BI-1358894 is an orally administered, first-in-class, small-molecule inhibitor of the Transient Receptor Potential Canonical 4 and 5 (TRPC4/5) ion channels. As a new molecular entity originated by Hydra Biosciences and advanced into clinical development by Boehringer Ingelheim, it represents a novel therapeutic approach for psychiatric disorders.[1] The drug’s therapeutic hypothesis is predicated on its ability to attenuate hyperreactivity of the amygdala, a central node in the brain's emotional processing circuits, which is implicated in the pathophysiology of depression, anxiety, and trauma-related conditions.[2]
The clinical development program for BI-1358894 has yielded a complex and dichotomous set of results. Phase I studies in healthy volunteers were highly successful, establishing a favorable safety and pharmacokinetic profile suitable for once-daily dosing and, critically, providing clear evidence of target engagement. Pharmacodynamic and neuroimaging studies demonstrated that BI-1358894 could blunt pharmacologically induced panic-like symptoms and reduce activation in key emotional brain regions, including the amygdala.[2]
This early promise, however, was not translated into clinical efficacy in subsequent Phase II trials. Large, well-controlled studies in patients with Major Depressive Disorder (MDD) and Borderline Personality Disorder (BPD) failed to demonstrate a statistically significant separation from placebo on their respective primary and secondary endpoints. Consequently, proof-of-concept (PoC) was not established in either indication.[2]
Despite the lack of efficacy, a consistent and highly favorable safety and tolerability profile has emerged as the drug's most significant asset. Across all clinical trials, including those in high-risk patient populations, BI-1358894 was well tolerated. Most notably, there was no observed increase in self-harm or suicidality compared to placebo, a finding of critical importance for any centrally-acting therapeutic.[2]
The future of the BI-1358894 program now hinges on the results of a completed Phase II trial in Post-Traumatic Stress Disorder (PTSD).[1] The disconnect between the drug's confirmed biological activity and its lack of clinical efficacy in broad psychiatric populations suggests a potential mismatch between its targeted mechanism and the heterogeneous nature of MDD and BPD. The PTSD indication, with its more defined pathophysiology centered on fear circuitry and amygdala dysfunction, represents a more mechanistically aligned and decisive test of the drug's core hypothesis. The viability of BI-1358894 as a therapeutic agent is therefore almost entirely contingent on the outcome of this pivotal study.
The development of BI-1358894 marks a deliberate move within psychopharmacology from broad, neurotransmitter-based strategies, such as those employed by selective serotonin reuptake inhibitors (SSRIs), towards more precise, circuit-based interventions. Standard-of-care antidepressants modulate entire neurotransmitter systems, often leading to systemic side effects and limited efficacy for a substantial portion of patients.[8] BI-1358894's mechanism, by contrast, is not predicated on modulating serotonin or norepinephrine but on inhibiting specific ion channels highly expressed in brain regions critical for fear and emotion. This targeted approach aims to correct a specific pathophysiological process—amygdala hyperreactivity—with the potential for improved efficacy in specific symptom domains and a more favorable tolerability profile, addressing a significant unmet need in modern psychiatry.[2]
BI-1358894 is an investigational small molecule classified as an antidepressant, anxiolytic, and neuroprotectant.[1] It is being developed by Boehringer Ingelheim, following its origination by Hydra Biosciences.[1] As a New Molecular Entity, it represents a novel chemical structure not previously approved for therapeutic use. The drug has not been granted Orphan Drug status, indicating its intended applications are for conditions with a broader prevalence.[1]
The primary mechanism of action of BI-1358894 is the inhibition of TRPC4 and TRPC5 cation channels.[1] These channels are members of the transient receptor potential canonical family of ion channels and have been implicated in the pathophysiology of mood and anxiety disorders.[4] Physiologically, TRPC4 and TRPC5 channels are typically closed but open in response to certain stimuli, allowing an influx of cations, including calcium (
Ca2+), into the neuron. This influx is a critical step in neuronal signal transmission.[4] By inhibiting these channels, BI-1358894 is designed to prevent or reduce this cation influx, thereby dampening neuronal activity and attenuating the physiological responses associated with fear, anxiety, and stress.[4]
The core therapeutic rationale for BI-1358894 is built upon the "amygdala hyperreactivity hypothesis".[2] This hypothesis posits that excessive neuronal activity in the amygdala and its associated cortico-limbic circuits is a key neurobiological substrate for the symptoms of several psychiatric disorders, including MDD, BPD, and PTSD.[4] The amygdala is a critical brain region for processing fear and other salient emotional stimuli. Hyperactivity in this region is thought to contribute to the negative emotional biases, heightened anxiety, and emotional dysregulation characteristic of these conditions. BI-1358894 is theorized to exert its therapeutic effects by directly attenuating this amygdala hyperreactivity, thereby normalizing cortico-limbic signaling and improving behaviors associated with dysfunctional emotional processing.[2] This novel, non-serotonergic mechanism positions BI-1358894 as a potential alternative or adjunctive treatment for patients who do not respond adequately to existing therapies.[2]
The Phase I clinical program for BI-1358894 was exceptionally robust and successful in establishing proof-of-mechanism. It went beyond standard safety and pharmacokinetic assessments to provide clear evidence that the drug was engaging its intended target and producing a measurable biological effect on the specific brain circuits and physiological systems it was designed to modulate. This comprehensive early-stage validation provided a strong scientific rationale to proceed to Phase II and makes the subsequent efficacy failures in later-stage trials all the more significant and perplexing.
Multiple Phase I studies, including single ascending dose (SAD) and multiple ascending dose (MAD) trials in healthy male volunteers of both Caucasian and Japanese ethnicity, established a favorable profile for BI-1358894.[5]
The drug was well tolerated in single oral doses up to 200 mg and in multiple-dose regimens.[2] The pharmacokinetic profile was found to be favorable, with exposure increasing in a dose-dependent and proportional manner.[5] The terminal elimination half-life was observed to be long, ranging from approximately 70 hours to over 200 hours at higher doses, supporting a once-daily dosing schedule.[14] A positive food effect was noted, with administration after a high-fat meal resulting in significantly higher drug exposure compared to the fasted state.[11] A drug-drug interaction study with the potent CYP3A4 inhibitor itraconazole showed a less than two-fold increase in BI-1358894 exposure, suggesting that co-administration with CYP3A4 inhibitors would not be a significant contraindication for Phase II studies.[15]
To establish early proof-of-principle and pharmacodynamic effect, a Phase I trial (NCT03904576) was conducted using the cholecystokinin-tetrapeptide (CCK-4) challenge model.[2] CCK-4 is a neuropeptide known to induce anxiety and panic-like symptoms by stimulating amygdala neuronal activity.[2] In this randomized, placebo-controlled, crossover study, healthy male volunteers received a single 100 mg oral dose of BI-1358894 or placebo prior to an intravenous CCK-4 challenge.[6]
The results provided clear evidence of the drug's anxiolytic and stress-dampening effects. Compared to placebo, BI-1358894 significantly reduced the psychological and physiological responses to the CCK-4 challenge. The primary endpoint, the maximum change from baseline in the Panic Symptom Scale (PSS) sum intensity score, was 24.4% lower after treatment with BI-1358894.[6] This subjective finding was corroborated by objective biomarker data; BI-1358894 reduced CCK-4-induced mean maximum plasma adrenocorticotropic hormone (ACTH) and serum cortisol values by 58.6% and 27.3%, respectively.[6]
Further confirmation of the drug's mechanism of action came from a functional magnetic resonance imaging (fMRI) study that directly visualized its effects on brain activity.[4] This study demonstrated that BI-1358894 reduced activation in several cortico-limbic brain regions involved in emotional processing, most notably the bilateral amygdala, when patients with MDD were presented with negative emotional stimuli (faces and scenes).[4] This finding provided direct evidence of target engagement within the intended neurocircuitry, confirming that the drug was actively modulating the biological substrate it was designed to affect.
The Phase II development program for BI-1358894 was ambitious, simultaneously investigating the drug's efficacy across three distinct psychiatric disorders: Major Depressive Disorder (MDD), Borderline Personality Disorder (BPD), and Post-Traumatic Stress Disorder (PTSD). The results from the MDD and BPD trials were negative, failing to establish proof-of-concept. This consistent failure across two broad indications suggests a potential mismatch between the drug's highly specific mechanism and the heterogeneous nature of these conditions. Consequently, the clinical and commercial viability of BI-1358894 has been refocused onto the PTSD indication, which represents a more direct and mechanistically plausible test of the drug's core therapeutic hypothesis.
Table 1: Summary of Phase II Clinical Trials for BI-1358894
| Parameter | Major Depressive Disorder (NCT04521478) | Borderline Personality Disorder (NCT04566601) | Post-Traumatic Stress Disorder (NCT05103657) |
|---|---|---|---|
| Indication | MDD with inadequate response to antidepressants | Borderline Personality Disorder (BPD) | Post-Traumatic Stress Disorder (PTSD) |
| Phase | Phase 2 | Phase 2 | Phase 2 |
| Design | Randomized, placebo-controlled, parallel group, dose-ranging (5, 25, 75, 125 mg), with quetiapine reference arm | Randomized, placebo-controlled, parallel group, dose-ranging (5, 25, 75, 125 mg) | Randomized, placebo-controlled, parallel group |
| N (Randomized) | 389 | 390 | 318 |
| Treatment Duration | 6 weeks | 12 weeks | 8 weeks |
| Primary Endpoint | Change from baseline in Montgomery-Åsberg Depression Rating Scale (MADRS) total score at Week 6 | Change from baseline in Zanarini Rating Scale for BPD (ZAN-BPD) total score at Week 10 | Change from baseline in Clinician-Administered PTSD Scale for DSM-5 (CAPS-5) total score at Week 8 |
| Status | Completed (Feb 2024) | Completed (Jan 2023) | Completed (Nov 2023) |
| Outcome | Negative Trial: Proof-of-Concept Not Established | Negative Trial: Proof-of-Concept Not Established | Results Not Yet Publicly Reported |
| Source(s) | 1 | 1 | 1 |
The investigation into MDD involved two separate Phase II trials, neither of which succeeded in establishing efficacy.
This was the primary efficacy-seeking study for BI-1358894 in MDD. It was a 6-week, multicenter, randomized, double-blind, placebo-controlled trial designed to assess a range of oral doses (5 mg, 25 mg, 75 mg, and 125 mg).[2] The study enrolled 389 patients diagnosed with MDD who had an inadequate response to their ongoing antidepressant monotherapy (SSRI, SNRI, or bupropion). A quetiapine extended-release arm was included as a reference comparator, a common adjunctive agent for this patient population.[2]
The trial was definitively negative. No statistically significant differences were observed between any of the BI-1358894 dose groups and the placebo group for the primary endpoint, which was the change from baseline in the Montgomery-Åsberg Depression Rating Scale (MADRS) total score at Week 6.[2] Furthermore, no significant differences were found for any of the secondary endpoints. As a result, the trial failed to meet its objectives, and proof-of-concept for BI-1358894 in this MDD population was not established.[2]
In parallel, Boehringer Ingelheim initiated a novel, fully remote decentralized clinical trial (DCT) to evaluate the efficacy and safety of a single 125 mg dose of BI-1358894 against placebo over 6 weeks in a similar patient population.[12] This study aimed to leverage digital technologies to increase accessibility for patients and investigators.[12]
However, the trial was terminated early due to deficient enrollment, which was attributed to stringent eligibility criteria.[12] Only 45 patients were randomized, and 43 received treatment. Due to this small sample size, a meaningful evaluation of efficacy was not possible.[18] Despite the failure to meet enrollment targets, the study provided valuable insights into the operational feasibility of conducting a fully remote, interventional psychiatric trial. The procedures were successfully executed, and the majority of participants (88%) reported a positive experience with the decentralized format.[12]
The Phase II trial in BPD (NCT04566601) mirrored the design of the MDD study. It was a 12-week, multinational, randomized, double-blind, placebo-controlled, parallel-group trial that evaluated the same four doses of BI-1358894 (5 mg, 25 mg, 75 mg, and 125 mg) against placebo.[3] The trial enrolled 390 patients with a DSM-5 diagnosis of BPD across 67 centers in 17 countries.[3]
The outcome was consistent with the findings in MDD. The trial failed to meet its primary endpoint, which was the change from baseline in the Zanarini Rating Scale for BPD (ZAN-BPD) total score at Week 10.[3] No significant differences were observed between the treatment and placebo groups for any secondary endpoints. Consequently, proof-of-concept for TRPC4/5 inhibition with BI-1358894 was not established for the treatment of BPD.[3] In their analysis, the study authors noted the inherent challenges of BPD research, including high placebo response rates, and suggested that future trials in this population should consider more targeted patient selection, alternative outcome assessments, and additional measures to mitigate placebo effects.[3]
Following the negative results in MDD and BPD, the development program has pivoted to focus on PTSD, an indication with a much stronger and more direct mechanistic link to the drug's established pharmacodynamic effects. The pathophysiology of PTSD is closely tied to a hyperactive fear circuit involving the amygdala, making it a more precise target for a therapy designed to attenuate amygdala hyperreactivity.[4]
The Phase II trial in PTSD (NCT05103657) was an 8-week, multicenter, randomized, double-blind, placebo-controlled study that enrolled 318 adult patients (aged 18-65) with a DSM-5 diagnosis of PTSD.[7] The trial was initiated in December 2021 and completed in November 2023.[7] The study's primary purpose is to evaluate the efficacy of orally administered BI-1358894 in improving PTSD symptoms compared to placebo. The primary efficacy measures are based on changes in scores on the Clinician-Administered PTSD Scale for DSM-5 (CAPS-5) and the self-reported PTSD Checklist for DSM-5 (PCL-5).[7] As of the latest updates, the results of this completed trial have not been made publicly available, and the developer's website continues to list the asset as "under investigation" for PTSD.[22]
A standout feature of the BI-1358894 clinical development program, consistent across all phases and patient populations, is its favorable safety and tolerability profile. In the context of modern psychopharmacology, where safety concerns and tolerability issues are major drivers of regulatory scrutiny and clinical prescribing behavior, this "clean" profile represents a primary strategic asset. It significantly de-risks the drug's future development and would provide a major commercial and clinical advantage should efficacy be demonstrated in any indication.
Table 2: Consolidated Profile of Treatment-Emergent Adverse Events (TEAEs) in Phase II Trials
| Adverse Event (AE) | BI 1358894 (Pooled MDD & BPD) | Placebo (Pooled MDD & BPD) | Quetiapine (MDD Trial Only) |
|---|---|---|---|
| Patients with any AE (%) | 66.7 (MDD), 77.9 (BPD) | 53.9 (MDD), 75.0 (BPD) | Not specified in detail, but higher than placebo |
| Patients with Serious AEs (SAEs) (%) | Not specified (MDD), 10.3 (BPD) | Not specified (MDD), 8.6 (BPD) | Not specified (MDD) |
| AEs Leading to Discontinuation (%) | 2.6 (MDD) | 5.5 (MDD) | 9.9 (MDD) |
| Headache (%) | 15.9 (MDD) | 10.2 (MDD) | Not specified |
| Dizziness (%) | 7.4 (MDD) | 2.3 (MDD) | Not specified |
| Somnolence (%) | 7.4 (MDD) | 3.1 (MDD) | Not specified |
| SAE of Suicidal Ideation (%) | 5.1 (MDD, 75mg group only), 4.2 (BPD, overall) | 4.7 (MDD), 6.3 (BPD) | 1.4 (MDD) |
| Source(s) | 2 | 2 | 2 |
Across all Phase I and Phase II trials, BI-1358894 was consistently described by investigators as being "well tolerated".[2] The most frequently reported treatment-emergent adverse events that occurred at a higher rate than placebo in the MDD trial were headache (15.9% vs 10.2%), dizziness (7.4% vs 2.3%), and somnolence (7.4% vs 3.1%).[2] Similar AEs, including headache, dizziness, and fatigue, were also noted in Phase I studies.[12]
A powerful indicator of the drug's superior tolerability is the rate of discontinuation due to adverse events. In the large MDD trial, patients receiving BI-1358894 were significantly less likely to discontinue treatment due to AEs (2.6%) than patients receiving placebo (5.5%) or the active comparator quetiapine (9.9%).[2] This suggests that, from a patient's perspective, the side effect burden of BI-1358894 was more manageable than that of either a sugar pill or a standard-of-care adjunctive therapy.
The overall incidence of serious adverse events was comparable between the BI-1358894 and placebo groups in the large Phase II trials. In the BPD study, 10.3% of patients in the combined BI-1358894 arms experienced an SAE, compared to 8.6% in the placebo arm.[3] No specific pattern of events or dose-dependent trend in SAEs was observed in the BI-1358894-treated groups.[2]
The most critical safety finding from the entire clinical program is the consistent lack of any signal for increased suicidality or self-harm. This is particularly noteworthy given that the trials were conducted in patient populations (MDD and BPD) with inherently elevated baseline risks for such events.
Across studies, investigators concluded there was "no worsening of Columbia-Suicide Severity Rating Scale (C-SSRS) for most patients" and "no increase in self-harm or suicidality".[2] This conclusion is supported by quantitative data on the most common SAE, suicidal ideation. In the BPD trial, the proportion of patients with an SAE of suicidal ideation was actually lower in the overall BI-1358894 group (4.2%) than in the placebo group (6.3%).[3] In the MDD trial, the rate in the 75 mg BI-1358894 group (5.1%) was comparable to that of the placebo group (4.7%).[2] No completed suicides were reported during the trials.[2] This neutral-to-favorable profile on suicidality is a significant advantage for a CNS drug and a key element of its overall risk-benefit assessment.
BI-1358894 stands at a critical juncture, embodying a common yet challenging narrative in CNS drug development where a promising, mechanistically elegant therapeutic fails to demonstrate clinical efficacy in its initial target indications. The program's trajectory reveals a central paradox: a drug with a strong scientific rationale and confirmed target engagement in the human brain did not produce a measurable clinical benefit in two large, well-designed Phase II trials. A critical analysis of this disconnect is essential for understanding the drug's future prospects.
The failure of BI-1358894 in MDD and BPD likely stems not from a failure of the drug's biological mechanism, but from a misapplication of that mechanism to overly broad and complex clinical disorders. Several factors may have contributed to this efficacy-mechanism disconnect:
The decision by Boehringer Ingelheim to complete the Phase II trial in PTSD represents a logical and necessary strategic pivot based on the learnings from the MDD and BPD studies. The PTSD trial (NCT05103657) is not merely another attempt in a different disorder; it is the program's decisive moment and a much more direct test of the core therapeutic hypothesis. Unlike the broad syndromes of MDD and BPD, the pathophysiology of PTSD is more clearly and consistently linked to a hyperactive fear circuit centered on the amygdala.[4] The core symptom clusters of PTSD, such as intrusive memories, hypervigilance, and exaggerated startle responses, are direct clinical manifestations of this underlying neurobiological dysfunction. Therefore, a drug proven to attenuate amygdala activity should, in theory, have a more direct and measurable impact on these core PTSD symptoms. Success in this trial would validate the amygdala-attenuation hypothesis in a highly relevant patient population. Conversely, failure in this mechanistically-aligned indication would cast serious doubt on the therapeutic utility of this mechanism for any psychiatric disorder and would likely lead to the termination of the program.
BI-1358894 is a high-risk, high-reward asset. Its development program has successfully de-risked its safety profile but has thus far failed to establish clinical efficacy. The continuation of its development with a clear focus on PTSD reflects a sound scientific strategy to give the drug's novel mechanism its best possible chance to succeed in a patient population where it is most likely to have an effect.
The most undeniable and valuable characteristic of BI-1358894 is its excellent safety and tolerability profile, particularly its neutral effect on suicidality. In the event of a positive efficacy outcome in the PTSD trial, this safety profile would become a powerful differentiating feature, positioning the drug as a potentially safe and well-tolerated option for a difficult-to-treat condition.
The final verdict on BI-1358894 now awaits the public disclosure of the results from the NCT05103657 trial. A positive outcome could resurrect the asset, validating its first-in-class mechanism and establishing a new therapeutic option for trauma-related disorders. A negative outcome would likely confirm that, despite its elegant mechanism and proven biological activity, BI-1358894 is unable to translate its effects into meaningful clinical improvement, relegating it to the long list of promising but ultimately unsuccessful CNS drug candidates.
Published at: September 26, 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.