C12H7Cl3O2
3380-34-5
Acne, Dental Cavity, Dermabrasion, Gingivitis, Plaque, Dental, Abrasions, Bacterial skin infections, Minor burns, Oral infections
Flonoltinib maleate is an orally administered, investigational small molecule inhibitor representing a new generation of targeted therapy for myeloproliferative neoplasms (MPNs). Developed by Chengdu Zenitar Biomedical Technology Co., Ltd., Flonoltinib is engineered to overcome the limitations of existing Janus kinase (JAK) inhibitors through a novel and highly selective mechanism of action. It functions as a potent dual inhibitor of JAK2 and FMS-like tyrosine kinase 3 (FLT3), with a unique molecular interaction that involves simultaneous binding to both the active kinase domain (JH1) and the regulatory pseudokinase domain (JH2) of JAK2. This dual-domain engagement is the structural basis for its exceptional selectivity for JAK2 over other JAK family members, a feature designed to minimize off-target toxicities.
Preclinical studies have validated this mechanism, demonstrating potent anti-neoplastic activity in MPN cell lines and patient-derived cells, as well as superior efficacy in murine models of myelofibrosis, where it significantly reduced splenomegaly, suppressed bone marrow fibrosis, and prolonged survival. The drug exhibits a favorable preclinical pharmacokinetic profile characterized by good oral bioavailability and a primary hepato-biliary excretion pathway.
The most compelling evidence for Flonoltinib's potential comes from a first-in-human Phase I/IIa clinical trial (NCT05153343) in patients with intermediate- to high-risk myelofibrosis. The study reported outstanding efficacy, with 77.3% of patients achieving a spleen volume reduction of at least 35% (SVR35) at 24 weeks—a rate substantially higher than historical benchmarks for the current standard of care, ruxolitinib. Furthermore, 76.7% of patients experienced a significant improvement in symptom burden (TSS50), and 26.1% showed an improvement in bone marrow fibrosis, suggesting potential disease-modifying activity. Critically, this high level of efficacy was observed in both treatment-naïve patients and those previously exposed to other JAK inhibitors, highlighting its potential in the difficult-to-treat second-line setting.
The clinical safety profile is described as manageable. While hematologic adverse events such as anemia and thrombocytopenia were the most common, the data suggest a differentiated profile characterized by stable mean platelet counts and normalized neutrophil levels, consistent with the drug's high on-target selectivity. Currently in Phase II development in China, Flonoltinib maleate is positioned as a potential best-in-class therapy for myelofibrosis and other MPNs, with the potential to redefine the standard of care by offering superior efficacy and an improved safety window.
Flonoltinib maleate, also identified by the synonym JAK2/FLT3-IN-1, is an orally active, small molecule investigational drug at the forefront of next-generation targeted therapies for hematologic malignancies.[1] It is being developed by Chengdu Zenitar Biomedical Technology Co., Ltd., a biopharmaceutical company founded in April 2019 and located in the Chengdu High-tech Zone Biomedical Incubation Park in Sichuan, China.[3] The emergence of this promising candidate from a relatively new company underscores the rapid growth and increasing innovation within China's biotechnology sector.
The primary therapeutic focus for Flonoltinib maleate is the treatment of myeloproliferative neoplasms (MPNs), a group of clonal hematopoietic disorders. The development program is actively investigating its utility in myelofibrosis (MF), polycythemia vera (PV), and essential thrombocythemia (ET).[3] The drug's mechanism of action has also prompted exploratory development for other conditions, including graft-versus-host disease, acute myeloid leukemia (AML), and even non-oncology indications such as COVID-19, although MPNs remain the core strategic focus.[3]
Flonoltinib maleate is the maleate salt form of the active pharmaceutical ingredient, Flonoltinib. The precise chemical structure and properties are essential for understanding its pharmacological behavior.
Active Moiety (Flonoltinib):
Salt Form (Flonoltinib Maleate):
While the maleate salt is the form advancing in clinical trials, other salt forms, such as Flonoltinib sulfate, have been synthesized for research purposes, as noted in commercial catalogs of bioactive molecules.[11]
Myeloproliferative neoplasms are a collection of blood cancers characterized by the overproduction of mature myeloid cells, driven by clonal proliferation of hematopoietic stem cells. The core molecular pathogenesis of the most common MPNs—polycythemia vera, essential thrombocythemia, and primary myelofibrosis—is the dysregulation of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway.[12]
A seminal discovery in the field was the identification of a somatic, gain-of-function mutation in the JAK2 gene, specifically a valine-to-phenylalanine substitution at codon 617 ().[13] This mutation is present in over 95% of patients with PV and approximately 50-60% of patients with ET and primary MF. The
mutation leads to constitutive, cytokine-independent activation of the JAK2 kinase, which in turn drives the uncontrolled cell proliferation and the clinical manifestations of MPNs, including splenomegaly (enlargement of the spleen due to extramedullary hematopoiesis), debilitating constitutional symptoms (such as fatigue, night sweats, and fever), and progressive bone marrow fibrosis.[13]
This clear genetic driver established the hyperactivated JAK2 protein as a prime therapeutic target. The development of JAK inhibitors, most notably the JAK1/JAK2 inhibitor ruxolitinib, revolutionized the management of intermediate- and high-risk myelofibrosis. Ruxolitinib was the first drug approved for MF and provides significant benefits in reducing spleen size and alleviating symptoms.[14] However, the efficacy of first-generation JAK inhibitors is not universal, and their use is associated with significant limitations. A substantial portion of patients achieve a suboptimal response, and nearly all patients eventually experience disease progression. Furthermore, these agents are associated with dose-limiting hematologic toxicities, particularly anemia and thrombocytopenia, which can be exacerbated by their inhibitory effects on JAK1 and other kinases.[16]
This therapeutic gap has created a clear and urgent unmet medical need for a next-generation JAK inhibitor with an improved efficacy and safety profile. The development of Flonoltinib maleate is a direct response to this need. It is not merely an incremental follow-on compound but has been strategically engineered as a "new generation" inhibitor.[19] The core design principle was to create a molecule with exceptionally high selectivity for JAK2 over other JAK family members. This enhanced selectivity is intended to maximize on-target efficacy against the primary driver of the disease while minimizing the off-target effects responsible for the dose-limiting toxicities of less selective agents. By aiming for a wider therapeutic window, Flonoltinib is positioned to potentially offer superior clinical outcomes for patients with MPNs.
Flonoltinib maleate is a potent, ATP-competitive small molecule inhibitor with a precisely defined kinase inhibition profile. Its primary activity is as a dual inhibitor of Janus kinase 2 (JAK2) and FMS-like tyrosine kinase 3 (FLT3).[1] This dual targeting is strategically relevant, as JAK2 is the core driver of MPNs, while activating mutations in FLT3 are common drivers in acute myeloid leukemia (AML), a potential and fatal transformation of MF.[3] Some evidence also suggests that Flonoltinib inhibits
Cyclin-dependent kinase 6 (CDK6), an activity that may contribute to its observed effects on reversing bone marrow fibrosis.[7]
The potency of Flonoltinib against its primary targets is exceptional. Multiple independent in vitro kinase assays have reported half-maximal inhibitory concentration () values for JAK2 in the sub-nanomolar range, typically between 0.7 nM and 0.8 nM.[1] It is equally potent against the pathogenic mutant form,
, with a reported of 1.4 nM.[2] Its inhibitory activity against FLT3 is also in the low nanomolar range, with reported
values between 4 nM and 15 nM.[1]
However, the most critical feature of Flonoltinib's profile is its high degree of selectivity. It was specifically designed to preferentially inhibit JAK2 over the other members of the JAK family (JAK1, JAK3, and TYK2). This is quantitatively demonstrated by its significantly higher values against these other kinases. For instance, one report cites values of 26 nM for JAK1 and 39 nM for JAK3, representing a ~37-fold and ~56-fold selectivity for JAK2, respectively.[1] Other reports from the clinical trial investigators claim an even more pronounced selectivity of over 600-fold for JAK2 compared to JAK1 and JAK3.[19] This high selectivity ratio is the cornerstone of its therapeutic hypothesis, aiming to reduce the off-target toxicities associated with broader JAK inhibition.
Table 1: In Vitro Kinase Inhibition Profile of Flonoltinib Maleate
| Kinase Target | (nM) | Selectivity Ratio (vs. JAK2) | Data Source(s) |
|---|---|---|---|
| JAK2 | 0.7 - 0.8 | 1.0x | 1 |
| 1.4 | ~1.8x | 2 | |
| FLT3 | 4 - 15 | ~5.7x - 21.4x | 1 |
| JAK1 | 26 - 690 | ~37x - >860x | 1 |
| JAK3 | 39 - 557 | ~56x - >690x | 1 |
| TYK2 | 65 | ~81x | 3 |
Note: values are compiled from multiple sources and may vary based on assay conditions. The selectivity ratio is calculated using the lowest reported for JAK2 (0.7 nM or 0.8 nM).
The molecular basis for Flonoltinib's remarkable selectivity lies in a unique binding mechanism that distinguishes it from all currently marketed JAK inhibitors. The JAK2 protein is composed of several domains, including a catalytically active kinase domain (JH1) and an adjacent, catalytically inactive pseudokinase domain (JH2).[13] The JH2 domain functions as a crucial negative regulator, essentially acting as a brake on the JH1 domain's activity. The disease-driving
mutation is located within this JH2 domain, and its presence disrupts the auto-inhibitory function, leading to a constitutively "on" state of the JH1 kinase.[13]
Marketed JAK inhibitors, such as ruxolitinib, primarily function by binding to the ATP-binding pocket within the active JH1 domain.[20] In contrast, preclinical structural biology studies have revealed that Flonoltinib employs a more sophisticated mechanism: it
simultaneously engages both the JH1 kinase domain and the JH2 pseudokinase domain.[13] Surface plasmon resonance assays, which measure real-time binding kinetics, have quantitatively confirmed this, showing that Flonoltinib has a stronger binding affinity for the JH2 domain than for the JH1 domain.[13] Co-crystal structure analysis has further elucidated the precise molecular interactions, confirming that Flonoltinib can stably bind within the JH2 domain.[13]
This dual-domain binding is not merely a scientific curiosity; it is the direct structural cause of Flonoltinib's high selectivity. The amino acid sequences and structures of the JH2 domains differ more significantly across the JAK family members than the highly conserved JH1 domains. By targeting this more variable JH2 region in addition to the JH1 domain, Flonoltinib achieves a level of specificity for JAK2 that is difficult to attain with inhibitors that only target the common JH1 domain. This elegant mechanism-based selectivity provides a strong molecular rationale for the favorable safety profile observed in clinical trials, particularly the relative sparing of hematologic cell lines that rely on signaling through other JAK kinases. The result is a clear and powerful "mechanism-to-clinic" narrative: the unique molecular interaction directly translates into a tangible clinical benefit.
By potently and selectively inhibiting the kinase activity of JAK2 and FLT3, Flonoltinib effectively severs the downstream signaling cascades that drive cancer cell proliferation and survival. Preclinical pharmacodynamic studies have confirmed that treatment with Flonoltinib leads to a dose-dependent down-regulation of the phosphorylation of key signaling intermediates. Specifically, it has been shown to reduce levels of phosphorylated JAK2 (p-JAK2), phosphorylated STAT3 (p-STAT3), and phosphorylated STAT5 (p-STAT5) in relevant cancer cell lines.[10] It also down-regulates phosphorylated FLT3 (p-FLT3) in cells dependent on that pathway.[1]
This blockade of critical survival signals translates directly into potent anti-cancer effects at the cellular level. A series of in vitro experiments using well-established hematologic cancer cell lines, such as MV4-11 (which harbors an FLT3 mutation) and SET-2 (which has a mutation), have demonstrated that Flonoltinib exerts profound and dose-dependent cytotoxic and cytostatic effects. At nanomolar concentrations, Flonoltinib was shown to:
These cellular effects provide the fundamental biological basis for the anti-tumor activity observed in subsequent animal models and human clinical trials.
The therapeutic potential of Flonoltinib maleate was first established through a comprehensive series of in vitro studies that demonstrated its potent activity against relevant cancer cell types. In assays measuring cell proliferation, Flonoltinib showed potent inhibitory effects on cell lines harboring the mutation, with values below 0.5 µM. Notably, its activity was stronger in these mutant cell lines compared to their wild-type counterparts, indicating a degree of selectivity for the pathogenic driver kinase.[10] Its dual-targeting nature was also confirmed, as it exhibited potent anti-proliferative activity against tumor cell lines driven by FLT3 mutations, with
values below 0.1 µM.[10]
Moving beyond established cell lines to a more clinically relevant model, researchers tested Flonoltinib's effect on primary cells isolated directly from patients with MPNs. In these experiments, Flonoltinib effectively suppressed the spontaneous, disease-characteristic formation of erythroid progenitor cell colonies (in vitro), providing direct proof-of-concept that the drug is active against the primary human cancer cells it is intended to treat.[13]
Following the promising in vitro results, the efficacy of Flonoltinib was evaluated in vivo using multiple, well-validated murine models that recapitulate the key features of human myelofibrosis. In these models, mice are engineered to express the human mutation, leading to the development of an MPN-like disease characterized by splenomegaly, bone marrow fibrosis, and shortened survival.[13]
When administered orally to these mice, Flonoltinib maleate demonstrated robust, dose-dependent, and therapeutically significant anti-cancer activity.[13] The key findings from these pivotal animal studies were:
Importantly, in preclinical studies that included head-to-head comparisons, Flonoltinib demonstrated superior efficacy and lower toxicity compared to existing, marketed JAK inhibitors, providing a strong rationale for its advancement into human clinical trials.[3]
A detailed investigation of the absorption, distribution, metabolism, and excretion (ADME) properties of Flonoltinib was conducted in rats using a radiolabeled () version of the drug, providing critical insights into its behavior in vivo.[6]
This preclinical ADME profile has significant clinical implications. The target patient population for myelofibrosis is generally older, with a median age at diagnosis around 67 years.[14] This demographic frequently presents with age-related comorbidities, including varying degrees of renal impairment. Drugs that are primarily cleared by the kidneys often require complex dose adjustments in these patients to avoid toxicity. The clear demonstration that Flonoltinib is overwhelmingly cleared via the hepato-biliary route provides a strong preclinical hypothesis that its pharmacokinetics will be less affected by renal function. While this must be confirmed in dedicated human studies, it suggests a potential for a more straightforward dosing regimen and a favorable safety profile in this important and common patient subgroup, which could represent a subtle but meaningful clinical and marketing advantage.
The clinical development of Flonoltinib maleate is being spearheaded by its developer, Chengdu Zenitar Biomedical Technology, with a primary focus on its application in hematologic malignancies within China.[7] As of late 2024, the drug has advanced to
Phase II clinical trials for several indications, including Primary Myelofibrosis, Polycythemia Vera, and Mycosis Fungoides.[6]
Several key clinical trials form the backbone of its current development program:
The NCT05153343 trial was a meticulously designed study intended to efficiently establish the safety, optimal dose, and preliminary efficacy of Flonoltinib in its primary target population.
The decision by Chengdu Zenitar to enroll both JAK inhibitor-naïve and previously treated patients in this inaugural human trial represents a notably confident and aggressive clinical development strategy. A more conventional and conservative approach would have been to first establish safety and a signal of efficacy in a single, homogeneous population (e.g., treatment-naïve only) before exploring more challenging patient groups. By simultaneously generating data for both first-line and second-line positioning, the company aimed to rapidly define the drug's potential across the full spectrum of the disease. This approach not only accelerates the overall development timeline but also addresses the significant unmet need in the post-ruxolitinib setting from the very beginning. This strategy suggests a high degree of confidence in the drug's differentiated mechanism and robust preclinical data, reflecting a clear intent to expedite its path to market for the broadest possible patient population.
The clinical data from the Phase I/IIa study (NCT05153343), presented at the 2024 ASH Annual Meeting, revealed a remarkably high level of clinical activity for Flonoltinib maleate in patients with intermediate- to high-risk myelofibrosis. The results demonstrated rapid, deep, and durable responses across the key domains of the disease: spleen size, symptom burden, and bone marrow pathology.
Reduction in spleen volume is the primary objective measure of efficacy in myelofibrosis clinical trials. In this domain, Flonoltinib produced exceptional results.
Beyond objective measures of organ size, a critical goal of myelofibrosis therapy is to alleviate the debilitating constitutional symptoms that severely impact patients' quality of life. Flonoltinib demonstrated a powerful effect on symptom improvement.
A key aspiration for new myelofibrosis therapies is the ability to modify the underlying biology of the disease, rather than just managing its symptoms. An improvement in the grade of bone marrow fibrosis is considered a strong indicator of such disease-modifying potential.
A pivotal question for any new myelofibrosis drug is its activity in patients who have already been treated with and failed the current standard of care. The study's inclusive design provided a clear answer to this question.
Table 2: Summary of Key Efficacy Outcomes from the Phase I/IIa Study (NCT05153343)
| Efficacy Endpoint | Dose-Escalation Cohort (n=15) | Dose-Expansion Cohort (n=15) | Overall Population (n=30) | JAKi-Exposed Subgroup | JAKi-Naïve Subgroup |
|---|---|---|---|---|---|
| SVR35 at Week 24 | 72.7% (8/11) | 81.8% (9/11) | 77.3% (17/22) | 70.0% (7/10) | 83.3% (10/12) |
| Best SVR35 Rate | 80.0% | 93.3% | 83.3% (25/30) | Not Reported | Not Reported |
| TSS50 Rate | 80.0% | 73.3% | 76.7% (23/30) | Not Reported | Not Reported |
| Bone Marrow Fibrosis Improvement | 16.7% | 36.4% | 26.1% | Not Reported | Not Reported |
Note: Percentages for SVR35 at Week 24 are based on the number of evaluable patients at that timepoint. Other percentages are based on the total number of patients in the cohort or subgroup. Data compiled from.[18]
The safety and tolerability of Flonoltinib maleate were primary objectives of the Phase I/IIa study. The data indicate that the drug has a manageable safety profile, consistent with its mechanism of action as a potent kinase inhibitor.
In the dose-escalation phase, the maximum tolerated dose (MTD) was established at 225 mg per day.[20] Importantly, no dose-limiting toxicities (DLTs) were observed during the critical first cycle of the Phase I portion of the trial, suggesting good initial tolerability.[20]
The most frequently reported treatment-related adverse events (TRAEs) of Grade 3 or higher were hematological in nature, which is expected for a potent inhibitor of JAK2, a kinase essential for normal hematopoiesis. The incidence rates across the overall study population were:
Grade 3 or higher non-hematological TRAEs were considerably less frequent. The most common included pneumonia (9.7%), abdominal pain (3.2%), hypertension (3.2%), decreased fibrinogen (3.2%), and abnormal liver function (3.2%).[18]
Table 3: Incidence of Grade ≥3 Treatment-Related Adverse Events (TRAEs) in the Phase I/IIa Study
| Adverse Event | Dose-Escalation Cohort (%) | Dose-Expansion Cohort (%) | Overall Population (%) |
|---|---|---|---|
| Hematological | |||
| Anemia | 50.0% | 50.0% | 50.0% |
| Thrombocytopenia | 13.3% | 43.8% | 29.0% |
| Leukopenia | 13.3% | 25.0% | 19.4% |
| Neutropenia | 6.7% | 25.0% | 16.1% |
| Non-Hematological | |||
| Pneumonia | 6.7% | 12.5% | 9.7% |
| Abdominal Pain | 0.0% | 6.3% | 3.2% |
| Hypertension | 0.0% | 6.3% | 3.2% |
Note: Incidence rates are based on the total number of enrolled patients (N=31). Data compiled from.[18]
While the incidence rates of Grade 3 anemia and thrombocytopenia are notable, the qualitative description of the hematologic safety profile reveals a crucial and potentially differentiating feature of Flonoltinib. Clinical investigators specifically reported that, despite individual instances of cytopenias, the overall platelet levels remained stable during treatment and that neutrophil levels tended to normalize.[18]
At first glance, a 29.0% rate of Grade 3 thrombocytopenia seems to contradict the claim of "stable platelet counts." This apparent discrepancy requires a nuanced interpretation. The 29.0% figure likely represents the proportion of patients who, at any point during the study, had a platelet count that fell below the threshold for a Grade 3 event. This can be influenced by several factors, including transient dips in platelet counts or the fact that a significant portion of the study population (22.6%) already had low baseline platelet counts (≤100 x /L) upon entering the trial.[18]
The more significant claim of "stability" likely refers to the trend in the mean platelet count across the entire patient population over time. Unlike less selective JAK inhibitors such as ruxolitinib, which are known to cause a predictable, dose-dependent, and often progressive decline in mean platelet counts, Flonoltinib does not appear to induce this same pattern of on-target myelosuppression. This favorable characteristic is directly attributed to its high selectivity for JAK2, which spares other signaling pathways (like those mediated by JAK1) that are important for maintaining hematopoiesis.[18] Therefore, the key safety advantage of Flonoltinib may not be the complete avoidance of cytopenias in all patients, but rather the prevention of the progressive, dose-limiting myelosuppression that often complicates treatment with other agents in this class.
The characterization of Flonoltinib's pharmacokinetic (PK) behavior in humans was a primary endpoint of the Phase I portion of the NCT05153343 study.[18] While these assessments were conducted, specific human PK parameters such as maximum concentration (
), time to maximum concentration (), elimination half-life (), and area under the curve (AUC) have not yet been made publicly available in the provided materials.
However, the progression of the clinical program provides indirect evidence of a well-behaved PK profile. The establishment of a 100 mg once-daily oral dose for the Phase II expansion suggests predictable absorption and an exposure profile suitable for convenient daily administration.[18] Furthermore, the initiation of a dedicated food-effect study (NCT07193576) in healthy volunteers using this 100 mg dose is a standard step in late-stage clinical development, aimed at providing clear guidance for patients on whether the drug should be taken with or without food.[25] This indicates that the developer is systematically characterizing the drug's PK properties in accordance with global regulatory expectations. The preclinical data, which showed good oral bioavailability and rapid clearance, provide a solid foundation for these ongoing human studies.[7]
Based on the available data, Flonoltinib maleate is strongly positioned to be a disruptive force in the treatment of myelofibrosis and other MPNs. Its potential competitive advantages are multi-faceted, spanning efficacy, safety, and breadth of application.
Flonoltinib maleate remains an investigational drug and has not yet received marketing authorization from any major regulatory agency. The provided materials contain no evidence of approval by the U.S. Food and Drug Administration (FDA), the European Medicines Agency (EMA), or the Australian Therapeutic Goods Administration (TGA).[26]
Its development is currently centered in China, where multiple Phase II trials are underway under the sponsorship of Chengdu Zenitar Biomedical Technology.[3] The logical and necessary next step in its development pathway is the initiation of a large, randomized, pivotal Phase III registration trial. This trial will likely compare Flonoltinib to the best available therapy (such as ruxolitinib) in patients with intermediate- to high-risk myelofibrosis. The success of such a trial would form the basis of a New Drug Application, first in China and potentially in other global territories thereafter.
Despite the highly promising data, several key questions remain that will be addressed as the development program matures:
The emergence of Flonoltinib is significant not only for its therapeutic potential but also for what it represents in the broader landscape of pharmaceutical innovation. The development of a potentially best-in-class molecule, characterized by a novel mechanism of action born from sophisticated, structure-based drug design, by a Chinese biotech company founded as recently as 2019, is a powerful testament to the maturation of China's biopharmaceutical ecosystem. It signals a notable shift from a historical focus on generics and "me-too" compounds towards the creation of highly differentiated, novel therapeutics capable of competing on a global stage. The successful development and presentation of Flonoltinib at major international scientific congresses like ASH indicates a clear ambition for global validation and impact. As such, the story of Flonoltinib is a compelling case study of the changing dynamics of global pharmaceutical R&D and a harbinger of future innovation from new geographic centers of excellence.
Published at: October 3, 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.