns about dosing and safety, particularly regarding thromboembolic events.[2] Following the submission of additional data, the FDA granted approval for RA in May 2018, but only for the 2 mg dose as the primary recommendation, with the 4 mg dose reserved for specific circumstances.[2]
The COVID-19 pandemic prompted a rapid expansion of Baricitinib's use. In November 2020, the FDA granted an Emergency Use Authorization (EUA) for its use in combination with remdesivir.[43] This was followed by full FDA approval in May 2022 for the treatment of hospitalized adults with COVID-19, making Baricitinib the first immunomodulatory drug to receive such approval.[2]
A landmark achievement occurred in June 2022, when the FDA approved Baricitinib for adults with severe alopecia areata, establishing it as the first-ever systemic treatment for this autoimmune disease.[12] Baricitinib is also approved for atopic dermatitis and JIA in the European Union.[6] The drug is marketed globally under the brand name Olumiant®, and the manufacturer offers patient support services, such as the Olumiant Together program, to assist with access and affordability.[13]
Baricitinib has established itself as a versatile and potent immunomodulatory agent with a complex benefit-risk profile that varies significantly by indication. Its therapeutic value is undeniable in specific patient populations, but its use demands a nuanced and individualized approach.
For rheumatoid arthritis, Baricitinib offers a valuable oral, second-line option for patients who have failed TNF blockers. Its strong efficacy, particularly the superior improvement in patient-reported outcomes like pain and function compared to a leading biologic, is a significant advantage. This must be weighed against the long-term, class-wide risks of MACE, malignancy, and thrombosis, which are especially relevant in the often older, comorbid RA population.
For severe alopecia areata, Baricitinib represents a breakthrough as the first approved systemic therapy. For many patients, the prospect of significant hair regrowth is a profound, life-altering benefit. This psychological and social benefit must be carefully balanced against the risks inherent to long-term systemic immunosuppression for a condition that is not life-threatening.
In the context of COVID-19, Baricitinib has a clearly defined niche. It is not a broad antiviral but a targeted immunomodulator that significantly reduces mortality in hospitalized patients with severe disease and hyperinflammation. Here, the clear mortality benefit in an acute, life-threatening setting generally outweighs the risks of short-term use.
Looking forward, several key questions remain. The most pressing is the need for large-scale, long-term safety studies specific to Baricitinib to precisely quantify its risk of MACE and malignancy, which would help to either confirm or differentiate its profile from other JAK inhibitors. The post-marketing study NCT04086745 is designed to compare its VTE risk against TNF inhibitors but may not address all safety concerns.[47] Further research into its potential neuromodulatory effects on pain perception could unlock new therapeutic avenues. Finally, as its use expands into pediatric populations for JIA and potentially AA, long-term efficacy and safety data in these groups will be crucial. The continued exploration of Baricitinib in other cytokine-mediated diseases, such as the ongoing trial in depression associated with HIV, demonstrates the enduring interest in harnessing the potential of targeted JAK inhibition.[48]
Published at: July 17, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.