What drugs are in development for Depressive Disorder?

12 March 2025
Overview of Depressive DisorderDefinitionon and Types Depressive disorder broadly refers to a heterogeneous spectrum of conditions that share the common feature of pervasive low mood and diminished interest or pleasure. Major depressive disorder (MDD) is the most frequently diagnosed form, with an estimated lifetime risk of 15–18% and a prevalence that affects hundreds of millions worldwide. Along with MDD, clinical presentations include subthreshold or minor depression, treatment‐resistant depression (TRD), bipolar depression (the depressive phase in bipolar disorder) and postpartum depression. The disorder is not only defined by psychological symptoms such as persistent sadness, hopelessness, guilt and anhedonia, but also by associated issues such as cognitive dysfunction, changes in appetite and sleep disturbances, and sometimes somatic complaints. Moreover, many patients exhibit comorbidities such as anxiety disorders, substance use disorders, and even physical illnesses, which complicate the clinical presentation further.

Current Treatment Landscape The predominant treatment for depressive disorders has historically revolved around pharmacotherapy with monoaminergic agents. These include selective serotonin reuptake inhibitors (SSRIs), serotonin–norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants and monoamine oxidase inhibitors (MAOIs). Although these agents benefit many, they have important limitations; delayed onset of action, modest response/remission rates (with up to one-third of patients remaining symptomatic after multiple treatment steps), and undesirable side effects often lead to a challenging management context—especially for patients with TRD or those with severe comorbidities. Because of these shortcomings, research has increasingly focused on innovative compounds and alternative mechanisms beyond the conventional monoamine theory.

Drugs in Development

Novel Drug Classes A large number of novel therapeutic classes are emerging that target non‐monoaminergic mechanisms to provide faster onset and improved efficacy. Data from multiple synapse sources have highlighted several promising classes:

• NMDA Receptor Antagonists: Ketamine and its related formulations (esketamine nasal spray) have already demonstrated rapid antidepressant effects in TRD and have spurred the development of related compounds that aim to act on the glutamatergic system without ketamine’s dissociative side effects. New agents such as GLYX-13 (rapastinel), NRX-1074 (Apimostinel) and AV-101 are being investigated as alternatives. These drugs target the NMDA receptor complex either by blocking specific subunits or modulating the receptor indirectly, resulting in a rapid onset of action.

• Triple Reuptake Inhibitors and Combination Compounds: AXS-05—an investigational fixed-dose combination of dextromethorphan and bupropion—has shown positive results in phase III trials, providing a novel approach by inhibiting the reuptake of serotonin, norepinephrine, and dopamine. Likewise, ansofaxine hydrochloride is another compound in development that also targets multiple monoaminergic reuptake sites but with an innovative pharmacodynamic profile.

• Psychedelic-Based Therapies: There has been a resurgence of interest in classical psychedelic compounds such as psilocybin, ayahuasca, and novel formulations of 5-MeO-DMT. These agents are being reformulated, micro-dosed, or altered chemically to reduce hallucinogenic effects while keeping the rapid antidepressant potential intact. The use of these therapies, supported by careful psychological support during administration, has produced promising early signals for TRD and severe depression.

• Neuroactive Steroids: Brexanolone, approved for postpartum depression, has paved the way for oral analogues like zuranolone (SAGE-217) that modulate GABA_A receptor function. These agents are intended to produce rapid and sustained antidepressant responses via neurosteroid mechanisms, which may also improve safety and ease of administration over infusions.

• Agents Acting on Inflammatory and Neurotrophic Pathways: Given the growing evidence for the role of inflammation and impaired neuroplasticity in depression, compounds that target anti-inflammatory and neuroprotective pathways are also in development. Some drugs are focused on modulating cytokine levels and reducing neuroinflammation, while others aim to enhance brain-derived neurotrophic factor (BDNF) signaling and promote neurogenesis. Although these drugs are not yet fully classified as a separate group, they represent a novel mechanistic approach.

• Cholinergic and Opioidergic Modulators: Recent research has also focused on agents interacting with the cholinergic system as well as partial opioid receptor agonists or antagonists to rebalance endogenous opioid signaling in depression. These compounds are increasingly recognized as having potential utility in both unipolar and bipolar depressive episodes.

• Other Combination Strategies and Prodrug Approaches: Beyond these more clearly defined classes, there is development of several combination strategies including formulations that blend novel mechanisms (such as combining a dextromethorphan derivative with a monoamine reuptake inhibitor) offering synergistic biological profiles. Moreover, prodrug strategies—such as bifunctional psilocin prodrugs (BPPs) and novel psilocin prodrugs (NPPs)—are designed to optimize pharmacokinetics while minimizing psychedelic adverse events.

Clinical Trial Phases The development pipeline for these drugs spans early-phase exploratory trials to larger phase III studies:

• Preclinical and Phase I: Many of the new compounds have undergone rigorous preclinical testing using animal models to evaluate efficacy and toxicity. Agents such as novel NMDA receptor modulators, selective triple reuptake inhibitors, and early iterations of neuroactive steroids have been evaluated in Phase I clinical trials. These studies focus on dose-escalation, tolerability, and pharmacokinetic profiling, establishing safe dose limits and preliminary signals of target engagement.

• Phase II Trials: In phase II studies, the focus shifts to establishing preliminary efficacy in patient populations along with continued safety assessment. For example, multiple Phase II trials for psilocybin and other psychedelic-based therapies have been reported, aiming to document improvements on standard depression rating scales. Several compounds acting on glutamatergic mechanisms (such as GLYX-13 or related modulators) have reached this stage and are currently being evaluated in randomized, placebo-controlled settings. Additionally, combination agents like AXS-05 have successful phase II data that support their move into larger trials.

• Phase III Trials: Phase III trials are underway for a number of candidates including newer triple reuptake inhibitors and NMDA receptor antagonists. Esketamine, which was advanced in earlier trials due to its rapid antidepressant action, is also inspiring parallel phase III studies of drugs with similar mechanisms but improved tolerability profiles. Compounds such as ansofaxine hydrochloride and AXS-05 are undergoing phase III evaluation to confirm efficacy in larger patient samples with TRD and to compare their performance against active controls and placebos. These larger studies are critical for regulatory approvals and assessing longer-term safety and maintenance of antidepressant effects over several weeks to months.

Mechanisms of Action

Targeted Biological Pathways The emerging drugs are being developed by targeting multiple biological systems that are thought to be at the heart of depressive pathophysiology:

• Glutamatergic System: One of the most transformative advances in the field has been the recognition that dysregulation of glutamate neurotransmission plays a key role in depression. Drugs such as ketamine and its derivatives target NMDA receptors to produce rapid effects. In addition, novel compounds are now targeting specific subunits (e.g., GluN2B) or using allosteric modulators to modulate the receptor activity in a way that enhances synaptic plasticity without the dissociative side effects traditionally seen with ketamine.

• Monoaminergic Systems – Expanded Beyond SSRIs: Although SSRIs and SNRIs remain mainstays in the current treatment landscape, innovative agents that inhibit the reuptake of three monoamine neurotransmitters simultaneously (serotonin, norepinephrine, and dopamine) have been developed. For instance, AXS-05 and ansofaxine hydrochloride provide a broader spectrum of neurotransmitter modulation which may contribute to both improved efficacy and a more rapid onset of action.

• GABAergic Neuroactive Steroids: Brexanolone and oral analogues such as zuranolone modulate the GABA_A receptor complex. Neuroactive steroids work by enhancing inhibitory neurotransmission and may also have the added benefit of modulating neuroplasticity and stress-related circuitry in the brain.

• Inflammatory Pathways and Neurotrophic Factors: A growing body of evidence supports the idea that increased inflammation and reduced neurotrophic support (for example, decreased BDNF signaling) contribute to depression. Novel drug candidates are being developed that either inhibit pro-inflammatory cytokines or enhance neurotrophic factor signaling. Such agents aim to restore homeostatic balance by reducing neuroinflammation and promoting synaptic repair and neurogenesis.

• Cholinergic and Opioidergic Systems: Recent studies have implicated the cholinergic system in mood regulation. New agents targeting cholinergic receptors or modulating their activity have emerged as novel treatments. Likewise, drugs that interact with opioid receptors in a selective manner (without producing the full spectrum of opioid effects) are being investigated to correct dysregulated endogenous opioid tone seen in depression.

Innovations in Drug Mechanisms The drugs in development are notable not only for their novel targets but also for innovative pharmacokinetic and pharmacodynamic approaches:

• Rapid-Acting Formulations: Unlike traditional antidepressants that require weeks for effect, many new compounds (especially those acting on the glutamatergic system) have demonstrated therapeutic changes within hours. This rapid onset is thought to involve both rapid modulation of synaptic connectivity and neuroplasticity as well as immediate downstream intracellular signaling cascades that reset dysfunctional neural circuits.

• Combination and Fixed-Dose Approaches: Innovative combination products such as AXS-05 exemplify the trend toward co-formulated agents that are designed to target multiple neurotransmitter systems concurrently. By combining agents with complementary mechanisms in a fixed-dose formulation, these drugs aim to achieve greater efficacy with possibly fewer side effects.

• Prodrug Strategies and Receptor Sub-type Specificity: Prodrug formulations such as bifunctional psilocin prodrugs (BPPs) have been engineered to optimize absorption and to release active compounds gradually, minimizing acute adverse effects like hallucinations while preserving therapeutic benefit. Similarly, efforts to improve receptor subtype specificity—such as targeting specific NMDA receptor subunits or selective activation/inhibition of subtypes of serotonin receptors—are at the forefront of modern drug design.

Challenges and Future Directions

Current Development Challenges Despite many advances, significant challenges persist in the current phase of drug development for depressive disorder:

• Heterogeneity of Depression: The complexity and heterogeneity of depressive disorders create challenges for clinical trial design and drug development. With multiple symptom dimensions and varying degrees of treatment resistance across patients, it is difficult to identify biomarkers that predict response to any single agent. This heterogeneity means that even promising drugs may only benefit a subset of patients, thereby complicating clinical trial outcomes and regulatory approval.

• Balancing Efficacy and Safety: Developing agents with rapid onset yet minimal side effects remains a daunting task. Ketamine, for instance, has shown impressive results but its potential for dissociation and abuse limits widespread use. Researchers now aim to replicate rapid-acting properties without such adverse effects—a challenge that requires novel compounds and optimized dosing regimens.

• Placebo Effects and Trial Design Issues: Depression trials are notorious for high placebo response rates. This phenomenon can obscure the true efficacy of a novel agent and necessitates highly powered studies with rigorous methodology to ensure that even modest differences can be statistically validated. Carefully selecting patients based on severity and treatment history, as well as designing trials for long-term outcomes, are critical aspects that must be addressed.

• Translational Bridging: Moving from successful preclinical results to efficacy in human subjects remains one of the greatest hurdles. Many compounds demonstrate strong antidepressant effects in animal models but then fail to produce similar outcomes in clinical studies. This “translation gap” is partly due to differences in neurobiology between species and the multifactorial nature of depression in humans.

Future Prospects in Drug Development Looking ahead, the future of antidepressant development is promising, driven by multiple innovations and a deeper understanding of the underlying molecular pathways:

• Biomarker-Driven Personalization: Advances in pharmacogenomics and neuroimaging offer the potential for stratifying patients based on biological markers. By identifying those who may benefit from a certain drug class (for example, patients with elevated inflammatory markers or specific glutamatergic dysfunction), treatment can become more personalized. This approach promises to improve success rates by matching drugs to patient subtypes rather than treating depression as a single homogeneous disorder.

• Rapid-Acting Therapies with Sustained Benefits: The development of rapid-acting compounds—especially those that work by modulating the NMDA receptor complex—has revolutionized the understanding of how quickly depression symptoms can be alleviated. The next frontier will be to sustain these rapid responses over longer durations with fewer relapses, possibly through combination regimens or adjunctive therapies that maintain neural circuit stability.

• Multimodal and Combination Therapeutics: Future drug development is likely to move toward multimodal strategies, where the combination of drugs that act on different systems (for example, combining a glutamatergic modulator with a neuroactive steroid) may provide synergistic effects. This could lead to a new generation of antidepressants that offer both rapid onset and durable efficacy, while also addressing the diverse biological abnormalities seen in depression.

• Innovation in Regulatory Pathways: As novel agents emerge with mechanisms outside traditional targets, regulatory bodies are adapting. The acceptance of compounds like esketamine and the growing number of breakthrough therapy designations for rapid-acting antidepressants signal a regulatory climate that could accelerate the approval of innovative drugs. Changes in trial endpoints, including the use of more sensitive biomarkers and functional imaging outcomes, will likely improve the detectability of drug effects in clinical studies.

• Expanding the Therapeutic Spectrum: Future research may also investigate adjunct therapies such as transcranial magnetic stimulation, vagal nerve stimulation, and even stem cell-based approaches in combination with pharmacotherapy. With ongoing studies into the neurobiological correlates of depression—including neurogenesis, synaptic plasticity, and neuroinflammation—the emerging pharmacological strategies will likely be complemented by these device-based and biological therapies to offer a holistic treatment plan.

• Longitudinal and Real-world Data Collection: Emerging clinical trials are increasingly designed to include long-term follow-up measurements to assess the durability of antidepressant responses. In addition, real-world evidence from post-marketing studies may help refine dosing regimens and further elucidate safety and effectiveness in diverse patient populations. This comprehensive approach should ultimately lead to improved treatment outcomes and decreased disease burden.

Conclusion In summary, drugs in development for depressive disorder are spanning a wide array of novel drug classes and mechanisms that go well beyond the conventional monoaminergic targets. The field now includes NMDA receptor antagonists and modulators, triple reuptake inhibitors such as AXS-05 and ansofaxine hydrochloride, psychedelic-based therapies (including micro-dosed psilocybin and 5-MeO-DMT formulations), neuroactive steroids like zuranolone, agents targeting neuroinflammatory and neurotrophic pathways, as well as compounds modulating cholinergic and opioidergic systems. Clinical trials are at various stages—from early phase I dose-finding studies to large phase III randomized controlled trials—with special emphasis on rapid-acting properties and improved tolerability. Innovative strategies include fixed-dose combination formulations, prodrug approaches designed to optimize drug release, and the leveraging of biomarkers to personalize treatment.

Current challenges in the development of these drugs relate to the heterogeneity of depressive disorders, the need to balance rapid efficacy with long-term safety, and difficulties in translating preclinical findings into clinical success. Despite these hurdles, future prospects are promising. Advances in genetic and neuroimaging biomarkers are expected to allow for more targeted, precision-based medicine, and regulatory developments will likely facilitate faster approvals for compounds with breakthrough potential.

In conclusion, the landscape of drug development for depressive disorders is evolving rapidly. With a multifaceted approach that combines novel biological targets with innovative clinical trial design, there is hope for more effective, rapid-acting, and personalized treatments for patients suffering from these debilitating conditions. The integration of multiple non-monoaminergic pathways promises to revolutionize how depression is managed and is likely to lead to substantial improvements in patient outcomes in the coming years.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.