MedPath

Fostamatinib Advanced Drug Monograph

Published:Aug 6, 2025

Generic Name

Fostamatinib

Brand Names

Tavalisse, Tavlesse

Drug Type

Small Molecule

Chemical Formula

C23H26FN6O9P

CAS Number

901119-35-5

Associated Conditions

Chronic immune thrombocytopenia

Comprehensive Monograph on Fostamatinib (DB12010)

Executive Summary and Key Insights

Fostamatinib is a first-in-class, orally administered small molecule that functions as a prodrug for its active metabolite, R406.[1] R406 is a potent and selective inhibitor of Spleen Tyrosine Kinase (Syk), a critical enzyme in the intracellular signaling pathways of various immune cells.[3] Marketed under the brand names Tavalisse in the United States and Tavlesse in Europe, fostamatinib is approved for the treatment of adult patients with chronic immune thrombocytopenia (ITP) who have had an insufficient response to previous therapies.[3] Its primary mechanism of action involves the inhibition of Fc receptor-mediated phagocytosis of antibody-coated platelets by macrophages, a novel therapeutic approach that directly targets a key pathological process in ITP.[3]

The clinical efficacy of fostamatinib in refractory ITP has been established through a pivotal Phase 3 program. While the overall rate of stable platelet response is modest, the data from two placebo-controlled trials and a long-term extension study demonstrate a statistically significant and clinically meaningful benefit for a difficult-to-treat patient population.[10] For patients who do respond, the increase in platelet count is often rapid, occurring within weeks of initiation, and has been shown to be durable with continued treatment.[11]

The safety and tolerability profile of fostamatinib is well-characterized and considered manageable. The most frequently reported adverse events include diarrhea, hypertension, nausea, and elevations in liver transaminases.[5] These toxicities are generally mild to moderate and can be managed effectively through supportive care and a structured dose-modification strategy, including dose reduction, interruption, or discontinuation, as outlined in the prescribing information.[14] Rigorous monitoring of blood pressure, liver function, and blood counts is essential for the safe use of the drug.

The therapeutic potential of fostamatinib extends beyond ITP, driven by its fundamental immunomodulatory mechanism. The drug is under active investigation for other Syk-mediated autoimmune conditions, including warm autoimmune hemolytic anemia (wAIHA) and IgA nephropathy (IgAN). Early-phase clinical data in these indications have shown promising signals, suggesting a significant opportunity for label expansion.[6] Fostamatinib holds regulatory approvals from the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) and is being commercialized globally through a strategic partnership model led by Rigel Pharmaceuticals in collaboration with Grifols and Kissei Pharmaceuticals.[3]

Molecular Profile and Chemical Characteristics

Nomenclature and Identifiers

Fostamatinib is identified by a comprehensive set of chemical and regulatory names, ensuring precise tracking across scientific literature, clinical trials, and commercial markets.

  • Generic Name: Fostamatinib.[3]
  • Brand Names: The drug is marketed as Tavalisse in the United States and as Tavlesse in the European Union.[3]
  • DrugBank ID: DB12010.[3]
  • CAS Number: The primary CAS Registry Number for the fostamatinib free acid is 901119-35-5.[4] The commercially available salt forms also have distinct identifiers, including 1025687-58-4 for the sodium salt and 945745-48-2 for a deprecated CAS number.[4]
  • Synonyms and Internal Codes: During its development, fostamatinib was known by several internal codes, most notably R788 and R935788. The active metabolite is designated R406.[1]
  • IUPAC Name: The systematic chemical name for the molecule is [6-({5-Fluoro-2-[(3,4,5-trimethoxyphenyl)amino]-4-pyrimidinyl}amino)-2,2-dimethyl-3-oxo-2,3-dihydro-4H-pyrido[3,2-b]oxazin-4-yl]methyl dihydrogen phosphate.[4] The sodium salt is chemically named sodium (6-((5-fluoro-2-((3,4,5-trimethoxyphenyl)amino)pyrimidin-4-yl)amino)-2,2-dimethyl-3-oxo-2H-pyrido[3,2-b]oxazin-4(3H)-yl)methyl phosphate.[20]

Chemical Structure and Properties

Fostamatinib is classified as a small molecule drug, characterized by its specific chemical structure and physicochemical properties that dictate its formulation, stability, and biological activity.[3]

  • Type and Modality: Small Molecule.[3]
  • Chemical Formula: The molecular formula for the free acid form of fostamatinib is C23​H26​FN6​O9​P.[4] The commercially available formulation, Tavalisse, is the disodium hexahydrate salt, with a molecular formula of C23​H24​FN6​Na2​O9​P⋅6H2​O.[14]
  • Molar Mass: The molar mass of the free acid is 580.466 g·mol⁻¹.[6] The molecular weight of the fostamatinib disodium hexahydrate salt is 732.52 g/mol.[14]
  • Physical Properties: Fostamatinib disodium presents as a white to off-white solid powder.[14] It exhibits solubility in dimethyl sulfoxide (DMSO) but is not soluble in water, a key property influencing its pharmaceutical formulation.[20]
  • Storage and Stability: For research purposes, the compound is stored in a dry, dark environment, typically at 2-8°C for short-term use and at -20°C for long-term stability.[20] The final commercial product (Tavalisse tablets) is stored at controlled room temperature (20°C to 25°C) in its original container, which includes desiccant canisters to protect against moisture.[14]

The pharmaceutical design of fostamatinib as a phosphate prodrug is a critical element of its clinical utility. The active molecule, R406, likely possesses suboptimal physicochemical properties for oral administration, such as poor aqueous solubility or limited membrane permeability, which are common challenges for kinase inhibitors. To overcome this, developers employed a prodrug strategy by attaching a phosphate ester group to the molecule. This modification increases the aqueous solubility of the compound, making it suitable for formulation into an oral tablet. Upon ingestion, this phosphate group is rapidly and efficiently cleaved in the gastrointestinal tract by ubiquitous enzymes, specifically intestinal alkaline phosphatases, releasing the active drug R406 for absorption into the portal circulation.[3] This elegant design bypasses the inherent biopharmaceutical limitations of the active compound itself.

This two-stage process has profound implications for the drug's overall pharmacokinetic profile. The initial activation step is dependent on gut enzymes, a process that is generally robust and less prone to the drug-drug interactions that affect hepatic enzyme systems. However, the subsequent metabolism and clearance of the now-active R406 molecule are indeed dependent on hepatic enzymes, primarily CYP3A4.[3] This creates a distinct metabolic pathway that must be fully understood to predict and manage potential drug-drug interactions safely. The further choice to formulate the drug as a disodium hexahydrate salt is a standard pharmaceutical practice aimed at optimizing the solid-state properties of the drug substance, enhancing its stability, handling, and dissolution characteristics to ensure consistent and reliable absorption after oral administration.[14]

Pharmacological Profile: Mechanism of Action and Pharmacodynamics

Primary Mechanism: Spleen Tyrosine Kinase (Syk) Inhibition

Fostamatinib functions as an orally administered prodrug that is rapidly converted in vivo to its active metabolite, R406.[1] The therapeutic effects of the drug are entirely attributable to R406, which is a potent, competitive, and reversible inhibitor of Spleen Tyrosine Kinase (Syk).[1]

R406 exhibits high-affinity binding to the adenosine triphosphate (ATP) binding pocket within the catalytic domain of the Syk enzyme, with a reported binding affinity (dissociation constant, Ki​) of 30 nM.[3] This competitive binding prevents ATP from accessing the kinase, thereby inhibiting its enzymatic activity with a half-maximal inhibitory concentration (

IC50​) of 41 nM.[1]

Syk is a non-receptor, cytosolic protein tyrosine kinase that serves as a central signaling molecule in hematopoietic cells.[4] It plays a pivotal role in coupling activated immunoreceptors to downstream intracellular events that mediate diverse cellular responses, including proliferation, differentiation, phagocytosis, and the production of inflammatory mediators.[3] Syk is essential for signal transduction downstream of a variety of critical immune receptors, most notably Fc receptors (FcR), B-cell receptors (BCR), and T-cell receptors (TCR).[2]

Pathophysiological Relevance in ITP

The mechanism of fostamatinib is uniquely suited to the underlying pathophysiology of immune thrombocytopenia. ITP is an autoimmune disorder characterized by the production of autoantibodies, primarily IgG, that bind to glycoproteins on the surface of platelets.[6] This opsonization marks the platelets for premature destruction.

The antibody-coated platelets are recognized and bound by Fc-gamma receptors (FcγR) expressed on the surface of phagocytic cells, particularly macrophages located in the spleen and liver.[6] The engagement of these FcγR initiates an intracellular signaling cascade that is critically dependent on Syk. Upon receptor clustering, Syk is recruited and activated, leading to the phosphorylation of downstream substrates. This cascade culminates in profound cytoskeletal rearrangement within the macrophage, a process essential for engulfment and phagocytosis of the platelet.[2]

By potently inhibiting Syk, fostamatinib's active metabolite R406 directly interrupts this pathological pathway. It blocks the signal transduction originating from the FcγR, thereby preventing the activation of macrophages and inhibiting the phagocytic destruction of platelets.[2] This reduction in platelet clearance allows the circulating platelet count to rise toward homeostatic levels, mitigating the bleeding risk associated with ITP.[3]

Broader Pharmacodynamic Effects and Off-Target Activity

The role of Syk extends beyond FcγR signaling in macrophages, giving fostamatinib a broad spectrum of immunomodulatory activity and also accounting for some of its off-target effects.

  • General Immunomodulation: By inhibiting Syk, R406 also effectively blocks signaling from B-cell receptors (BCRs) and T-cell receptors (TCRs). This action can suppress the activation and proliferation of B and T lymphocytes, as well as interfere with dendritic cell maturation and antigen presentation.[3] This broader immunosuppressive effect contributes to a reduction in the production of pro-inflammatory cytokines and mediators, such as tumor necrosis factor-alpha (TNF-α), interleukin-8 (IL-8), leukotriene C4, and granulocyte-macrophage colony-stimulating factor.[3]
  • Kinase Selectivity: While fostamatinib is considered a selective Syk inhibitor, in vitro kinase assays have shown that R406 can inhibit other kinases at concentrations comparable to its Syk IC50​. These include Flt3, Lyn (IC50​ = 63 nM), and Lck (IC50​ = 37 nM).[1] However, in more physiologically relevant cell-based assays, R406 demonstrates significantly greater potency against Syk-mediated pathways compared to others, suggesting a functional selectivity in a cellular context.[20]
  • Off-Target Effects: Fostamatinib treatment is associated with a notable incidence of hypertension, which is believed to be an off-target effect unrelated to Syk inhibition.[3] The precise mechanism is not fully elucidated but may relate to the inhibition of other kinases or transporters involved in the regulation of vascular tone. Furthermore, R406 has been found to interact with several other biological targets, acting as an antagonist at the adenosine A3 receptor and as an inhibitor of various enzymes and transporters, including UDP-glucuronosyltransferase 1A1 (UGT1A1), phosphodiesterase 5 (PDE5), fatty acid amide hydrolase, and cathepsins L and S.[3] These interactions may contribute to the overall pharmacodynamic and safety profile of the drug.

The therapeutic rationale for fostamatinib in ITP is a direct and logical consequence of its ability to block FcγR signaling in macrophages. This same fundamental mechanism, however, positions the drug as a potential platform molecule for a range of other autoimmune diseases where Syk plays a central pathological role. The clinical investigations into its use for warm autoimmune hemolytic anemia (wAIHA), IgA nephropathy (IgAN), and rheumatoid arthritis are not speculative endeavors but are mechanistically driven extensions of its core pharmacology. In wAIHA, FcγR-mediated destruction of red blood cells is the primary driver of disease; in IgAN, Syk is implicated in renal inflammation; and in rheumatoid arthritis, Syk is involved in the inflammatory cascades within the synovium.[3]

This broad biological activity is a double-edged sword. While it provides a strong basis for pipeline expansion, it is also the likely source of the drug's characteristic adverse effect profile. The development of hypertension is explicitly noted as an off-target effect, likely resulting from the inhibition of other kinases or transporters that regulate blood pressure.[3] The extensive list of secondary targets identified in preclinical screening, such as adenosine receptors and PDE5, paints a picture of a molecule that, while primarily aimed at Syk, interacts with numerous other biological systems.[3] This explains why its clinical use necessitates more intensive monitoring for side effects like hypertension and liver enzyme elevations compared to a more highly specific therapeutic like a monoclonal antibody. The clinical benefit in ITP is thus intrinsically linked to a predictable set of risks derived from its systemic, multi-faceted mechanism of action.

Clinical Pharmacokinetics (ADME)

The clinical behavior of fostamatinib is governed by the absorption, distribution, metabolism, and excretion (ADME) properties of its active metabolite, R406. Fostamatinib itself is a prodrug with negligible systemic exposure.

Absorption

Fostamatinib is designed for oral administration and undergoes rapid and extensive conversion to R406 in the gastrointestinal tract. This hydrolysis is mediated by intestinal alkaline phosphatase.[3]

  • Bioavailability: The active metabolite R406 has an absolute oral bioavailability of 55%, indicating efficient absorption following its formation in the gut.[3]
  • Time to Peak Concentration (Tmax​): Peak plasma concentrations of R406 are typically reached at a median of 1.5 hours (range: 1 to 4 hours) after administration under fasting conditions.[3]
  • Effect of Food: Administration of fostamatinib with a high-calorie, high-fat meal results in a modest increase in R406 exposure, with the area under the curve (AUC) increasing by 23% and the maximum concentration (Cmax​) increasing by 15%.[3] Due to this limited effect, the drug can be taken with or without food, providing flexibility for patients.[14]

Distribution

Once absorbed, R406 distributes extensively throughout the body.

  • Protein Binding: R406 is highly bound to human plasma proteins, with a binding fraction of 98.3%.[3] This high degree of binding can influence its distribution and potential for drug interactions.
  • Volume of Distribution (Vd​): The mean apparent volume of distribution of R406 at steady state is approximately 256 L, indicating significant distribution into tissues beyond the plasma compartment.[5]

Metabolism

R406 is the major pharmacologically active component and undergoes extensive metabolism before excretion.

  • Metabolic Pathways: The primary routes of metabolism for R406 are oxidation, mediated by the cytochrome P450 3A4 (CYP3A4) enzyme system, and glucuronidation, mediated by UDP glucuronosyltransferase 1A9 (UGT1A9).[3]
  • Metabolites: Major metabolites identified in plasma include an O-glucuronide conjugate, an N-glucuronide conjugate, and an O-desmethyl metabolite.[3]

Excretion

The metabolites of R406 are eliminated from the body through both fecal and renal routes.

  • Route of Elimination: Approximately 80% of the administered dose is excreted in the feces, while the remaining 20% is excreted in the urine.[3] The primary forms found in feces are the O-glucuronide conjugate and an O-desmethyl metabolite further processed by gut bacteria, whereas the N-glucuronide conjugate is the main form in urine.[3]
  • Half-Life (t1/2​): The mean terminal elimination half-life of R406 is approximately 15 hours.[3] This pharmacokinetic property supports a twice-daily dosing regimen to maintain therapeutic concentrations.

Drug-Drug Interaction Profile

The metabolism of R406 via CYP3A4 makes it susceptible to interactions with potent inhibitors and inducers of this enzyme.

  • Strong CYP3A4 Inhibitors: Co-administration with strong CYP3A4 inhibitors, such as ketoconazole, significantly increases exposure to R406. Studies have shown a 102% increase in AUC and a 37% increase in Cmax​.[14] This may elevate the risk of fostamatinib-related toxicities. Therefore, careful monitoring and potential dose reduction of fostamatinib are required when used concomitantly with strong CYP3A4 inhibitors.[5]
  • Strong CYP3A4 Inducers: Co-administration with strong CYP3A4 inducers, such as rifampicin, markedly reduces exposure to R406, with a reported 75% decrease in AUC.[14] This can compromise the efficacy of fostamatinib, and therefore, concomitant use is not recommended.[5]
  • Effect on Other Drugs: Fostamatinib and its metabolite R406 can also act as inhibitors of other drug-metabolizing enzymes and transporters. Concomitant use may increase the concentrations of substrates for CYP3A4 (e.g., simvastatin), the Breast Cancer Resistance Protein (BCRP) transporter (e.g., rosuvastatin), and the P-glycoprotein (P-gp) transporter (e.g., digoxin). Dose reductions of these co-administered drugs may be necessary to avoid toxicity.[7]

The following table provides a consolidated summary of the key pharmacokinetic parameters for R406, the active metabolite of fostamatinib.

ParameterValueClinical ImplicationSource(s)
Absolute Bioavailability55%Indicates good oral absorption of the active metabolite after prodrug conversion.3
Time to Peak (Tmax​)~1.5 hours (fasting)Rapid onset of absorption.3
Effect of FoodAUC ↑ 23%, Cmax​ ↑ 15% (high-fat meal)Minor food effect allows for dosing with or without food, enhancing patient convenience.3
Plasma Protein Binding98.3%High binding may limit free drug concentration but also creates a reservoir; potential for displacement interactions.3
Volume of Distribution (Vd​)256 LExtensive tissue distribution, suggesting the drug acts systemically and not just within the plasma.5
Terminal Half-Life (t1/2​)~15 hoursSupports a twice-daily (BID) dosing schedule to maintain steady-state concentrations.5
Metabolism PathwaysCYP3A4 (oxidation), UGT1A9 (glucuronidation)Primary reliance on CYP3A4 makes it susceptible to significant drug-drug interactions with inhibitors/inducers.3
Route of Excretion~80% Feces, ~20% UrinePrimarily hepatic/biliary clearance, with a smaller contribution from renal clearance.3

Clinical Efficacy in Chronic Immune Thrombocytopenia (ITP)

The approval of fostamatinib for chronic ITP was based on a comprehensive clinical development program, highlighted by two pivotal Phase 3 trials and a long-term open-label extension study.

Pivotal Phase 3 Program (FIT-1 and FIT-2)

The core evidence for fostamatinib's efficacy comes from two identically designed, multicenter, randomized, double-blind, placebo-controlled trials: FIT-1 (NCT02076399) and FIT-2 (NCT02076412).[10]

  • Study Design: A total of 150 adult patients with persistent or chronic ITP were enrolled across the two studies. To be eligible, patients must have had an insufficient response to at least one prior ITP therapy. Patients were randomized in a 2:1 ratio to receive either fostamatinib or a matching placebo for 24 weeks.[11]
  • Patient Population: The trials enrolled a heavily pre-treated and refractory patient population. The median duration of ITP was 8.5 years, and patients had failed multiple prior lines of therapy, including corticosteroids, immunoglobulins, rituximab, thrombopoietin receptor agonists (TPO-RAs), and/or splenectomy.[11] The median baseline platelet count was severely low at 16,000/μL.[11]
  • Dosing Regimen: Patients initiated treatment with fostamatinib 100 mg twice daily (BID). For those who did not show an adequate platelet response after 4 weeks, the dose was escalated to 150 mg BID.[10]
  • Primary Efficacy Endpoint: The primary endpoint for both studies was the rate of stable platelet response. This was stringently defined as achieving a platelet count of at least 50,000/μL on at least four of the six scheduled clinic visits between weeks 14 and 24 of treatment, without the use of rescue medication.[6]

Efficacy Results

The results of the two trials showed a consistent effect for fostamatinib, although they differed in statistical significance for the primary endpoint.

  • FIT-1 Trial (NCT02076399): This trial successfully met its primary endpoint. The stable platelet response rate was 18% (9 out of 51 patients) in the fostamatinib arm, compared to 0% (0 out of 25 patients) in the placebo arm. This difference was statistically significant (p=0.03).[6]
  • FIT-2 Trial (NCT02076412): This trial did not formally meet its primary endpoint. The stable response rate was 16% (8 out of 50 patients) in the fostamatinib arm, compared to 4% (1 out of 25 patients) in the placebo arm. While the response rate in the fostamatinib group was consistent with FIT-1, the single responder in the placebo group rendered the difference not statistically significant (p=0.26).[10]
  • Combined Analysis: Recognizing the consistency of the treatment effect, a pre-planned pooled analysis of the data from both trials was conducted. This combined analysis demonstrated a highly statistically significant benefit for fostamatinib. The stable response rate in the pooled fostamatinib group (n=101) was 18%, compared to just 2% in the pooled placebo group (n=49), with a p-value of 0.0003.[11]

Key Secondary and Other Endpoints

  • Overall Response: A retrospective analysis defined an "overall response" as achieving at least one platelet count of ≥50,000/μL within the first 12 weeks of treatment. By this measure, 43% of patients treated with fostamatinib achieved a response, compared to only 14% of patients on placebo (p=0.0006).[11] This indicates that a broader group of patients experienced at least a transient benefit.
  • Time to Response: The response to fostamatinib was notably rapid. For those who responded, the median time to the first platelet count increase to ≥50,000/μL was only 15 days, with the majority of responders (83%) achieving this milestone within 8 weeks of initiating therapy.[11]

Long-Term Efficacy (FIT-3 Open-Label Extension)

The FIT-3 study (NCT02077192) was an open-label extension trial that allowed patients from FIT-1 and FIT-2 to receive long-term fostamatinib treatment, providing crucial data on the durability of response and the drug's effect in patients previously on placebo.[14]

  • Durability of Response: Patients who had responded to fostamatinib in the parent trials and continued into the extension study largely maintained their response. The median platelet count for these durable responders was 96,000/μL after a median exposure of 13 months, demonstrating the long-term sustainability of the treatment effect.[28] Across all three trials, 18 subjects were documented to have maintained a platelet count of at least 50,000/μL for 12 months or longer.[14]
  • Efficacy in Placebo Crossover Patients: A critical finding from FIT-3 was the response observed in patients who had been on placebo in the pivotal trials and subsequently initiated fostamatinib. In a prospectively defined analysis, 17% to 23% of these previously placebo-treated patients achieved a stable platelet response after starting active treatment, confirming the drug's therapeutic activity in a new cohort of patients.[10]

The divergent outcomes of the FIT-1 and FIT-2 trials, where one met its primary endpoint and the other did not, highlight the inherent statistical challenges of conducting clinical trials in rare and refractory diseases with small patient populations. The failure of FIT-2 to reach statistical significance was not due to a lack of efficacy in the fostamatinib arm—the response rate was nearly identical to that in FIT-1 (16-18%). Instead, the outcome was driven by the random chance of a single patient in the small placebo group (n=25) achieving a stable response, which was enough to push the p-value above the traditional 0.05 threshold.[10]

This scenario underscores why regulatory agencies like the FDA and EMA evaluate the "totality of the evidence" rather than relying on a single p-value from one trial.[28] In the case of fostamatinib, the evidence was compelling. The consistent response rate for the active drug across both studies, the highly significant result from the pooled analysis, the rapid and durable nature of the responses, and the confirmatory data from the FIT-3 extension study (where placebo patients responded to active drug) collectively built a strong case for approval.[10] This regulatory journey serves as a key example of how a drug with a modest but clinically meaningful benefit for a niche, high-need population can successfully navigate a complex development path. It positions fostamatinib not as a universal solution for ITP, but as an important and mechanistically novel option for patients who have exhausted other available therapies.

Investigational Applications and Future Directions

The mechanism of fostamatinib, centered on the inhibition of Syk-mediated immune processes, provides a strong rationale for its investigation in a variety of other autoimmune and inflammatory diseases beyond ITP.

IgA Nephropathy (IgAN)

  • Scientific Rationale: The investigation in IgAN is supported by preclinical evidence showing increased expression of Syk in the kidney biopsies of patients with the disease. Inhibition of Syk was shown to reduce the production of inflammatory cytokines from mesangial cells stimulated by IgA immune complexes, directly targeting a key pathological driver.[30]
  • Phase 2 Trial (SIGN - NCT02112838): A Phase 2, randomized, double-blind, placebo-controlled trial evaluated fostamatinib in 76 patients with IgAN who had persistent proteinuria despite being on standard-of-care renin-angiotensin system inhibitors.[30]
  • Clinical Findings: The study did not meet its primary endpoint of a statistically significant reduction in proteinuria across the entire study population. However, a pre-specified subgroup analysis of high-risk patients—those with a baseline urine protein to creatinine ratio (UPCR) greater than 1000 mg/g—revealed a promising and dose-dependent trend. In this high-risk group, the 150 mg BID dose of fostamatinib was associated with a 36% median reduction in proteinuria from baseline to 24 weeks, compared to a 14% reduction in the placebo group.[30] The drug was generally well-tolerated, and repeat kidney biopsies in a subset of patients showed a reduction in Syk staining, providing histological evidence of target engagement.[30]
  • Future Outlook: While the primary endpoint was not met, the positive signal in the high-risk subgroup is considered a valuable "proof of principle." These findings may support the design of a larger, longer-term Phase 3 trial specifically targeting this patient population, for whom there remains a high unmet medical need.[15]

Warm Autoimmune Hemolytic Anemia (wAIHA)

  • Scientific Rationale: wAIHA shares a common pathophysiology with ITP. It is driven by autoantibodies that coat red blood cells, leading to their destruction by macrophages via the same FcγR-Syk signaling pathway. Therefore, Syk inhibition is a highly rational therapeutic strategy.[6]
  • Clinical Development: Promising results from an initial Phase 2 open-label study prompted the advancement to a larger pivotal program.[6]
  • Phase 3 Program (NCI-2019-05092): A multi-center, randomized, double-blind, placebo-controlled Phase 3 study is currently underway to formally assess the efficacy of fostamatinib in patients with wAIHA. Key inclusion criteria include a hemoglobin level of ≤10 g/dL and having failed prior therapies. An associated open-label extension study is also active to gather long-term data.[16]

COVID-19

  • Scientific Rationale: Fostamatinib was investigated as a potential treatment for severe COVID-19 based on the hypothesis that Syk inhibition could modulate the hyperinflammatory response (cytokine storm) that leads to acute respiratory distress syndrome (ARDS).[3]
  • Phase 3 Trial (NCT04924660): A large, multicenter, randomized, placebo-controlled trial was conducted in 400 adults hospitalized with COVID-19 and hypoxemia during the Omicron variant era. The primary outcome was the number of oxygen-free days at day 28.[34]
  • Clinical Findings: The trial did not meet its primary endpoint. There was no significant difference in the mean number of oxygen-free days between the fostamatinib group and the placebo group. The results do not support the use of fostamatinib for treating hospitalized patients with COVID-19-related hypoxemia.[34]

Other Investigational Indications

  • Rheumatoid Arthritis (RA): An early Phase 2 study in RA patients who had failed biologic agents showed limited efficacy overall compared to placebo. However, a potential treatment signal was observed in a subgroup of patients with a high inflammatory burden, as measured by elevated C-reactive protein levels.[6]
  • Lymphoma: Fostamatinib has been evaluated in Phase 2 clinical trials for hematologic malignancies, including Diffuse Large B-Cell Lymphoma (DLBCL).[6]
  • Sickle Cell Disease (SCD): A Phase 1 study sponsored by the National Institutes of Health (NIH) is currently evaluating the safety and tolerability of fostamatinib in patients with SCD. The rationale is that Syk inhibition may help mitigate the chronic inflammation and downstream consequences, such as vaso-occlusive crises (VOCs), that characterize the disease.[36]

Safety, Tolerability, and Risk Management

The safety profile of fostamatinib has been extensively characterized through its clinical development program in ITP and other indications. The adverse reactions are generally predictable and can be managed with appropriate monitoring and intervention.

Comprehensive Adverse Reaction Profile

  • Most Common Adverse Reactions: The most frequently reported adverse reactions, occurring in ≥5% of patients and more commonly than placebo in the pivotal ITP trials, are:
  • Gastrointestinal: Diarrhea (31%), nausea (19%), abdominal pain (6%).[6]
  • Cardiovascular: Hypertension (28%), chest pain (6%).[6]
  • Hepatic: Increased Alanine Aminotransferase (ALT) and/or Aspartate Aminotransferase (AST) (9-11%).[6]
  • General/Neurological: Dizziness (11%), fatigue (6%).[6]
  • Infectious/Hematologic: Respiratory infection (11%), neutropenia (6%).[6]
  • Dermatologic: Rash (9%).[14]
  • Serious Adverse Reactions: In the ITP clinical trials, serious adverse drug reactions each occurred with an incidence of 1%. These included febrile neutropenia, severe diarrhea, pneumonia, and hypertensive crisis.[7]

Warnings, Precautions, and Clinical Monitoring

The prescribing information for fostamatinib includes several important warnings and precautions that necessitate a structured risk management plan, including regular clinical and laboratory monitoring.

  • Hypertension: New or worsening hypertension is a common and potentially severe side effect. Blood pressure must be monitored every two weeks after initiation until a stable dose is achieved, and monthly thereafter. Standard antihypertensive therapy should be initiated or adjusted as needed to maintain blood pressure control. Persistent or severe hypertension may require fostamatinib dose interruption, reduction, or discontinuation.[7]
  • Hepatotoxicity: Elevations in liver transaminases (ALT and AST) are common. Liver function tests (LFTs) must be monitored monthly throughout treatment. If ALT or AST levels rise to ≥3 times the upper limit of normal (ULN), management requires dose interruption, reduction, or discontinuation, depending on the severity and presence of symptoms or hyperbilirubinemia.[4]
  • Diarrhea: Diarrhea is the most common gastrointestinal side effect and can be severe in a small percentage of patients. Patients should be advised on supportive care measures, including hydration, dietary changes, and the use of anti-diarrheal medications. If diarrhea becomes severe (Grade 3 or higher), dose interruption or reduction is necessary.[7]
  • Neutropenia: Decreases in white blood cell counts, specifically the absolute neutrophil count (ANC), are common and can be severe, potentially increasing the risk of infection. The ANC must be monitored monthly. A significant decrease in neutrophils requires dose modification or discontinuation.[7]
  • Embryo-Fetal Toxicity: Based on its mechanism of action and findings in animal studies, fostamatinib can cause fetal harm. It is contraindicated for use during pregnancy. Females of reproductive potential must have a negative pregnancy test before starting treatment and must use effective contraception during therapy and for at least one month after the final dose.[7]
  • Lactation: It is not known if fostamatinib or its metabolites are present in human milk. Due to the potential for serious adverse reactions in a breastfed child, lactating women are advised not to breastfeed during treatment and for at least one month after the last dose.[7]

The following table provides a summary of the recommended dose modifications for managing the key adverse reactions associated with fostamatinib, as outlined in the prescribing information.

Adverse ReactionSeverity / ThresholdRecommended ActionSource(s)
HypertensionStage 1 (Systolic 130-139 or Diastolic 80-89 mmHg)Initiate or increase antihypertensive medication. If target not met after 8 weeks, reduce TAVALISSE dose.14
Stage 2 (Systolic ≥140 or Diastolic ≥90 mmHg)Initiate or increase antihypertensive medication. If BP remains high, reduce TAVALISSE dose. If BP remains ≥160/100 mmHg for >4 weeks despite therapy, interrupt or discontinue.14
Hypertensive Crisis (Systolic >180 and/or Diastolic >120 mmHg)Interrupt or discontinue TAVALISSE. Manage BP aggressively. May resume at same dose if BP normalizes.14
HepatotoxicityAST/ALT ≥3x to <5x ULNIf symptomatic, interrupt TAVALISSE. If asymptomatic, monitor closely and consider dose reduction. Resume at next lower dose once LFTs normalize.14
AST/ALT ≥5x ULN (Total Bilirubin <2x ULN)Interrupt TAVALISSE. Monitor LFTs. If levels decrease, may resume at next lower dose. If levels persist for ≥2 weeks, discontinue.14
AST/ALT ≥3x ULN (Total Bilirubin >2x ULN)Discontinue TAVALISSE permanently.14
DiarrheaSevere (Grade ≥3)Interrupt TAVALISSE. Manage with supportive care. If symptoms improve to mild (Grade 1), resume at the next lower daily dose.14
NeutropeniaAbsolute Neutrophil Count (ANC) <1.0 x 10⁹/LInterrupt TAVALISSE. Monitor ANC. Once ANC resolves to >1.5 x 10⁹/L, resume at the next lower daily dose.14

Regulatory Status and Commercialization

Fostamatinib has successfully navigated the regulatory pathways in major global markets and is being commercialized through a strategic, multi-partner approach.

Global Regulatory Approvals

  • U.S. Food and Drug Administration (FDA): Fostamatinib was approved by the FDA on April 17, 2018. It is marketed in the United States under the brand name Tavalisse. The approved indication is for the treatment of thrombocytopenia in adult patients with chronic immune thrombocytopenia (ITP) who have had an insufficient response to a previous treatment. The FDA also granted fostamatinib Orphan Product designation for this indication, which provides incentives to encourage the development of drugs for rare diseases.[3]
  • European Medicines Agency (EMA): Following a positive opinion from the Committee for Medicinal Products for Human Use (CHMP) in November 2019, the EMA granted a full marketing authorization for fostamatinib on January 9, 2020.[4] In the European Union, the drug is marketed under the brand name Tavlesse. The EMA's assessment concluded that while the drug's efficacy was "modestly effective," its benefits outweighed the risks for adult patients with chronic ITP who are refractory to other treatments and for whom there is a high unmet need.[19]

Corporate and Licensing Landscape

The global development and commercialization of fostamatinib is led by its originator, Rigel Pharmaceuticals, in collaboration with key regional partners.

  • Originator/Developer: Rigel Pharmaceuticals, Inc., a U.S.-based biotechnology company headquartered in South San Francisco, California, is the original developer of fostamatinib and holds the primary rights to the molecule.[17]
  • Commercial Partners: To maximize the drug's global reach, Rigel has entered into several strategic licensing agreements:
  • Grifols: In January 2019, Rigel granted Grifols exclusive rights to commercialize fostamatinib in Europe, Turkey, the Middle East, North Africa, and CIS countries. This partnership leverages Grifols' established commercial infrastructure and expertise in hematology and rare diseases in these territories.[18]
  • Kissei Pharmaceuticals: Rigel has granted commercial rights for fostamatinib in Japan, China, Taiwan, and the Republic of Korea to Kissei Pharmaceuticals, a Japanese pharmaceutical company.[18]

This global commercialization strategy reflects a pragmatic and capital-efficient approach for a biotechnology company of Rigel's size. Launching a new drug on a global scale is a formidable logistical and financial challenge. By focusing its own commercial efforts on the U.S. market, where it launched Tavalisse independently, Rigel was able to establish a direct presence in a key market.[40] Simultaneously, the company de-risked and accelerated its international expansion by partnering with established pharmaceutical players in other major regions.

The licensing deals provide Rigel with a significant influx of non-dilutive capital through upfront payments, regulatory and commercial milestone payments, and a steady stream of future revenue from royalties on net sales.[18] This model allows Rigel to fund its ongoing research and development pipeline without bearing the full, substantial cost of building and maintaining a global sales and marketing organization. For the partners, such as Grifols and Kissei, the agreements provide access to a novel, first-in-class product that complements their existing portfolios, particularly in hematology and rare diseases. This symbiotic relationship is a common and effective business strategy in the pharmaceutical industry, enabling innovative medicines from smaller companies to reach patients worldwide. The two-year gap between the FDA and EMA approvals likely reinforced this strategy, allowing Rigel to focus on the U.S. launch while Grifols managed the latter stages of the European regulatory and reimbursement processes.[17]

Dosing, Administration, and Patient Guidance

The safe and effective use of fostamatinib requires adherence to specific guidelines for dosing, administration, and ongoing patient monitoring and counseling.

Prescribing and Dosing Regimen

  • Approved Indication: Fostamatinib is indicated for the treatment of thrombocytopenia in adult patients with chronic immune thrombocytopenia (ITP) who have had an insufficient response to a previous treatment.[3] The European indication specifies its use in patients who are refractory to other treatments.[39]
  • Initial Dose: The recommended starting dose is 100 mg administered orally twice daily.[14]
  • Dose Escalation: After one month (4 weeks) of treatment, the patient's platelet count should be assessed. If the count has not increased to at least 50 x 10⁹/L, the dose should be increased to 150 mg twice daily.[14]
  • Maintenance Dosing: The therapeutic goal is to use the lowest effective dose of fostamatinib necessary to achieve and maintain a platelet count of at least 50 x 10⁹/L, as this level is considered sufficient to reduce the risk of clinically significant bleeding.[14]
  • Discontinuation for Lack of Efficacy: If an adequate platelet response is not achieved after 12 weeks of treatment (including a period at the 150 mg BID dose), the treatment should be discontinued.[14]

Administration

  • Dosage Forms: Fostamatinib is supplied as film-coated tablets in two strengths: 100 mg (round, orange) and 150 mg (oval, orange).[14]
  • Method of Administration: The tablets should be swallowed whole with water. They may be taken with or without food.[14] Taking the tablets with food may help to mitigate potential gastrointestinal upset, such as nausea.[45]
  • Missed Dose: If a patient misses a dose, they should be instructed to skip that dose and take their next dose at its regularly scheduled time. Patients should not take two doses at once to make up for a missed dose.[14]

Patient Counseling Information

Effective patient education is critical for ensuring adherence, managing side effects, and promoting the safe use of fostamatinib. Healthcare providers should counsel patients on the following key points:

  • Risks and Monitoring: Patients must be informed about the primary risks associated with treatment, including hypertension, hepatotoxicity (liver problems), diarrhea, and neutropenia. They must understand the importance of adhering to the schedule of regular monitoring, which includes frequent blood pressure checks and monthly blood tests for liver function and complete blood counts.[14]
  • Reporting Symptoms: Patients should be instructed to immediately report any signs or symptoms of potential serious side effects. This includes symptoms of hypertensive crisis (e.g., severe headache, confusion, chest pain, shortness of breath), liver injury (e.g., jaundice, dark urine, upper right abdominal pain), severe diarrhea, or signs of infection (e.g., fever, chills, sore throat).[23]
  • Embryo-Fetal Toxicity and Contraception: The potential for fetal harm must be clearly communicated to all female patients of reproductive potential. They must be advised to use effective contraception throughout the duration of treatment and for at least one month after the final dose. A pregnancy test is required before initiating therapy.[7]
  • Lactation: Lactating women should be advised not to breastfeed during treatment and for at least one month after their last dose due to the potential risk of serious adverse reactions in the infant.[7]
  • Drug Interactions: Patients should be advised to inform their healthcare providers of all medications they are taking, including prescription and over-the-counter drugs, vitamins, and herbal supplements, to avoid potentially harmful drug interactions.[38]

Conclusion

Fostamatinib represents a significant, albeit specialized, advancement in the management of chronic immune thrombocytopenia. As the first-in-class Spleen Tyrosine Kinase (Syk) inhibitor, it offers a novel mechanism of action that directly targets the antibody-mediated platelet destruction central to ITP's pathophysiology. This distinguishes it from other therapies that either broadly suppress the immune system or stimulate platelet production.

The clinical evidence from the FIT pivotal trial program demonstrates that fostamatinib provides a meaningful benefit for a subset of the most difficult-to-treat adult patients with chronic ITP who are refractory to multiple prior therapies. While the overall response rates are modest, the responses, when they occur, are typically rapid and durable, leading to a sustained increase in platelet counts that can reduce bleeding risk and improve quality of life for these individuals. The drug's safety profile is well-defined and characterized by manageable adverse events, primarily hypertension, diarrhea, and hepatotoxicity, which necessitate a structured monitoring and dose-management strategy but are not prohibitive for most patients.

Beyond its established role in ITP, the immunomodulatory mechanism of fostamatinib provides a strong rationale for its development in other autoimmune disorders. Promising early-phase data in IgA nephropathy and an ongoing Phase 3 trial in warm autoimmune hemolytic anemia highlight its potential as a platform therapy for conditions driven by similar Syk-mediated pathways. The global commercialization strategy, leveraging partnerships with established regional players, ensures that this important therapeutic option is made available to patients worldwide.

In summary, fostamatinib (Tavalisse/Tavlesse) has carved out a crucial niche in the hematology landscape. It is not a first-line agent but rather a valuable tool for clinicians treating refractory ITP, offering a mechanistically distinct option when others have failed. Its future value will be determined by its success in ongoing and future clinical trials in other indications, which could significantly broaden its therapeutic impact.

Works cited

  1. Fostamatinib (R788) | Syk/FLT3 Inhibitor | MedChemExpress, accessed August 6, 2025, https://www.medchemexpress.com/r788.html
  2. What is the mechanism of Fostamatinib Disodium? - Patsnap Synapse, accessed August 6, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-fostamatinib-disodium
  3. Fostamatinib: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed August 6, 2025, https://go.drugbank.com/drugs/DB12010
  4. Fostamatinib | C23H26FN6O9P | CID 11671467 - PubChem, accessed August 6, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Fostamatinib
  5. Fostamatinib in chronic immune thrombocytopenia: a profile of its use in the USA - PMC, accessed August 6, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6223701/
  6. Fostamatinib - Wikipedia, accessed August 6, 2025, https://en.wikipedia.org/wiki/Fostamatinib
  7. Homepage | TAVALISSE® (fostamatinib disodium hexahydrate) tablets, accessed August 6, 2025, https://www.tavalissehcp.com/
  8. en.wikipedia.org, accessed August 6, 2025, https://en.wikipedia.org/wiki/Fostamatinib#:~:text=This%20enzyme%20is%20involved%20in,the%20likelihood%20of%20excessive%20bleeding.
  9. Full article: Fostamatinib: A Review of its Clinical Efficacy and Safety in the Management of Chronic Adult Immune Thrombocytopenia, accessed August 6, 2025, https://www.tandfonline.com/doi/full/10.2217/imt-2020-0215
  10. FDA approves fostamatinib tablets for ITP, accessed August 6, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-fostamatinib-tablets-itp
  11. Fostamatinib for the Treatment of Adult Persistent and Chronic Immune Thrombocytopenia: Results of Two Phase 3, Randomized, Placebo-Controlled Trials - ResearchGate, accessed August 6, 2025, https://www.researchgate.net/publication/324779011_Fostamatinib_for_the_Treatment_of_Adult_Persistent_and_Chronic_Immune_Thrombocytopenia_Results_of_Two_Phase_3_Randomized_Placebo-Controlled_Trials
  12. Fostamatinib for the treatment of adult persistent and chronic immune thrombocytopenia: Results of two phase 3, randomized, placebo-controlled trials - PubMed, accessed August 6, 2025, https://pubmed.ncbi.nlm.nih.gov/29696684/
  13. Fostamatinib More Effective Earlier in Treatment for Immune Thrombocytopenia | ASH Clinical News, accessed August 6, 2025, https://ashpublications.org/ashclinicalnews/news/5401/Fostamatinib-More-Effective-Earlier-in-Treatment
  14. TAVALISSE® (fostamatinib disodium ... - TAVALISSE.com, accessed August 6, 2025, https://www.tavalisse.com/downloads/pdf/Tavalisse-Full-Prescribing-Information.pdf
  15. Trial offers treatment hope for IgA nephropathy - Kidney Research UK, accessed August 6, 2025, https://www.kidneyresearchuk.org/2024/02/12/ground-breaking-clinical-trial-offers-treatment-hope-for-iga-nephropathy/
  16. A Phase 3, Multi-Center, Randomized, Double-Blind, Placebo ..., accessed August 6, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2019-05092
  17. Tavalisse (fostamatinib) FDA Approval History - Drugs.com, accessed August 6, 2025, https://www.drugs.com/history/tavalisse.html
  18. Grifols Announces Collaboration and License Agreement with Rigel Pharmaceuticals, accessed August 6, 2025, https://www.grifols.com/en/view-news/-/news/grifols-announces-collaboration-and-license-agreement-with-rigel-pharmaceuticals-to-commercialize-fostamatinib-in-europe
  19. Tavlesse | European Medicines Agency (EMA), accessed August 6, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/tavlesse
  20. Fostamatinib sodium | R788 | CAS#1025687-58-4 | 901119-35-5 - MedKoo Biosciences, accessed August 6, 2025, https://www.medkoo.com/products/4655
  21. Fostamatinib (R788) | 99.59%(HPLC) | In Stock | Syk inhibitor - Selleck Chemicals, accessed August 6, 2025, https://www.selleckchem.com/products/R7935788-Fostamatinib.html
  22. Fostamatinib | 901119-35-5 - Sigma-Aldrich, accessed August 6, 2025, https://www.sigmaaldrich.com/US/en/product/ambeedinc/ambh9a260500?context=bbe
  23. Tavalisse (fostamatinib): Side Effects, Dosage & Uses - Drugs.com, accessed August 6, 2025, https://www.drugs.com/tavalisse.html
  24. Fostamatinib for the treatment of chronic immune thrombocytopenia | Blood | American Society of Hematology, accessed August 6, 2025, https://ashpublications.org/blood/article/133/19/2027/273805/Fostamatinib-for-the-treatment-of-chronic-immune
  25. Showing BioInteractions for Fostamatinib (DB12010) | DrugBank Online, accessed August 6, 2025, https://go.drugbank.com/drugs/DB12010/biointeractions
  26. Fostamatinib for the treatment of adult persistent and chronic ..., accessed August 6, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6055608/
  27. Drug fails to meet endpoint in phase 3 ITP trial | MDedge - The Hospitalist, accessed August 6, 2025, https://community.the-hospitalist.org/content/drug-fails-meet-endpoint-phase-3-itp-trial
  28. Rigel Announces Results from the Second FIT Phase 3 Study and ..., accessed August 6, 2025, https://www.rigel.com/news-media/press-releases/detail/183/rigel-announces-results-from-the-second-fit-phase-3-study
  29. EMA Panel Backs Fostamatinib (Tavalisse) for Chronic ITP - Medscape, accessed August 6, 2025, https://www.medscape.com/viewarticle/921452
  30. Randomized Trial on the Effect of an Oral Spleen Tyrosine Kinase Inhibitor in the Treatment of IgA Nephropathy, accessed August 6, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10719605/
  31. Randomized Trial on the Effect of an Oral Spleen Tyrosine Kinase Inhibitor in the Treatment of IgA Nephropathy - PubMed, accessed August 6, 2025, https://pubmed.ncbi.nlm.nih.gov/38106605/
  32. Safety and Efficacy Study of Fostamatinib to Treat Immunoglobin A (IgA) Nephropathy - Clinical Trials | Stanford Medicine, accessed August 6, 2025, https://clinicaltrials.stanford.edu/trials/s/NCT02112838.html
  33. Clinical Trials Using Fostamatinib Disodium - NCI, accessed August 6, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/fostamatinib-disodium?pn=1
  34. Fostamatinib for Hospitalized Adults With COVID-19 and Hypoxemia: A Randomized Clinical Trial - PubMed Central, accessed August 6, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11615712/
  35. Fostamatinib Completed Phase 2 Trials for Diffuse Large B-Cell Lymphoma (DLBCL) Treatment | DrugBank Online, accessed August 6, 2025, https://go.drugbank.com/drugs/DB12010/clinical_trials?conditions=DBCOND0028221&phase=2&purpose=treatment&status=completed
  36. Rigel's Fostamatinib Being Studied by National Institute of Health in Patients with Sickle Cell Disease, accessed August 6, 2025, https://www.rigel.com/news-media/press-releases/detail/408/rigels-fostamatinib-being-studied-by-national-institute-of
  37. Fostamatinib Side Effects: Common, Severe, Long Term - Drugs.com, accessed August 6, 2025, https://www.drugs.com/sfx/fostamatinib-side-effects.html
  38. Homepage | TAVALISSE® (fostamatinib disodium hexahydrate ..., accessed August 6, 2025, https://www.tavalisse.com/
  39. Tavlesse 100 mg film-coated tablets - Summary of Product Characteristics (SmPC) - (emc), accessed August 6, 2025, https://www.medicines.org.uk/emc/product/11479/smpc
  40. Rigel Announces FDA Approval of TAVALISSE™ (fostamatinib disodium hexahydrate) for Chronic Immune Thrombocytopenia (ITP) in Adult Patients, accessed August 6, 2025, https://www.rigel.com/news-media/press-releases/detail/228/rigel-announces-fda-approval-of-tavalisse-fostamatinib
  41. EMA Panel Backs Fostamatinib (Tavalisse) for Chronic ITP - International ITP Alliance, accessed August 6, 2025, https://www.globalitp.org/index.php/about-itp/global-itp-news/154-ema-fostamatinib
  42. TAVLESSE - Orphanet, accessed August 6, 2025, https://www.orpha.net/en/drug/trade_name/642344?name=&mode=pat®ion=&status=all
  43. Grifols' TAVLESSE®(fostamatinib) receives NICE recommendation for treating refractory chronic immune thrombocytopenia, accessed August 6, 2025, https://www.grifols.com/documents/3627767/4667722/np-20221019-en.pdf/bd91d6be-c9cc-c924-05e8-3562ac3ac484?t=1666168690580
  44. Rigel Pharmaceuticals - Fostamatinib - AdisInsight - Springer, accessed August 6, 2025, https://adisinsight.springer.com/drugs/800021937
  45. Tavlesse 100 mg film-coated tablets - Patient Information Leaflet (PIL) - (emc) | 11479, accessed August 6, 2025, https://www.medicines.org.uk/emc/product/11479/pil
  46. Tavalisse (fostamatinib): Uses, Side Effects, Interactions, Pictures, Warnings & Dosing, accessed August 6, 2025, https://www.webmd.com/drugs/2/drug-175200/tavalisse-oral/details
  47. Fostamatinib Uses, Side Effects & Warnings - Drugs.com, accessed August 6, 2025, https://www.drugs.com/mtm/fostamatinib.html

Published at: August 6, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.