MedPath

DB-959Na Advanced Drug Monograph

Published:Jun 11, 2025

Generic Name

DB-959Na

Comprehensive Report on DB-959Na (T3D-959)

Executive Summary

DB-959Na, the sodium salt of T3D-959, is an orally administered small molecule investigational drug being developed primarily for Alzheimer's Disease (AD). It functions as a dual peroxisome proliferator-activated receptor (PPAR) agonist, with a notable 15-fold higher potency for PPARδ compared to PPARγ. This mechanism targets fundamental metabolic dysfunctions, including impaired glucose and lipid metabolism and insulin resistance, which are increasingly recognized as core components of AD pathology. Originally developed for Type 2 Diabetes and dyslipidemia, T3D-959 was repositioned by T3D Therapeutics for AD, leveraging its brain-penetrant properties and its potential to address neuroinflammation and other upstream pathological processes.

Preclinical studies, predominantly in intracerebral streptozotocin (i.c. STZ)-induced rat models of sporadic AD, demonstrated broad efficacy. T3D-959 improved cognitive and motor functions, reduced Aβ and phospho-tau pathology, normalized metabolic signaling pathways, and decreased neuroinflammation. These promising results, coupled with a favorable early safety profile in Phase 1 studies, supported its progression into AD patient trials.

The Phase 1/2a exploratory study (NCT02560753) in mild-to-moderate AD patients provided initial signals of target engagement, including positive changes in brain glucose metabolism (FDG-PET) and plasma metabolic markers, alongside preliminary cognitive improvements. However, the subsequent Phase 2 PIONEER trial (NCT04251182), a larger, placebo-controlled study, faced significant challenges. While the original intent-to-treat (ITT) analysis failed to meet its primary cognitive and functional endpoints, post-hoc analysis of a modified ITT (mITT) population (N=141), established after excluding data from five sites with significant irregularities (including lack of drug in patient plasma and improbable AD diagnoses), showed that the primary endpoints approached statistical significance. Specifically, the 30mg dose of T3D-959 showed a trend towards slowing cognitive decline on the ADAS-Cog11, and the 15mg dose showed a similar trend on the ADCS-CGIC.

Importantly, in the mITT population, T3D-959 demonstrated statistically significant improvements in key AD biomarkers, including the plasma Aβ42/40 ratio (comparable in effect size to lecanemab at 6 months) and neurogranin, a marker of synaptic degeneration. Effects were also seen on markers of inflammation, insulin resistance, and oxidative stress. A patient subgroup with high baseline pTau-217, indicative of AD pathology, showed more pronounced cognitive benefits with the 30mg dose. Critically, T3D-959 has consistently shown a favorable safety profile across trials, with no evidence of amyloid-related imaging abnormalities (ARIA), a significant concern with amyloid-targeting antibody therapies.

T3D Therapeutics plans to advance T3D-959 into a Phase 2b/3 trial, focusing on the 30mg dose and utilizing plasma biomarkers for patient selection. While the data irregularities in the PIONEER trial necessitate cautious interpretation of the mITT results, the drug's unique metabolic mechanism, oral administration, and favorable safety profile, particularly the absence of ARIA, position it as a potentially valuable and differentiated therapeutic candidate for AD, especially if efficacy can be robustly demonstrated in future, well-controlled trials targeting biomarker-defined populations. Its potential also extends to other neurodegenerative conditions like Huntington's Disease.

1. Introduction to DB-959Na (T3D-959)

DB-959Na, also widely referred to by its free acid form T3D-959, is an investigational small molecule drug currently under clinical development primarily for the treatment of Alzheimer's Disease (AD). Its development represents a shift in therapeutic strategy, focusing on the metabolic underpinnings of neurodegeneration.

**1.1. Chemical Profile and Pharmaceutical Properties** DB-959Na is the sodium salt of the active pharmaceutical ingredient (1S)-5-(2-(5-Ethyl-2-(4-methoxyphenyl)-4-oxazolyl)ethoxy)-2,3-dihydro-1H-indene-1-acetic acid (T3D-959).[1, 2] The use of a sodium salt form is a common pharmaceutical practice often employed to enhance physicochemical properties such as solubility, stability, or bioavailability of the active free acid. T3D-959 (the free acid) has a molecular formula of <span class="math-inline">C\_\{25\}H\_\{27\}NO\_5</span> and a molecular weight of 421.4856 g/mol.[1] The corresponding sodium salt, DB-959Na, has a molecular formula of <span class="math-inline">C\_\{25\}H\_\{26\}NNaO\_5</span> and a molecular weight of approximately 443.5 g/mol.[3, 4] The CAS Registry Number for DB-959Na is 1258076-66-2.[2, 4] Synonyms for the compound include T3D-959 and DB-959 (referring to the free acid).[3, 4]

Pharmacokinetically, T3D-959 is designed for once-daily oral administration and is reported to be blood-brain barrier (BBB) penetrant, a critical characteristic for drugs targeting central nervous system (CNS) disorders like AD.[3, 5, 6, 7, 8, 9, 10, 11] The half-life of T3D-959 has been reported to be in the range of 14.8-19.9 hours.[3] This pharmacokinetic profile supports the once-daily dosing regimen explored in clinical trials. The ability to penetrate the BBB is fundamental for T3D-959 to exert its proposed neuroprotective effects directly within the brain.

**Table 1: Chemical and Pharmacokinetic Properties of DB-959Na (T3D-959)**

PropertyDetailReference(s)
Common Name(s)DB-959Na, T3D-959, DB-959 (free acid)3
Chemical Name (free acid)(1S)-5-(2-(5-Ethyl-2-(4-methoxyphenyl)-4-oxazolyl)ethoxy)-2,3-dihydro-1H-indene-1-acetic acid1
Molecular FormulaC25​H27​NO5​ (free acid)1
C25​H26​NNaO5​ (sodium salt)2
Molecular Weight421.4856 g/mol (free acid)1
443.5 g/mol (sodium salt)2
CAS Registry Number1258076-66-2 (sodium salt)2
Route of AdministrationOral3
Half-life14.8-19.9 hours3
BBB PenetrationPenetrant3

**1.2. Development Timeline and Key Stakeholders** The developmental history of DB-959Na (T3D-959) involves a strategic repositioning. The compound, originally known as DB959, was first developed by Dara Therapeutics (under license from Bayer HealthCare Pharmaceuticals) as a potential treatment for Type 2 Diabetes and dyslipidemia.[1, 3, 12] This initial focus aligns with its mechanism of action as a PPAR agonist, given the central role of these receptors in metabolic regulation.

In June 2013, a significant shift occurred when T3D Therapeutics, Inc. acquired exclusive worldwide rights from DARA BioSciences to develop and commercialize DB959 and its related compounds.[3, 12] T3D Therapeutics redirected the clinical development program towards AD, based on the growing understanding of metabolic dysfunction as a key pathological component of this neurodegenerative disorder and the compound's specific PPAR agonistic profile.[3, 5, 13] The compound was subsequently renamed T3D-959. T3D Therapeutics is the current developer, spearheading its investigation for AD and also exploring its potential for other neurodegenerative conditions, such as Huntington's Disease.[1, 4, 5, 6, 7, 8, 9, 10, 11, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22] This multi-company involvement, from originator (Bayer) to initial developer (DARA) and current specialized developer (T3D), reflects a common trajectory in the pharmaceutical industry where assets are transferred to entities with specific expertise or strategic focus.

2. Pharmacological Profile

**2.1. Mechanism of Action: Dual PPARδ/γ Agonism** T3D-959 is characterized as a dual peroxisome proliferator-activated receptor (PPAR) agonist, exerting its effects through the activation of two PPAR subtypes: PPARδ (delta) and PPARγ (gamma).[3, 4, 6, 11, 12, 22, 23] A key feature of its pharmacological profile is its biased agonism; T3D-959 exhibits a 15-fold greater potency for PPARδ (human <span class="math-inline">ED\_\{50\}</span> = 19 nM) compared to PPARγ (human <span class="math-inline">ED\_\{50\}</span> = 297 nM).[3, 4, 6, 11, 12, 22]

PPARs are ligand-activated transcription factors belonging to the nuclear hormone receptor superfamily. They play crucial roles in the regulation of numerous biological processes, most notably metabolic homeostasis (including glucose and lipid metabolism) and inflammation.[3, 6, 9, 10, 11, 12, 22, 23, 24] The expression pattern of these receptors is tissue-specific, with PPARγ being highly expressed in adipose tissue and involved in adipogenesis and insulin sensitization, while PPARδ is more ubiquitously expressed, including high levels in the brain, particularly in regions critical for energy homeostasis such as the mediobasal hypothalamus.[6, 9, 10]

The preferential activation of PPARδ by T3D-959, combined with its BBB penetrance, suggests a mechanism tailored for CNS effects. This specific agonistic profile might offer a distinct therapeutic advantage. While PPARγ agonism is linked to insulin sensitization (beneficial in AD's context of brain insulin resistance), strong and selective PPARγ agonists like older thiazolidinediones have been associated with side effects such as weight gain and fluid retention.[3] T3D-959's biased agonism, favoring PPARδ, may allow it to harness the neuroprotective and metabolic benefits associated with both receptor subtypes while potentially mitigating the adverse effects primarily linked to potent PPARγ activation. Indeed, preclinical studies with T3D-959 did not report body weight increases typically seen with some chronic PPARγ agonists.[25]

The therapeutic strategy of T3D-959 is aimed at addressing what are considered upstream metabolic dysfunctions in AD, rather than directly targeting the downstream pathological hallmarks such as amyloid plaques and neurofibrillary (tau) tangles. This approach aligns with the growing "metabolic hypothesis" of AD, which posits that impaired brain energy metabolism and insulin resistance are fundamental drivers of the disease.[1, 3, 4, 5, 6, 7, 8, 9, 10, 11, 13, 14, 16, 17, 22, 25, 26, 27]

**2.2. Effects on Neuro-Metabolism and Inflammation** T3D-959's primary therapeutic goal in AD is the amelioration of dysfunctional brain glucose and lipid metabolism.[4, 5, 6, 7, 8, 9, 10, 11, 13, 16, 17, 22] This includes addressing the phenomenon of insulin resistance within the brain, a condition increasingly linked to AD pathogenesis and sometimes referred to as "Type 3 diabetes" or a "brain form of diabetes".[1, 3, 4, 17, 25, 26, 27] By improving insulin sensitivity and glucose utilization, T3D-959 aims to restore energy homeostasis in compromised neurons.

In addition to its effects on glucose and lipid metabolism, T3D-959 is also reported to reduce neuroinflammation.[3, 4, 8, 9, 10, 16, 17, 18, 19, 21, 22, 23, 27] Neuroinflammation is a critical component of AD, contributing to neuronal damage and disease progression. PPARs, particularly PPARγ, are known to have anti-inflammatory properties.[23]

Clinical evidence from the Phase 1/2a trial (NCT02560753) supports T3D-959's impact on metabolic profiles. Treatment led to dose-dependent changes in the plasma metabolome, including decreases in branched-chain amino acids (isoleucine, leucine, and valine), which are often elevated in insulin-resistant states. Furthermore, an increase in the ratio of glycine levels (end-of-treatment/baseline), another marker suggestive of improved insulin sensitivity, was observed at the highest dose tested.[3, 4] These systemic metabolic changes suggest target engagement and provide a peripheral correlate to the intended CNS metabolic improvements. The multifaceted action of T3D-959—simultaneously addressing glucose metabolism, lipid metabolism, insulin sensitivity, and neuroinflammation—positions it as a therapy that could potentially counteract several interconnected pathological pathways in AD. This contrasts with many therapeutic approaches that focus on a single target, and may offer a more holistic and potentially more effective means of modifying the disease course.

3. Preclinical Evaluation

**3.1. Efficacy in Alzheimer's Disease Models** The preclinical efficacy of T3D-959 has been extensively evaluated, primarily utilizing the intracerebral (i.c.) administration of streptozotocin (STZ) in rats as a model for sporadic AD. This model is favored because STZ induces brain insulin resistance and metabolic dysfunction, mimicking key aspects of human AD pathology.[1, 3, 4, 6, 21, 22, 25, 27]

In these i.c. STZ-treated rat models, T3D-959 demonstrated a broad range of beneficial effects: *   **Cognitive and Motor Improvements:** Significant improvements were observed in spatial learning and memory, as assessed by the Morris water maze test.[1, 3, 4, 27] Motor performance, evaluated using the rotarod test, was also enhanced.[3, 12, 21, 22] *   **Neuropathological Attenuation:** T3D-959 treatment led to improved brain morphology and increased viability in ex vivo frontal lobe slice cultures derived from STZ-treated rats.[1, 4, 27] It also reduced levels of oxidative stress and amyloid-beta (Aβ) accumulation.[1, 4, 21, 27] Furthermore, the drug normalized the expression of hyperphosphorylated tau (phospho-tau), choline acetyltransferase (ChAT, an enzyme critical for acetylcholine synthesis and thus cholinergic function), and myelin-associated glycoprotein (MAG, indicative of myelin integrity).[1, 4, 27] *   **Restoration of Metabolic Signaling:** A key finding was the restoration of insulin/Insulin-like Growth Factor (IGF) signaling pathways. T3D-959 increased or normalized the expression/activity of crucial signaling molecules such as IGF-1R, IRS-1, Akt, p70S6K, and S9-GSK-3β (an inactive form of GSK-3β, an enzyme implicated in tau hyperphosphorylation and insulin resistance).[3, 4] *   **Anti-inflammatory Effects:** The treatment decreased the expression of multiple pro-inflammatory cytokines in the brain, underscoring its neuroinflammatory modulating capacity.[3, 4, 21]

These protective effects were observed even at relatively low doses of T3D-959 (e.g., 0.3 mg/kg/day in rats) and, importantly, when the drug was administered therapeutically—that is, after the STZ-induced brain insult had occurred (e.g., treatment initiated 1 or 7 days post-STZ).[3, 4, 22, 25, 27] The ability to show efficacy in a therapeutic paradigm, rather than solely prophylactic, is particularly relevant for clinical translation to AD patients who typically present after disease onset. The consistency of these findings across cognitive, motor, neuropathological, and molecular biomarker endpoints provided a robust preclinical foundation for advancing T3D-959 into human clinical trials for AD.

**Table 2: Summary of Key Preclinical Findings for T3D-959 in AD Models**

Animal ModelT3D-959 Dose(s) (Route)Key Outcome MeasuresKey ResultsReference(s)
i.c. STZ Rats0.3-3.0 mg/kg/day (oral gavage)Spatial learning & memory (Morris water maze), Motor function (rotarod)Significantly improved spatial learning and memory; Improved motor performance.1
i.c. STZ Rats0.3-3.0 mg/kg/day (oral gavage)Brain morphology, Oxidative stress, Aβ levels, Phospho-tau, ChAT, MAGImproved brain morphology & culture viability; Reduced oxidative stress & Aβ; Normalized phospho-tau, ChAT, MAG.1
i.c. STZ Rats0.3-3.0 mg/kg/day (oral gavage)Insulin/IGF signaling (IGF-1R, IRS-1, Akt, p70S6K, S9-GSK-3β), Neuroinflammation (pro-inflammatory cytokines)Restored/normalized insulin/IGF signaling pathways; Decreased expression of multiple pro-inflammatory cytokines.3
i.c. STZ Rat (Ex vivo frontal lobe slice cultures)0.5-1.0 µMCulture viability, Molecular markers of ADSignificantly improved culture viability; Normalized AD markers.4

**3.2. Other Relevant Preclinical Findings** Consistent with its original developmental focus, T3D-959 demonstrated anti-diabetic activity in preclinical models.[3] An important observation from the rat studies was the lack of body weight increase following T3D-959 administration.[25] This is a noteworthy distinction from some traditional PPARγ agonists, such as certain thiazolidinediones, which are often associated with weight gain as a side effect. This favorable aspect of T3D-959's preclinical profile may be attributable to its biased agonism, particularly its stronger relative activity at the PPARδ receptor, and suggests a potentially improved safety and tolerability profile for chronic administration in human populations.

4. Clinical Development for Alzheimer's Disease

The clinical development of T3D-959 for Alzheimer's Disease has progressed through early-phase studies to a significant Phase 2 trial, with plans for further late-stage investigation.

**Table 3: Overview of Clinical Trials for DB-959Na (T3D-959) in Alzheimer's Disease**

Trial IDPhaseStudy Title/ObjectivePatient Population (Stage, N)Doses TestedDurationKey Endpoints (Primary, Secondary)Current StatusKey Findings SummaryReference(s)
N/A1Safety, tolerability, MTDNormal subjects (N=96)Up to >200mgUp to 7 daysSafety, PKCompletedMTD >200mg, no drug-related AEs.6
NCT025607531/2aFeasibility study: safety, tolerability, PK, PD, exploratory cognitionMild-to-moderate AD (N=34-36, MMSE 14-26)3, 10, 30, 90 mg/day14 days (+22-week OLE for some)Primary: Cerebral glucose metabolism (FDG-PET). Secondary/Exploratory: ADAS-Cog11, DSST, safety, PK/PD.CompletedSafe, well-tolerated. Dose-dependent metabolic changes. Preliminary cognitive improvements.3
NCT04251182 (PIONEER)2Safety, tolerability, efficacyMild-to-moderate AD (N=250 ITT; N=141 mITT, MMSE 14-26)Placebo, 15mg, 30mg, 45mg/day24 weeksPrimary: ADAS-Cog11, ADCS-CGIC. Secondary: Plasma Aβ42/40, DSST.CompletedITT failed primary endpoints. mITT: primary endpoints approached significance; significant biomarker improvements (Aβ42/40, neurogranin). Safe, well-tolerated, no ARIA.3
Planned2b/3Efficacy and safetyMild-to-moderate AD, biomarker-confirmed (p-tau217) (N~376)30mg/day vs Placebo78 weeksCognition, functionPlannedTo affirm PIONEER mITT findings.7

**4.1. Early Phase Clinical Studies (Phase 1 and Phase 1/2a NCT02560753)**

   **4.1.1. Design, Objectives, and Patient Population**    Initial Phase 1 studies involving 96 healthy volunteers established a maximum tolerated dose (MTD) greater than 200mg over 7 days of dosing, with no drug-related adverse events reported, indicating a favorable early safety profile.[6]

   Following this, an exploratory/feasibility Phase 1/2a study (NCT02560753) was conducted in 34 to 36 patients diagnosed with mild-to-moderate AD (Mini-Mental State Examination score 14-26).[3, 6, 20, 21, 22, 25, 26, 27, 28] This open-label study randomized participants to one of four oral doses of T3D-959 (3 mg, 10 mg, 30 mg, or 90 mg) administered once daily for 14 days. A few completers also participated in a 22-week open-label extension.[6] The primary objective was to assess changes in cerebral glucose metabolism using FDG-PET imaging. Secondary and exploratory objectives included cognitive assessments (Alzheimer's Disease Assessment Scale-Cognitive Subscale 11-item, Digit Symbol Substitution Test), safety, pharmacokinetics (PK), and pharmacodynamics (PD).[11]

   **4.1.2. Pharmacokinetic, Pharmacodynamic, and Preliminary Efficacy Results (NCT02560753)**    Pharmacokinetic analyses from this study indicated dose-dependent exposure to T3D-959.[4] Evidence also suggested drug penetration into the human brain and target engagement, crucial for its intended CNS effects.[6]

   Pharmacodynamic assessments revealed dose-dependent effects on systemic metabolism. Changes in the plasma metabolome profile, including decreases in branched-chain amino acids (isoleucine, leucine, valine) and an increased ratio of glycine levels, were consistent with improved insulin sensitivity.[3, 4] FDG-PET imaging demonstrated dose-dependent, regional effects of T3D-959 on regional cerebral metabolic rate for glucose (<span class="math-inline">R\_\{CMRgl\}</span>), indicating an impact on brain energy metabolism.[3, 4]

   Preliminary cognitive assessments showed rapid improvements. In 53% of subjects, T3D-959 treatment led to improvements in ADAS-Cog11 scores (average improvement of 4.14 points from baseline for those improving by ≥1 point), and 56% of subjects improved on the DSST. These improvements were, on average, sustained 7 days after dosing cessation. Improvements were observed in both mild and moderate AD subjects, with no apparent relationship to baseline disease severity.[21] However, subjects in the highest dose cohort (90mg) showed an average improvement on DSST but not on ADAS-Cog11.[21] This lack of a clear linear dose-response on ADAS-Cog11 at the highest dose suggested a complex dose-efficacy relationship or limitations due to the small sample size, warranting further investigation in larger, controlled trials.

   **4.1.3. Safety and Tolerability**    The Phase 1 studies in healthy volunteers established an MTD greater than 200mg with no drug-related adverse events.[6] In the Phase 1/2a study (NCT02560753), T3D-959 was generally safe and well-tolerated across all doses tested. No significant safety findings were reported, and the drug did not have negative effects on cognitive function.[3, 4, 6, 21, 28] General literature mentions possible low-frequency side effects such as muscle weakness and cramps for T3D-959.[3] Overall, these early phase studies, despite the open-label design and small sample size of the Phase 1/2a trial, provided encouraging preliminary evidence of target engagement (metabolic and FDG-PET changes) and potential cognitive benefits, alongside a good safety profile, thereby supporting advancement to the Phase 2 PIONEER study.

**4.2. Phase 2 PIONEER Study (NCT04251182)**

   **4.2.1. Study Design, Endpoints, and Baseline Characteristics**    The PIONEER study (NCT04251182) was a Phase 2, multi-center, randomized, double-blind, placebo-controlled trial designed to evaluate the efficacy and safety of T3D-959 in patients with mild-to-moderate AD.[3, 6, 7, 8, 9, 10, 16, 17, 18, 19] The study enrolled 250 participants (Intent-to-Treat, ITT population) with mild-to-moderate AD as defined by NIA-AA criteria and MMSE scores between 14-26.[9, 10, 16, 17] Notably, AD biomarker confirmation was not an initial enrollment criterion for the ITT population.[10, 16, 17] Participants were randomized to receive one of three doses of T3D-959 (15mg, 30mg, or 45mg) or placebo, administered orally once daily for 24 weeks.[3, 8, 9, 10, 16, 17, 18, 19]

   The co-primary endpoints were the change from baseline to week 24 in the Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog11) and the Alzheimer's Disease Cooperative Study-Clinical Global Impression of Change (ADCS-CGIC).[3, 9, 10, 16, 17] Secondary endpoints included changes in plasma Aβ42/40 ratio and the Digit Symbol Coding Test (DSST).[10, 16, 17] A range of exploratory endpoints, including plasma biomarkers such as p-tau217, neurogranin, neurofilament light chain (NfL), and various metabolomic and proteomic markers, were also assessed.[9, 10, 16, 17, 18, 19]

   **4.2.2. Reported Data Irregularities and Establishment of mITT Population**    The initial analysis of the full ITT population (N=250) for the PIONEER study did not meet its primary endpoints.[3, 10] However, post-unblinding, significant data irregularities were identified at a cluster of five study sites.[10, 16, 17] These irregularities were threefold:    1.  **Pharmacokinetic Anomalies:** At these "excluded sites," 55% of subjects assigned to receive T3D-959 had no detectable drug in their plasma at the end of treatment, as confirmed by PK analysis and other sensitive biomarkers. This strongly suggests non-compliance or issues with drug administration/absorption for a majority of these participants.[10]    2.  **Scientifically Improbable AD Diagnosis:** Over 75% of subjects at these excluded sites did not meet the threshold for AD pathology based on baseline plasma p-tau217/non-p-tau217 ratio (a validated biomarker for AD pathology), whereas over 75% of subjects in the subsequently defined mITT population did meet this threshold.[10, 16, 17] This indicates that many participants from these sites may not have had the underlying AD pathology the drug aims to treat.    3.  **Atypical Placebo Response:** The placebo response observed at these sites was reported to be outside of historic norms for mild-to-moderate AD populations.[16, 17]

   Due to these substantial data integrity concerns, which confounded the ability to accurately evaluate therapeutic effect, data from all subjects enrolled at these five sites were removed from the dataset. This led to the formation of a Modified Intent-to-Treat (mITT) population comprising 141 subjects.[9, 10, 16, 17] This mITT population was independently corroborated using blinded study data by Pentara Corp. and, following formal discussion with the FDA, will be reported in the Clinical Study Report (CSR) as a post-hoc efficacy analysis.[10] The use of the pTau-217 ratio post-hoc to identify subjects with improbable AD diagnosis also underscores the evolving importance of biological confirmation of AD in clinical trials, even if not initially part of inclusion criteria.

   **4.2.3. Efficacy Outcomes in the mITT Population (Cognitive, Functional, and Biomarker Endpoints)**    In the mITT population (N=141; Placebo n=32; T3D-959 15mg n=38, 30mg n=39, 45mg n=32), the co-primary endpoints showed trends towards benefit:    *   **ADAS-Cog11:** At week 24, the 30mg T3D-959 group showed a mean worsening of 0.73 points from baseline, compared to a 2.70-point worsening in the placebo group (p=0.073). This difference suggests a slowing of clinical decline.[9, 10, 16, 17] One report highlighted this as a 1.97-point benefit, corresponding to a 73% slowing in cognitive decline for the 30mg dose.[10]    *   **ADCS-CGIC:** The 15mg T3D-959 group showed a mean worsening of 0.39 points, compared to 0.86 points for placebo (p=0.060).[9, 10, 16, 17]

   While these results for the primary endpoints did not achieve conventional statistical significance (p<0.05), they approached it, and the observed effect sizes, particularly for ADAS-Cog11 in the 30mg group, are considered indicative of potential clinical benefit, especially given the relatively small sample sizes in the mITT analysis per dose group.

   **Table 4: Detailed Efficacy Results from the PIONEER (NCT04251182) mITT Analysis (Selected Endpoints)**

EndpointTreatment GroupChange from Baseline at Week 24 (LS Mean)Placebo-Adjusted Difference (LS Mean)P-valueReference(s)
ADAS-Cog11 (Worsening is positive)Placebo (n=32)2.70N/AN/A9
T3D-959 15mg (n=38)N/A (focus on 30mg for ADAS-Cog11 primary)N/AN/A
T3D-959 30mg (n=39)0.73-1.970.0739
T3D-959 45mg (n=32)N/AN/AN/A
ADCS-CGIC (Worsening is positive)Placebo (n=32)0.86N/AN/A9
T3D-959 15mg (n=38)0.39-0.470.0609
Plasma Aβ42/40 Ratio (Improvement is positive)PlaceboN/AN/AN/A3
T3D-959 30mgSignificant ImprovementSignificant0.0113
T3D-959 45mgSignificant ImprovementSignificant0.0333
Plasma Neurogranin (Improvement is reduction)PlaceboN/AN/AN/A9
T3D-959 30mgSignificant ImprovementSignificant0.0359
T3D-959 45mgSignificant ImprovementSignificant0.0519

    *(Note: N/A indicates data not explicitly provided in the snippets for that specific cell in a comparable format or for that specific dose group's primary endpoint contribution. "Significant Improvement" refers to statistically significant changes vs. placebo as reported.)*

   **4.2.4. Analysis of Key Biomarkers (Aβ42/40, Neurogranin, pTau-217)**    Biomarker analysis from the PIONEER mITT population yielded several important findings:    *   **Plasma Aβ42/40 Ratio:** This secondary endpoint, a marker of amyloid plaque burden, showed statistically significant improvement in both the 30mg (p=0.011) and 45mg (p=0.033) T3D-959 groups compared to placebo. The magnitude of this effect was reported to be similar to that observed with the anti-amyloid antibody lecanemab at 6 months of treatment. Furthermore, in the subgroup of patients with a high baseline pTau-217 ratio (indicative of AD pathology), the improvement in the Aβ42/40 ratio was nearly twofold greater than in the overall mITT group, suggesting a more pronounced effect on amyloid pathology in this biomarker-defined population.[3, 9, 10, 16, 17, 18, 19]    *   **Plasma Neurogranin:** Levels of neurogranin, a postsynaptic protein and marker of synaptic degeneration, were significantly improved (reduced) in the 30mg (p=0.035) and 45mg (p=0.051) T3D-959 groups.[9, 10, 16, 17, 18, 19]    *   **Plasma pTau-217/non-pTau-217 Ratio:** This ratio was utilized to define a subgroup with high AD pathology, comprising 55% of the mITT population. In this subgroup, placebo-treated patients exhibited cognitive decline consistent with AD. Treatment with T3D-959, particularly the 30mg dose, showed evidence of slowing cognitive decline (an improvement of 2 points vs. placebo on ADAS-Cog11) and a trend towards slowing disease progression as measured by this tau biomarker (trend at 15mg).[9, 10, 18, 19]    *   **Other Proteomic and Metabolomic Markers:** T3D-959 treatment led to changes in multiple plasma proteomic and metabolomic biomarkers, suggesting pluripotent effects on dysregulated AD pathways, including inflammation, oxidative stress, metabolism, insulin resistance, and mitochondrial dysfunction.[9, 10, 16, 17, 18, 19] These findings support the drug's proposed multi-faceted mechanism of action. The statistically significant effects on established AD fluid biomarkers like Aβ42/40 and neurogranin provide objective evidence of biological activity in the CNS, lending credence to the potential for disease modification. The identification of a "responder" subgroup based on baseline pTau-217 levels is particularly important, as it suggests that a precision medicine approach, targeting patients with confirmed AD pathology and perhaps a specific metabolic profile, could optimize the chances of demonstrating efficacy in future trials.

   **4.2.5. Safety and Tolerability Profile (including ARIA assessment)**    T3D-959 was reported to be generally safe and well-tolerated in the PIONEER mITT population across the 24-week treatment period.[9, 10, 16, 17, 18, 19] Crucially, there were no drug-related adverse events related to stroke and no clinical signs of Amyloid-Related Imaging Abnormalities (ARIA-E for edema or ARIA-H for microhemorrhages/hemosiderin deposits).[10, 19] This lack of ARIA is a highly significant differentiating safety feature compared to many amyloid-targeting monoclonal antibody therapies for AD, which often carry a risk of ARIA that necessitates careful patient monitoring and can limit treatment eligibility or continuation. The favorable safety profile of an oral, once-daily medication without the ARIA risk associated with infused antibodies would represent a substantial clinical advantage if efficacy is confirmed.

**4.3. Future Clinical Development (Phase 2b/3)** Based on the outcomes of the PIONEER Phase 2 study, particularly the mITT analysis and biomarker findings, T3D Therapeutics plans to advance T3D-959 into a larger and longer Phase 2b/3 clinical trial.[7, 9, 10, 19] The 30mg once-daily dose has been identified as providing optimal safety and efficacy for this further investigation.[7, 9, 10, 19]

The planned Phase 2b/3 study will target patients with mild-to-moderate AD. A key refinement in the trial design will be the inclusion of a biological diagnosis of AD pathology, using a validated plasma biomarker (specifically, the %p-tau217 plasma biomarker), as an enrollment criterion.[7] This addresses one of the confounding factors in the PIONEER study by ensuring that enrolled participants have the target pathology. The study is designed as a seamless group sequential trial, with an estimated enrollment of 376 subjects who will be randomized 1:1 to receive either T3D-959 30mg or placebo once daily for 78 weeks. Stratification will be by sex and ApoE4 genotype. An interim analysis will re-estimate the sample size to ensure sufficient power to demonstrate efficacy on cognitive and functional scales. Safety, tolerability, efficacy, and exploratory assessments will be evaluated as changes from baseline to the end of the 78-week treatment period, followed by a four-week follow-up.[7] This adaptive design and biomarker-driven patient selection strategy reflect a considered approach to de-risk late-stage development and increase the probability of demonstrating a clear therapeutic benefit.

5. Other Potential Indications

Beyond Alzheimer's Disease, the mechanism of action of T3D-959, particularly its potent PPARδ agonism and ability to modulate metabolic pathways, suggests therapeutic potential in other neurodegenerative disorders where metabolic dysfunction and neuroinflammation are implicated.

T3D Therapeutics has indicated an interest in exploring T3D-959 for Huntington's Disease (HD).[4] Extensive research in HD has highlighted interference with PPARδ function as a contributor to its pathogenesis. Preclinical studies by other groups have shown that PPARδ agonists can ameliorate disease phenotypes and extend lifespan in HD mouse models, and can prevent cell death of medium spiny-like neurons derived from HD patient-induced pluripotent stem cells (iPSCs).[6] Given that PPARδ is highly expressed in the CNS and T3D-959 is brain-penetrant, this represents a rational area for pipeline expansion.

More broadly, the company suggests that T3D-959 may offer therapeutic benefits in other CNS or neurodegenerative disorders characterized by dysfunctional brain glucose and lipid metabolism.[5] The drug's ability to improve metabolic health at a cellular level, reduce oxidative stress, and dampen inflammation are all properties that could be advantageous across a spectrum of neurological conditions.

6. Intellectual Property

The intellectual property surrounding T3D-959 is rooted in its initial development and subsequent licensing. T3D Therapeutics, Inc. holds the exclusive worldwide rights to develop and commercialize T3D-959 (formerly DB959) and related compounds. These rights were obtained through a licensing agreement with DARA BioSciences, Inc. (now Midatech Pharma US) in June 2013.3 DARA BioSciences had previously developed DB959 through Phase 1 clinical trials after licensing it from the original originator, Bayer HealthCare Pharmaceuticals.

The core composition of matter patents for T3D-959 likely originated from Bayer and/or DARA BioSciences. For instance, WIPO patent WO2007038528A2, associated with T3D-959 in some databases, appears to be from Bayer Schering Pharma AG and pertains to oxazole derivatives and their use as PPAR agonists for metabolic disorders [[4] (mentioning the patent without full title/assignee), [29] (general patent from Bayer for similar compounds)]. PubChem also lists patents available for the chemical structure of DB-959 sodium.[2]

Since acquiring the rights, T3D Therapeutics would be expected to build its own patent portfolio around T3D-959, focusing on new uses (e.g., for Alzheimer's Disease, Huntington's Disease), specific formulations, methods of treatment, and potentially biomarkers for patient selection. Dr. Roberta Brinton, a member of T3D Therapeutics' Scientific Advisory Board, holds multiple patents targeting Alzheimer's disease and age-associated neurodegenerative diseases, which may complement or inform T3D's IP strategy.[15] The strategic development of a robust patent estate is critical for protecting the commercial potential of T3D-959 as it advances through clinical trials.

7. Discussion and Future Outlook

DB-959Na (T3D-959) represents a novel therapeutic approach for Alzheimer's Disease, distinguished by its mechanism as a dual PPARδ/γ agonist targeting underlying metabolic dysfunctions rather than directly addressing amyloid or tau pathology. Preclinical studies in AD models were robust, demonstrating improvements in cognition, motor function, and a range of neuropathological and biomarker endpoints. Early human studies (Phase 1 and Phase 1/2a) suggested good safety, brain penetration, target engagement (evidenced by metabolic changes), and preliminary cognitive benefits.

The Phase 2 PIONEER trial (NCT04251182) encountered significant operational challenges, leading to data irregularities at several sites that confounded the primary ITT analysis. However, a post-hoc mITT analysis, excluding data from these compromised sites, revealed trends toward efficacy on co-primary cognitive and functional endpoints, particularly at the 30mg dose. More compellingly, the mITT analysis showed statistically significant improvements in important AD biomarkers, including plasma Aβ42/40 ratio and neurogranin, and favorable changes in markers of inflammation and metabolism. The identification of a patient subgroup with high baseline pTau-217 (indicative of AD pathology) that showed more pronounced cognitive benefits with T3D-959 is a critical finding that informs a precision medicine strategy for future development.

A key strength of T3D-959 is its consistently favorable safety and tolerability profile, most notably the absence of ARIA, which is a common and serious concern with amyloid-targeting antibody therapies. As an oral, once-daily medication, T3D-959 also offers a significant convenience advantage over infused biologics.

The path forward for T3D-959 hinges on the successful execution and positive outcome of the planned Phase 2b/3 trial. This trial will incorporate crucial learnings from PIONEER, including the selection of the 30mg dose and the use of plasma pTau-217 for patient selection to ensure enrollment of individuals with confirmed AD pathology. Challenges remain, including the need to robustly demonstrate clinical efficacy in a larger, longer trial, further validate the biomarker-defined responder population, and navigate an increasingly competitive AD therapeutic landscape. However, if its unique metabolic mechanism translates into clear clinical benefits with a superior safety profile, T3D-959 could emerge as a valuable and differentiated disease-modifying therapy for AD, potentially for specific patient subpopulations or in combination with other agents. Its potential utility in other neurodegenerative disorders like Huntington's Disease also warrants further exploration.

8. Conclusion

DB-959Na (T3D-959) is an investigational oral dual PPARδ/γ agonist with a primary focus on treating Alzheimer's Disease by addressing underlying metabolic dysfunctions, including brain insulin resistance, impaired glucose and lipid metabolism, and neuroinflammation. Its pharmacological profile, characterized by a 15-fold higher potency for PPARδ and BBB penetration, supports its potential for CNS therapeutic effects.

Preclinical studies in AD models demonstrated broad neuroprotective effects, improving cognition, motor function, and multiple neuropathological markers. Early clinical trials (Phase 1 and 1/2a) indicated good safety, target engagement, and preliminary cognitive benefits. The Phase 2 PIONEER study (NCT04251182) yielded complex results due to significant data irregularities at several sites. While the ITT analysis did not meet primary endpoints, a pre-specified mITT analysis showed trends towards efficacy on cognitive/functional measures and statistically significant improvements in key AD biomarkers (plasma Aβ42/40, neurogranin) and markers of metabolic health and inflammation. A subgroup of patients with high baseline pTau-217 appeared to derive greater cognitive benefit.

A critical differentiating factor for T3D-959 is its favorable safety profile, notably the absence of ARIA, which positions it advantageously against amyloid-targeting antibodies. The upcoming Phase 2b/3 trial, employing the 30mg dose and biomarker-based patient selection, will be pivotal in determining its efficacy.

While the PIONEER trial's complexities require careful consideration, T3D-959's unique mechanism, oral administration, and promising safety and biomarker data suggest it holds potential as a novel disease-modifying therapy for AD, particularly for biomarker-defined patient populations. Its development underscores the importance of exploring diverse therapeutic targets beyond amyloid and tau in the multifaceted pathology of Alzheimer's Disease. Successful late-stage development could offer a much-needed alternative or complementary treatment strategy for this devastating neurodegenerative condition.

Works cited

  1. DB-959 - Inxight Drugs - ncats, accessed June 11, 2025, https://drugs.ncats.io/substance/4TW7R18VOJ
  2. DB-959 sodium | C25H26NNaO5 | CID 49839871 - PubChem, accessed June 11, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/DB-959-sodium
  3. T3D-959 - Alzheimer's Drug Discovery Foundation, accessed June 11, 2025, https://www.alzdiscovery.org/uploads/cognitive_vitality_media/T3D-959.pdf
  4. DB-959Na - Drug Targets, Indications, Patents - Patsnap Synapse, accessed June 11, 2025, https://synapse.patsnap.com/drug/e2a0a9d893594c9daab675cf877f4512
  5. About Us - T3D Therapeutics, accessed June 11, 2025, https://www.t3dtherapeutics.com/about-us/
  6. Lead Product Candidate T3D-959 - T3D Therapeutics, accessed June 11, 2025, https://www.t3dtherapeutics.com/lead-product-candidate-t3d-959/
  7. A Phase 2b/3 Clinical Study Evaluating T3D-959 in Mild-to-Moderate Alzheimer's Disease Subjects - ClinicalTrials.Veeva, accessed June 11, 2025, https://ctv.veeva.com/study/a-phase-2b-3-clinical-study-evaluating-t3d-959-in-mild-to-moderate-alzheimers-disease-subjects
  8. A Novel Way to Reduce Alzheimer's Amyloid Plaque Burden, Phase 2 Clinical Trial Results of T3D-959 to be Presented by T3D Therapeutics at AAIC - BioSpace, accessed June 11, 2025, https://www.biospace.com/a-novel-way-to-reduce-alzheimers-amyloid-plaque-burden-phase-2-clinical-trial-results-of-t3d-959-to-be-presented-by-t3d-therapeutics-at-aaic
  9. Alzheimer Agent T3D-959 Demonstrates Disease-Modifying Effects in Phase 2 PIONEER Trial - NeurologyLive, accessed June 11, 2025, https://www.neurologylive.com/view/alzheimer-agent-t3d-959-demonstrates-disease-modifying-effects-phase-2-pioneer-trial
  10. Phase 2 PIONEER Trial of Oral T3D-959 for the Treatment of Patients Diagnosed with Mild-to-Moderate Alzheimer's Disease, accessed June 11, 2025, https://www.t3dtherapeutics.com/wp-content/uploads/2024/07/AAIC24_Poster_Final-1.pdf
  11. Abstracts of 20th Annual ASENT Meeting - PMC - PubMed Central, accessed June 11, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6095775/
  12. T3D Therapeutics Signs Exclusive License Agreement for T3D-959 (DB959) and Related Compounds, accessed June 11, 2025, https://www.t3dtherapeutics.com/2013/06/18/t3d-therapeutics-signs-exclusive-license-agreement-for-t3d-959-db959-and-related-compounds/
  13. T3D Therapeutics | T3D Therapeutics, accessed June 11, 2025, https://www.t3dtherapeutics.com/
  14. Mission | T3D Therapeutics, accessed June 11, 2025, https://www.t3dtherapeutics.com/about-us/mission/
  15. Scientific Advisory Board - T3D Therapeutics, accessed June 11, 2025, https://www.t3dtherapeutics.com/about-us/advisory-board/
  16. Phase 2 PIONEER Trial of Oral T3D‐959 for the Treatment of Patients Diagnosed with Mild‐to‐Moderate Alzheimer's Disease: New Results in a Modified Intent‐to‐Treat Population - PMC, accessed June 11, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11713541/
  17. (PDF) Phase 2 PIONEER Trial of Oral T3D‐959 for the Treatment of Patients Diagnosed with Mild‐to‐Moderate Alzheimer's Disease: New Results in a Modified Intent‐to‐Treat Population - ResearchGate, accessed June 11, 2025, https://www.researchgate.net/publication/387856000_Phase_2_PIONEER_Trial_of_Oral_T3D-959_for_the_Treatment_of_Patients_Diagnosed_with_Mild-to-Moderate_Alzheimer's_Disease_New_Results_in_a_Modified_Intent-to-Treat_Population
  18. T3D Therapeutics presents top-line results from its phase 2 PIONEER trial, accessed June 11, 2025, https://www.alzheimer-europe.org/news/t3d-therapeutics-presents-top-line-results-its-phase-2-pioneer-trial?language_content_entity=en
  19. T3D Therapeutics Announces Positive Top-Line Results from the Phase 2 PIONEER Trial of T3D-959 in Mild-To-Moderate Alzheimer's Disease - PR Newswire, accessed June 11, 2025, https://www.prnewswire.com/news-releases/t3d-therapeutics-announces-positive-top-line-results-from-the-phase-2-pioneer-trial-of-t3d-959-in-mild-to-moderate-alzheimers-disease-301976115.html
  20. Targeting Alzheimer's Disease Neuro-Metabolic Dysfunction with a Small Molecule Nuclear Receptor Agonist (T3D-959) Reverses Dise ase Pathologies | OMICS International, accessed June 11, 2025, https://www.omicsonline.org/open-access/targeting-alzheimers-disease-neurometabolic-dysfunction-with-asmall-molecule-nuclear-receptor-agonist-t3d959-reverses-dise-asepath-2161-0460-1000238.php?aid=75764
  21. T3D Therapeutics Announces Presentation of Phase 2a Preliminary Results of T3D-959 in Alzheimer's Subjects at the 2016 Alzheimer's Association International Conference - PRWeb, accessed June 11, 2025, https://www.prweb.com/releases/t3d_therapeutics_announces_presentation_of_phase_2a_preliminary_results_of_t3d_959_in_alzheimer_s_subjects_at_the_2016_alzheimer_s_association_international_conference/prweb13590398.htm
  22. Targeting Alzheimer's Disease Neuro-Metabolic Dysfunction with a Small Molecule Nuclear Receptor Agonist (T3D-959) Reverses Dise ase Pathologies - ResearchGate, accessed June 11, 2025, https://www.researchgate.net/publication/305036272_Targeting_Alzheimer's_Disease_Neuro-Metabolic_Dysfunction_with_a_Small_Molecule_Nuclear_Receptor_Agonist_T3D-959_Reverses_Dise_ase_Pathologies
  23. (PDF) PPARs in Alzheimer's disease - ResearchGate, accessed June 11, 2025, https://www.researchgate.net/publication/51419498_PPARs_in_Alzheimer's_disease
  24. Central role of PPARγ in Alzheimer's disease: From pathophysiology to potential therapies, accessed June 11, 2025, https://accscience.com/journal/AN/articles/online_first/4652
  25. T3D-959: A Multi-Faceted Disease Remedial Drug Candidate for the Treatment of Alzheimer's Disease - PMC - PubMed Central, accessed June 11, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5577391/
  26. Targeting Alzheimer's Disease Neuro-Metabolic Dysfunction with a Small Molecule Nuclear Receptor Agonist (T3D-959) Reverses Disease Pathologies - PubMed, accessed June 11, 2025, https://pubmed.ncbi.nlm.nih.gov/27525190/
  27. T3D-959: A Multi-Faceted Disease Remedial Drug Candidate for the Treatment of Alzheimer's Disease - PubMed, accessed June 11, 2025, https://pubmed.ncbi.nlm.nih.gov/26836193/
  28. Novel Therapy for Alzheimer's Disease Receives NIA Support - - PracticalNeurology, accessed June 11, 2025, https://practicalneurology.com/news/novel-therapy-for-alzheimers-disease-receives-nia-support/2468907/
  29. IL190217A0 - Combination of rosiglitazone and donepezil for improvement of cognitive function - Google Patents, accessed June 11, 2025, https://patents.google.com/patent/IL190217A0/es

Published at: June 11, 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.