Small Molecule
C21H15ClF3NO2
1606974-33-7
Zatolmilast, also known as BPN-14770, is an investigational, first-in-class, orally administered small molecule that functions as a selective, allosteric inhibitor of phosphodiesterase-4D (PDE4D).[1] It is currently in late-stage clinical development, with its primary therapeutic indication being the treatment of Fragile X Syndrome (FXS), the leading inherited cause of intellectual disability and a common single-gene cause of autism.[1] The drug's novel mechanism of action involves the targeted modulation of the cyclic adenosine monophosphate (cAMP) signaling pathway within the central nervous system. By selectively inhibiting the activated form of PDE4D, Zatolmilast increases intracellular cAMP levels, which in turn promotes a cascade of events leading to enhanced synaptic plasticity, neuronal connectivity, and the production of critical neurotrophic factors like BDNF.[2]
A pivotal Phase 2 clinical trial (NCT03569631) in adult males with FXS yielded unequivocally positive results, demonstrating statistically significant and clinically meaningful improvements in both direct, performance-based cognitive measures and caregiver-reported assessments of language and daily functioning.[7] Importantly, the drug was found to be safe and well-tolerated, with an adverse event profile comparable to placebo, notably avoiding the gastrointestinal side effects that have limited the development of previous, less selective PDE4 inhibitors.[4]
Originally discovered by Tetra Therapeutics, Zatolmilast's development was strategically advanced following the company's acquisition by Shionogi & Co., Ltd. in 2020.[5] Shionogi is now conducting the pivotal Phase 2b/3 "EXPERIENCE" clinical trial program to confirm the Phase 2 findings in larger adolescent and adult male populations, with regulatory submissions anticipated in 2026.[12] With a portfolio of strategic regulatory designations from both the U.S. FDA and the European Commission, Zatolmilast is positioned as a potential breakthrough, disease-modifying therapy for FXS, a condition with a profound unmet medical need and no currently approved targeted treatments.[14]
Zatolmilast represents a significant advancement in the pursuit of targeted therapies for neurodevelopmental disorders. As an investigational new drug, its development has progressed from foundational academic research to a late-stage clinical program under the stewardship of a major pharmaceutical company, reflecting its considerable therapeutic promise.
Zatolmilast is a synthetic small molecule belonging to the class of acetamides, benzeneacetamides, and pyridines.[3] Its fundamental identifying information and key physicochemical properties are consolidated from multiple chemical and drug databases, as summarized in Table 2.1.
Table 2.1: Zatolmilast Drug Profile Summary
| Identifier Type | Value | Source(s) |
|---|---|---|
| Generic Name | Zatolmilast | 1 |
| Code Names/Synonyms | BPN-14770, BPN14770 | 1 |
| DrugBank ID | DB14790 | 1 |
| CAS Number | 1606974-33-7 | 1 |
| Drug Type | Small Molecule, Investigational | 1 |
| IUPAC Name | 2-[[2-(3-chlorophenyl)-6-(trifluoromethyl)pyridin-4-yl]methyl]phenyl]acetic acid | 1 |
| Chemical Formula | $C_{21}H_{15}ClF_{3}NO_{2}$ | 1 |
| Molecular Weight | 405.80 g·mol−1 | 1 |
| PubChem CID | 90111638 | 1 |
| UNII | G786V328X6 | 1 |
| InChI Key | LTSUMTMGJHPGFX-UHFFFAOYSA-N | 1 |
| Physical Appearance | A solid | 19 |
| Solubility | Soluble in Methanol, DMSO | 19 |
The trajectory of Zatolmilast from concept to late-stage asset exemplifies a successful modern paradigm for rare disease drug development, characterized by a collaborative ecosystem of patient advocacy, biotechnology innovation, and pharmaceutical investment.
The foundational science pointing toward phosphodiesterase inhibitors as a viable therapeutic target for FXS originated from academic research funded for many years by patient advocacy groups, most notably the FRAXA Research Foundation.[11] This early-stage, high-risk funding was crucial in validating the biological target.
The drug itself, Zatolmilast, was discovered and advanced through initial clinical development by Tetra Discovery Partners (later Tetra Therapeutics), a Michigan-based biotechnology company specializing in CNS disorders.[3] Tetra successfully navigated the compound through preclinical studies and into early-phase human trials, culminating in the landmark Phase 2 proof-of-concept study (NCT03569631) in adults with FXS.[9]
The announcement of unequivocally positive results from this trial in November 2020 served as a major de-risking event and a critical inflection point.[9] Shortly thereafter, in a strategic move that highlights the "build-to-buy" model prevalent in the biopharmaceutical industry, Tetra Therapeutics was acquired and became a wholly-owned subsidiary of Shionogi & Co., Ltd., a global, research-driven pharmaceutical company based in Japan.[5] This acquisition aligned with Shionogi's corporate mission to address unmet medical needs and its strategic focus on CNS disorders, providing the necessary capital and infrastructure to advance Zatolmilast into larger, more complex pivotal trials.[5] Under Shionogi's leadership, the late-stage clinical program, now branded as "EXPERIENCE," was launched to confirm the Phase 2 findings and prepare for global regulatory submissions.[11]
Zatolmilast's therapeutic potential is rooted in its precise and highly selective mechanism of action, which targets a fundamental signaling deficit in the brains of individuals with FXS and other neurological disorders.
Cyclic adenosine monophosphate (cAMP) is a ubiquitous and vital intracellular second messenger that plays a pivotal role in mediating neuronal function. Within brain cells, cAMP is integral to processes of synaptic transmission, the molecular basis of learning and memory known as synaptic plasticity, and the regulation of gene expression for neuronal growth and survival.[6] The concentration and activity of cAMP are tightly regulated by a superfamily of enzymes called phosphodiesterases (PDEs), which catalyze its degradation. The PDE4 family is particularly important in the CNS, with the PDE4D subtype being highly expressed in brain regions critical for cognition, such as the hippocampus and cortex.[2] In pathological states like FXS, the cAMP signaling pathway is known to be dysregulated, presenting a key target for therapeutic intervention.[13]
Zatolmilast is a highly differentiated PDE4 inhibitor that achieves its therapeutic effect through a sophisticated and selective mechanism.
This state-dependent mechanism represents a significant pharmacological advance. First-generation, non-selective PDE4 inhibitors failed in clinical development for CNS indications due to dose-limiting side effects, particularly severe nausea and emesis, which are caused by broad inhibition of PDE4 subtypes in the gastrointestinal tract and brainstem.[9] By selectively targeting only the activated form of the PDE4D subtype, Zatolmilast is hypothesized to enhance cAMP signaling primarily in neurons where the pathway is already active, thus providing the desired cognitive benefit while largely avoiding the off-target effects that plagued its predecessors. This creates a viable therapeutic window, separating CNS efficacy from peripheral toxicity.
The inhibition of activated PDE4D by Zatolmilast initiates a well-defined downstream signaling cascade that is believed to correct the underlying neurobiological deficits in FXS.
The rationale for advancing Zatolmilast into human trials was supported by robust preclinical data. In studies using healthy mice, oral administration of Zatolmilast at doses of 0.3 mg/kg and higher significantly enhanced performance in the novel object recognition task, a standard behavioral assay for learning and memory.[19]
More importantly, in the Fmr1-/- mouse model, which genetically recapitulates FXS, Zatolmilast demonstrated disease-modifying potential. At a low dose of 0.3 mg/kg, the drug produced anxiolytic-like effects, reducing hyperarousal and increasing social interaction time, and also normalized other characteristic behaviors like nesting and marble-burying.[19] The successful translation of these findings to the positive human Phase 2 trial was a significant achievement, validating the predictive utility of the Fmr1-/- mouse model for this specific therapeutic mechanism, a point of contrast with previously failed trials for other drug classes in FXS.[9]
A critical attribute for any CNS-targeted therapeutic is a favorable pharmacokinetic profile that ensures adequate drug exposure at the target site within the brain. Preclinical and early clinical studies indicate that Zatolmilast possesses such properties.
Studies in mice have characterized the absorption, distribution, metabolism, and excretion (ADME) profile of Zatolmilast, revealing features highly conducive to a CNS drug.
While a comprehensive, peer-reviewed publication of the human ADME profile is not yet available, the ongoing clinical program is designed to fully characterize these properties. Phase 1 single ascending dose (NCT02648672) and multiple ascending dose (NCT02840279) studies in healthy young and elderly volunteers have been completed, providing the initial safety and pharmacokinetic data necessary to proceed to later-stage trials.[17]
To support a future New Drug Application (NDA), Shionogi is conducting a standard battery of dedicated Phase 1 pharmacokinetic studies in specific populations, including participants with severe renal impairment (NCT07012005) and those with mild, moderate, or severe hepatic impairment (NCT07018492).[31] Furthermore, a drug-drug interaction study (NCT07011992) assessing the effect of Zatolmilast on rosuvastatin has been completed, providing insight into its potential interactions with common metabolic pathways and transporters.[32] Pharmacokinetic and biomarker data continue to be collected as secondary objectives in the ongoing Phase 2 and 3 trials.[33]
The clinical development of Zatolmilast is most advanced in FXS, where it has progressed to pivotal late-stage trials based on a strong therapeutic rationale and compelling proof-of-concept data. The overall clinical program is extensive, spanning multiple indications and phases, as summarized in Table 5.0.
Table 5.0: Summary of Zatolmilast Clinical Trials Program
| Trial ID (NCT) | Phase | Indication(s) | Status | Purpose | Design Summary |
|---|---|---|---|---|---|
| NCT03569631 | 2 | Fragile X Syndrome (FXS) | Completed | Treatment | Randomized, Double-Blind, Placebo-Controlled, 2-Period Crossover |
| NCT05163808 | 2/3 | FXS | Active, Not Recruiting | Treatment | Randomized, Double-Blind, Placebo-Controlled, Parallel Group (Adolescents) |
| NCT05358886 | 2/3 | FXS | Active, Not Recruiting | Treatment | Randomized, Double-Blind, Placebo-Controlled, Parallel Group (Adults) |
| NCT05367960 | 3 | FXS | Active, Not Recruiting | Treatment | Open-Label Extension |
| NCT06717438 | 2 | Jordan's Syndrome | Recruiting | Treatment | Randomized, Double-Blind, Placebo-Controlled |
| NCT03817684 | 2 | Alzheimer's Disease (AD) | Completed | Treatment | Randomized, Placebo-Controlled |
| NCT03030105 | 1 | AD (Cognitive Impairment Model) | Completed | Treatment | Randomized, Crossover |
| NCT02840279 | 1 | Healthy Volunteers | Completed | Treatment | Multiple Ascending Dose |
| NCT02648672 | 1 | Healthy Volunteers | Completed | Treatment | Single Ascending Dose |
| NCT03861000 | 1/2 | Depressive Illness | Completed | Diagnostic | PET Radioligand Study |
| NCT07012005 | 1 | Renal Impairment | Not Yet Recruiting | Treatment | Pharmacokinetics |
| NCT07018492 | 1 | Hepatic Impairment | Recruiting | Treatment | Pharmacokinetics |
| NCT07011992 | 1 | Healthy Volunteers | Completed | Treatment | Drug-Drug Interaction |
FXS is a neurodevelopmental disorder arising from a CGG trinucleotide repeat expansion (over 200 repeats) in the 5' untranslated region of the Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene on the X chromosome.[13] This genetic anomaly leads to hypermethylation and transcriptional silencing of the gene, resulting in a profound deficiency or complete absence of its protein product, FMRP.[13] FMRP is a critical RNA-binding protein that regulates local protein synthesis at synapses, a process essential for the development and plasticity of neuronal connections.[13] The loss of FMRP disrupts this delicate balance, leading to immature synaptic structures and impaired signaling, including a well-documented deficit in the cAMP pathway.[9]
Clinically, FXS manifests as a spectrum of intellectual disability, behavioral challenges (anxiety, hyperactivity, aggression), and features of autism spectrum disorder.[4] Currently, there are no approved pharmacological agents that treat the core cognitive deficits of FXS.[15] The standard of care is supportive, relying on educational interventions, various therapies (speech, occupational, behavioral), and medications to manage co-occurring symptoms like ADHD and anxiety.[13] Zatolmilast is designed to address the underlying pathophysiology by correcting the cAMP signaling deficit, thereby offering the potential for disease modification.
The clinical proof-of-concept for Zatolmilast in FXS was established in a landmark Phase 2 study that produced exceptionally strong and consistent results.
Table 5.2: Detailed Efficacy Outcomes of the Phase 2 FXS Trial (NCT03569631)
| Outcome Domain | Assessment Tool | Least Squares (LS) Mean Difference (Drug vs. Placebo) | p-value | Interpretation |
|---|---|---|---|---|
| Cognition | NIH Toolbox Cognition Crystallized Composite (CCC) Score | +5.31 | 0.0018 | Statistically significant improvement in overall crystallized intelligence. |
| Cognition | NIH Toolbox Oral Reading Recognition | +2.81 | 0.0157 | Statistically significant improvement in reading ability. |
| Cognition | NIH Toolbox Picture Vocabulary | +5.81 | 0.0342 | Statistically significant improvement in vocabulary. |
| Function | Caregiver Visual Analog Scale (VAS) - Language | +14.04 | 0.0051 | Clinically meaningful and statistically significant improvement in language skills as observed by caregivers. |
| Function | Caregiver Visual Analog Scale (VAS) - Daily Functioning | +14.53 | 0.0017 | Clinically meaningful and statistically significant improvement in the ability to perform daily tasks as observed by caregivers. |
| Data sourced from.7 |
Building on the success of the Phase 2 trial, Shionogi launched the "EXPERIENCE" (Evaluation of Fragile X Experience in Cognition Expression) program, a set of late-stage studies designed to provide the confirmatory evidence needed for regulatory approval.[7]
The primary endpoint for both pivotal trials is the change from baseline in the NIH-TCB Cognition Crystallized Composite score, the same key cognitive measure that showed a robust effect in the Phase 2 study.[4]
Across all completed studies in the FXS program, Zatolmilast has demonstrated a favorable safety profile. It has been consistently well-tolerated, with an adverse event profile that is largely indistinguishable from placebo.[4] The absence of significant dose-limiting gastrointestinal side effects remains a key differentiating feature of this second-generation PDE4 inhibitor, supporting its potential for chronic administration in this patient population.[9] Long-term safety will be further defined by the ongoing OLE study.
Leveraging its mechanism of action and established safety profile, Zatolmilast is also being investigated for other CNS disorders characterized by cognitive or synaptic dysfunction, demonstrating a potential "pipeline-in-a-product" strategy.
The therapeutic rationale for Zatolmilast in AD is strong, as synaptic loss and dysfunction are core pathological features of the disease, and enhancing the cAMP-CREB-BDNF pathway could be neuroprotective and procognitive.[6] The clinical development for AD has reached Phase 2.[3] Several Phase 1 studies in healthy volunteers have been completed, including one (NCT03030105) that used a scopolamine-induced cognitive impairment model relevant to AD.[30] A Phase 2 study in subjects with early AD, the "PICASSO AD Trial" (NCT03817684), has also been completed.[41] However, the results of this trial have not been publicly disclosed. The fact that the developer lists AD as an indication available for licensing suggests that while the drug may have potential, the company has strategically prioritized the FXS program, which has a clearer and potentially faster regulatory path, and may be seeking a partner to fund the larger, more expensive trials required for an AD indication.[3]
Zatolmilast is being explored in Jordan's Syndrome, an ultra-rare genetic disorder caused by mutations in the PPP2R5D gene that also leads to global developmental delays and intellectual disability.[7] Preclinical research in animal models of the disorder provided a scientific basis for its evaluation in humans.[33] This expansion demonstrates a strategic "rare disease roll-up" approach, where a promising therapy for one neurodevelopmental disorder is systematically tested in others with phenotypic overlap and high unmet need.
A Phase 2 clinical trial (NCT06717438) was initiated in May 2025 to evaluate Zatolmilast in this population.[7] The study is a randomized, double-blind, placebo-controlled trial designed to enroll 30 participants aged 9-45.[7] The primary endpoint is safety and tolerability, with secondary endpoints assessing preliminary efficacy and collecting pharmacokinetic data.[33] The study includes a 24-week double-blind period followed by a 24-week open-label extension and is expected to conclude in late 2026.[7]
Early-stage interest in Zatolmilast's potential for psychiatric disorders is evidenced by a completed Phase 1/2 study (NCT03861000) that used the compound as part of a PET radioligand diagnostic evaluation for depressive illness.[47] This aligns with the broader literature on PDE4 inhibitors in depression, though this indication does not appear to be in active late-stage development for Zatolmilast.
Zatolmilast's path through clinical development has been significantly facilitated by a comprehensive suite of strategic regulatory designations from global health authorities, reflecting its potential to address a serious unmet medical need.
The development program for Zatolmilast has been awarded multiple designations that expedite its review and provide significant commercial incentives. This collection of designations signals a strong alignment between the developer and regulatory agencies regarding the drug's potential. The key designations are summarized in Table 7.1.
Table 7.1: Summary of Global Regulatory Designations for Zatolmilast
| Designation | Regulatory Body | Indication | Date Granted | Key Benefits/Implications |
|---|---|---|---|---|
| Orphan Drug Designation | U.S. FDA | Fragile X Syndrome | Mar 2018 | 7 years of market exclusivity post-approval, tax credits for R&D, waiver of PDUFA fees. |
| Rare Pediatric Disease (RPD) Designation | U.S. FDA | Fragile X Syndrome | Sep 2023 | Eligible for a Priority Review Voucher (PRV) upon approval, which can be used to expedite review of another drug or sold. |
| Fast Track Designation | U.S. FDA | Fragile X Syndrome | - | More frequent meetings with FDA, eligibility for Accelerated Approval and Priority Review. |
| Orphan Medicinal Product Designation | European Commission | Fragile X Syndrome | Apr 2024 | 10 years of market exclusivity in the EU, protocol assistance, and fee reductions. |
| Data sourced from.3 |
The current standard of care for FXS is entirely supportive and symptomatic, as there are no approved therapies that target the underlying disease mechanism or its core cognitive symptoms.[13] Management is multidisciplinary and includes developmental and educational interventions (e.g., special education, speech-language therapy, occupational therapy) and symptomatic pharmacotherapy for co-morbid conditions.[13] Medications such as stimulants (methylphenidate) are used for ADHD symptoms, while selective serotonin reuptake inhibitors (SSRIs) like sertraline and fluoxetine are used for anxiety and obsessive-compulsive behaviors.[13]
Zatolmilast is not positioned to replace these supportive measures but rather to be the first therapy to address the core intellectual and cognitive deficits of the syndrome.[2] If approved, it would represent a paradigm shift, moving beyond symptom management to a potentially disease-modifying treatment. It is a leading candidate in the FXS pipeline, distinguished by its unique, well-tolerated mechanism and strong Phase 2 clinical data.[9]
Zatolmilast stands as a leading candidate in the quest for a targeted therapy for Fragile X Syndrome. A comprehensive assessment of its development program reveals a profile with significant strengths and a clear path forward, albeit with the inherent risks of late-stage clinical trials.
The future of Zatolmilast hinges on the outcomes of the pivotal EXPERIENCE program. The readouts from the adolescent (NCT05163808) and adult (NCT05358886) studies are the most critical near-term milestones. Positive results would support regulatory submissions, which the developer plans for the first half of 2026.[13]
Key questions that these trials will answer include whether the magnitude of cognitive and functional improvement seen in Phase 2 can be replicated in a larger, more diverse population, and whether the therapeutic effect is consistent across both adolescent and adult age groups. Concurrently, the long-term open-label extension study (NCT05367960) will be crucial for understanding the durability of the treatment effect and establishing a long-term safety profile.
Zatolmilast represents one of the most scientifically sound and clinically promising therapeutic candidates for Fragile X Syndrome to date. Its development is built upon a strong biological rationale, a sophisticated and well-differentiated molecular mechanism, compelling preclinical validation, and a landmark Phase 2 trial that demonstrated a robust, consistent, and clinically meaningful impact on cognition and function. While the challenges of late-stage development should not be underestimated, the totality of the evidence—from its molecular design to its regulatory status—suggests a high probability of success in the pivotal program. If approved, Zatolmilast has the potential to become a foundational, first-in-class therapy that fundamentally alters the treatment landscape for FXS. It would offer the first pharmacological intervention aimed at improving the core intellectual disability, providing tangible hope for enhanced quality of life for thousands of individuals and their families.
Published at: October 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.