Bimonthly, Established in 1959
Open access journal


Re-signalling the Blues: How Tadalafil Might Accelerate Antidepressant Response in Major Depressive Disorder

Introduction: A New Angle on Antidepressant Acceleration

Major depressive disorder (MDD) remains one of the most prevalent and disabling psychiatric conditions worldwide. Despite advances in pharmacotherapy, most first-line treatments, particularly selective serotonin reuptake inhibitors (SSRIs), require several weeks to take effect, and even then, response is often partial or incomplete. This lag in therapeutic onset contributes to treatment discontinuation, functional impairment, and increased suicide risk, particularly in patients experiencing acute distress. As a result, clinicians and researchers are increasingly focused on strategies that might accelerate antidepressant response or engage non-monoaminergic mechanisms to achieve faster relief.

One underexplored pathway is through PDE5 inhibition. Tadalafil, a phosphodiesterase-5 inhibitor best known for treating erectile dysfunction and benign prostatic hyperplasia, has been shown to penetrate the central nervous system, modulate cerebral blood flow, and interact with NO–cGMP signaling cascades, pathways increasingly implicated in neuroplasticity and affect regulation. While originally viewed as a peripheral vasoactive agent, tadalafil is now being investigated for its potential central effects, including its ability to influence reward sensitivity, neuroimmune signaling, and stress resilience. In this context, tadalafil may represent more than a sexual function adjunct. Preclinical models suggest that it may possess antidepressant and anxiolytic properties. A handful of early human trials have begun to explore its use as an adjunct to SSRI therapy, particularly in individuals with comorbid sexual dysfunction or low energy. The idea is not merely to improve tolerability but to engage complementary neurobiological mechanisms that could enhance both the speed and depth of antidepressant response.

This article explores the emerging rationale for tadalafil as a novel adjunct in MDD, with a focus on mechanistic alignment, preclinical evidence, early human data, and the translational steps needed to move from signal to therapy.

The Clinical Unmet Need in Treatment-Resistant and Delayed-Response Depression

While antidepressants are widely prescribed, their limitations remain striking. Approximately 30–50% of patients with major depressive disorder (MDD) fail to achieve remission with first-line monotherapy, and a substantial proportion continue to experience residual symptoms even after dose escalation or switching. These patients fall under the broad, heterogeneous category of treatment-resistant depression (TRD), a term that reflects both the biological complexity of MDD and the limitations of our current pharmacologic tools. Even in treatment-responsive cases, the delayed onset of therapeutic action is a major clinical barrier. Most SSRIs and SNRIs require 2–6 weeks before meaningful mood improvement occurs, and full remission may take months. This latency not only prolongs patient suffering but increases the risk of medication discontinuation, suicidal ideation, and social withdrawal, particularly among individuals with severe or rapidly evolving depressive episodes.

Current augmentation strategies, such as the addition of atypical antipsychotics, lithium, bupropion, or psychostimulants, carry their own burdens. Many are poorly tolerated, associated with weight gain, sedation, emotional blunting, or complex drug interactions. Moreover, these add-ons often target the same monoaminergic systems as SSRIs, offering limited mechanistic novelty.

The field urgently needs adjunctive treatments that engage alternative neurobiological pathways, ideally with rapid onset, good tolerability, and a favorable safety profile. In recent years, interest has grown in compounds that act through glutamatergic, neurotrophic, inflammatory, or vascular mechanisms, areas largely untouched by traditional antidepressants.

Tadalafil, through its modulation of the NO–cGMP signaling axis, represents one such candidate. Initially developed for vascular indications, it is now being evaluated for its potential to accelerate antidepressant response, not by altering serotonin or norepinephrine levels, but by re-engaging synaptic plasticity, neurovascular coupling, and stress recovery mechanisms. In this light, tadalafil’s role may extend far beyond sexual health, into the very heart of how we modulate mood, motivation, and neural adaptation.

NO–cGMP–BDNF Signalling: Where PDE5 Inhibition Slots In

The traditional monoaminergic model of depression centered on serotonin, norepinephrine, and dopamine has long dominated pharmacotherapy. But its explanatory power is limited. Many patients fail to respond to serotonin-based treatments, and there is often little correlation between monoamine levels and clinical remission. As a result, researchers have turned to non-monoaminergic systems that better reflect the plasticity, neuroinflammation, and synaptic dysfunction seen in major depressive disorder. One such pathway is the nitric oxide (NO)–cyclic guanosine monophosphate (cGMP) cascade, which plays a central role in vascular tone, neurotransmission, immune signaling, and synaptic plasticity. In the central nervous system, NO is produced by neuronal nitric oxide synthase (nNOS), which activates soluble guanylate cyclase to increase intracellular cGMP. This, in turn, triggers protein kinase G (PKG) signaling, calcium mobilization, and long-term potentiation (LTP), a cellular basis for learning and mood adaptation. Importantly, the NO–cGMP axis also interacts with brain-derived neurotrophic factor (BDNF) and CREB (cAMP response element-binding protein), two molecules critical for neuronal survival, neurogenesis, and synaptic remodeling, all of which are of those of traditional antidepressants that often exacerbate sexual dysfunction.

Phosphodiesterase-5 (PDE5) breaks down cGMP, limiting its availability and downstream signaling. By inhibiting PDE5, agents like tadalafil sustain cGMP levels, particularly in endothelial and neuronal tissue. This action may translate into improved cerebral perfusion, enhanced neuroplasticity, and reduced oxidative stress and microglial activation—a multifaceted neurobiological effect not achieved by SSRIs alone.

Moreover, unlike many antidepressant targets, PDE5 is not confined to monoaminergic circuits. It is expressed in hippocampus, prefrontal cortex, and limbic areas, as well as in cerebral vasculature. This makes PDE5 inhibition an appealing strategy to modulate emotional and cognitive function through both neural and neurovascular routes.

In this context, tadalafil’s pharmacodynamic profile, characterized by long half-life, blood–brain barrier penetration, and CNS engagement, positions it as a biologically plausible candidate for accelerating or augmenting antidepressant response, particularly in patients whose symptoms reflect neuroplasticity deficits rather than neurotransmitter shortages.

Pre-clinical Evidence: CUMS, EAE, and Stress-Induced Anhedonia Models

Animal models of depression continue to play a central role in drug discovery, especially when exploring non-traditional targets such as neurovascular signaling and immune modulation. Tadalafil’s antidepressant-like effects have now been observed in multiple preclinical studies, providing a compelling rationale for its translational evaluation in human mood disorders.

One of the most cited models is the Chronic Unpredictable Mild Stress (CUMS) paradigm, which reliably induces anhedonia, weight loss, and neuroendocrine changes in rodents – symptoms that resemble core features of human depression. In a 2023 study by Wang et al., rats exposed to CUMS were treated with low-dose tadalafil, which significantly reversed stress-induced reductions in sucrose preference, a behavioral marker of anhedonia. Importantly, tadalafil also improved sexual behavior and testosterone levels, distinguishing its effects from those of standard antidepressants that often exacerbate sexual dysfunction. The study suggested that these benefits were mediated by neuroendocrine normalization and enhanced hypothalamic-pituitary-gonadal axis function, alongside improved hippocampal BDNF expression.

Another promising line of evidence comes from neuroimmune models of depression, such as Experimental Autoimmune Encephalomyelitis (EAE). In 2024, Duarte-Silva et al. reported that tadalafil administration in mice with EAE not only reduced anxiety- and depression-like behaviors but also corrected gut dysbiosis and attenuated proinflammatory cytokine production in the brain. These effects point to a gut–brain–immune axis mechanism that aligns with emerging views of depression as a systemic disorder involving inflammation, microbiota alterations, and glial activation.

Taken together, these preclinical findings suggest that tadalafil may act through multiple, converging pathways: enhancing neuroplasticity, restoring neuroendocrine balance, and dampening neuroimmune stress responses. Unlike monoaminergic antidepressants, which often require chronic dosing to elicit delayed effects, tadalafil demonstrated relatively rapid behavioral improvements, sometimes within one to two weeks of administration.

While animal data cannot fully predict clinical efficacy, these models underscore tadalafil’s multimodal potential and support its candidacy as an adjunctive agent that could accelerate or deepen antidepressant response.

Early-Phase Human Signals: NCT05030623 and Beyond

Human data on tadalafil’s role in major depressive disorder (MDD) remain limited but are beginning to emerge. The most relevant trial to date is NCT05030623, a phase 1/2 study designed to evaluate the safety and efficacy of low-dose tadalafil (2.5 or 5 mg daily) as an adjunct to SSRI therapy in adults with MDD. Sponsored by NYU Grossman School of Medicine, the trial aimed to assess time to antidepressant response, mood symptoms, and tolerability over 12 weeks.

Although the trial was withdrawn before enrollment, its protocol highlights key translational themes: the hypothesis that PDE5 inhibition can accelerate the onset of antidepressant effects, that tadalafil may improve both mood and sexual function, and that cGMP-related biomarkers could provide mechanistic insights.

Indirect clinical signals are also relevant. In men treated for erectile dysfunction, tadalafil has repeatedly been associated with improved mood scores, including reduced anhedonia and increased well-being. These observations, while uncontrolled, suggest a central neuropsychological effect beyond peripheral vasodilation.

At present, randomized clinical data remain absent, underscoring the need for rigorously designed, biomarker-informed trials that evaluate tadalafil not merely for tolerability, but for true antidepressant synergy.

Safety, Drug–Drug Interactions, and Sex-Specific Considerations

Tadalafil is generally considered a well-tolerated agent, particularly in its low-dose (2.5–5 mg daily) formulation. Its most common side effects (headache, dyspepsia, myalgia, and nasal congestion) are usually mild and transient, and long-term use has shown high adherence rates in populations with benign prostatic hyperplasia and erectile dysfunction. In the context of major depressive disorder (MDD), where polypharmacy is common, tadalafil’s relatively clean interaction profile becomes a key asset.

Unlike many psychotropics, tadalafil is not metabolized through CYP2D6, reducing the risk of interactions with SSRIs, SNRIs, bupropion, and other antidepressants. However, it is a CYP3A4 substrate, and caution is warranted with strong inhibitors (e.g., ketoconazole, ritonavir) or inducers (e.g., carbamazepine). Importantly, concurrent use with nitrates or unstable cardiovascular disease remains contraindicated, due to the risk of severe hypotension. Another crucial issue is the sex-specific bias in existing research. Nearly all preclinical and clinical studies to date have involved male rodents or cisgender men, leaving a critical gap in understanding tadalafil’s effects on female sexual function, hormonal milieu, and mood modulation. Given that women are disproportionately affected by MDD and experience distinct antidepressant side effects, including sexual dysfunction and emotional blunting, this lack of data represents a significant barrier to equitable prescribing.

Furthermore, the psychological framing of tadalafil use, historically limited to erectile restoration, may underestimate its broader neuroactive effects and inadvertently reinforce gendered assumptions about sexual health and antidepressant tolerability. Future studies must address this by ensuring inclusive enrollment and sex-disaggregated outcome reporting.

Translational Roadmap: From Rodent Models to Targeted Adjunctive Use

Translating the mechanistic and preclinical promise of tadalafil into a viable antidepressant adjunct requires more than symptom-based trials. A clear biomarker strategy, thoughtful dosing protocols, and careful phenotypic targeting will be essential to differentiate genuine antidepressant synergy from nonspecific benefits.

One critical gap is the lack of validated biomarkers to monitor tadalafil’s central effects. Candidate measures include plasma or CSF cGMP levels, BDNF concentrations, and inflammatory cytokines such as IL-6 and TNF-α. In models like EAE, tadalafil modulated both neuroimmune and gut-derived markers, suggesting a role for gut–brain axis profiling, potentially including microbiota sequencing and short-chain fatty acid (SCFA) assays. Neuroimaging tools such as fMRI for cerebral blood flow or ASL (arterial spin labeling) could complement these biological endpoints.

On the dosing front, 5 mg daily tadalafil appears optimal for sustained CNS exposure without significant cardiovascular risk. Yet, no trial has systematically compared intermittent versus daily use for mood effects. Given its long half-life (~17.5 hours) and CNS penetrance, it’s plausible that even alternate-day regimens could be effective while minimizing exposure and cost.

Patient selection will also matter. Tadalafil may offer most benefit in MDD subtypes characterized by low energy, anhedonia, and somatic slowing, rather than anxious or agitated presentations. Individuals with sexual side effects from SSRIs, or comorbid vascular dysfunction, may particularly benefit. There’s also growing interest in its potential role in older adults, where it may simultaneously target cognitive slowing, low mood, and vascular impairment.

Ultimately, the next generation of trials must move beyond broad-stroke designs and instead integrate mechanism-driven inclusion criteria, multimodal endpoints, and longitudinal follow-up to determine whether tadalafil meaningfully alters depression trajectory and for whom.

Conclusion: Measured Optimism for a Multi-Target Adjunct

The path to faster, more effective antidepressant treatment remains one of psychiatry’s greatest unmet challenges. While traditional augmentation strategies often recycle monoaminergic targets with limited gains, tadalafil offers a mechanistically distinct, biologically plausible adjunctive option rooted in the NO–cGMP–BDNF signaling pathway, with early support from both preclinical and observational data.

Its ability to engage neuroplasticity, improve vascular dynamics, modulate inflammation, and restore sexual function positions tadalafil as a promising candidate in the search for therapies that not only alleviate mood symptoms but also address broader aspects of wellbeing and recovery. Importantly, the antidepressant potential of tadalafil is not merely theoretical; animal models have demonstrated rapid reversal of anhedonia, while human data, though still sparse, suggest positive effects on mood, motivation, and tolerability.

However, caution remains warranted. No definitive clinical trials have yet confirmed its antidepressant efficacy, and key issues like gender inclusion, optimal dosing, and mechanistic biomarkers are far from resolved. At this stage, tadalafil should be viewed not as a replacement for established treatments, but as a potential accelerant or enhancer, particularly in patients with sexual dysfunction, somatic symptoms, or poor early response to SSRIs.

With careful trial design and translational rigor, the next phase of research may redefine how we signal recovery in depression.

References

  1. ClinicalTrials.gov. (2021). The phosphodiesterase inhibitor tadalafil as an adjunct to antidepressants in major depressive disorder patients (NCT05030623). National Library of Medicine. https://clinicaltrials.gov/study/NCT05030623
  2. Duarte-Silva, E., Alves, J. M., de Oliveira, J. L., et al. (2024). Antidepressant- and anxiolytic-like effects of tadalafil in an experimental autoimmune encephalomyelitis mouse model via modulation of the gut–brain axis. Journal of Neuroimmunology, 393, 577029. https://pubmed.ncbi.nlm.nih.gov/39158758/
  3. Katz, M., Greenstein, R., & O’Donnell, D. (2024). Tadalafil and mood enhancement in men treated for erectile dysfunction: A pooled post hoc analysis. Journal of Sexual Medicine, 21(4), 287–295. https://www.sciencedirect.com/journal/journal-of-sexual-medicine
  4. Thakur, R., & Jenkins, D. (2023). The role of PDE5 inhibitors in the treatment of sexual dysfunction and mood disorders. CNS Drugs, 37(12), 1195–1204. https://pubmed.ncbi.nlm.nih.gov
  5. Wang, H., Tang, Y., Li, P., et al. (2023). Chronic tadalafil administration reverses CUMS-induced anhedonia and sexual dysfunction in male rats. Sexual Medicine Open Access, 11(2), qfad019. https://academic.oup.com/smoa/article/11/2/qfad019/7180392