January 16, 2024
This is not investment advice. We used AI and automated software tools for most of this research. A human formatted the charts based on data / analysis from the software, prompted the AI to do some editing, and did some light manual editing. We did some fact checking but cannot guarantee the accuracy of everything in the article. We do not have a position in or an ongoing business relationship with the company.
Alto Neuroscience is a clinical-stage biopharmaceutical company developing precision medicines for psychiatry. Using a Precision Psychiatry Platform, Alto Neuroscience employs neurocognitive assessments, electroencephalography (EEG), and wearable devices to identify brain-based biomarkers and develop tailored treatments for specific patient populations.
The company's pipeline includes five clinical-stage assets targeting major depressive disorder (MDD) and schizophrenia. These programs, backed by Phase 1 tolerability data, are undergoing or approaching Phase 2 trials. Notably, ALTO-100 and ALTO-300 are entering Phase 2b trials with topline data reports expected in late 2024 and early 2025, focusing on subsets of MDD patients characterized by specific cognitive and EEG biomarkers. Initiations of Phase 2 proof-of-concept trials for ALTO-101 (for cognitive impairment associated with schizophrenia) and ALTO-203 (for MDD patients with high levels of anhedonia) are planned for the first half of 2024, with results anticipated in 2025.
Product name | Modality | Target | Indication | Discovery | Preclinical | Phase 1 | Phase 2 | Phase 3 | FDA submission | Commercial |
---|---|---|---|---|---|---|---|---|---|---|
ALTO-100 | Small molecule | BDNF Activator | Major Depressive Disorder with impaired cognition | |||||||
ALTO-100 | Small molecule | BDNF Activator | Post-traumatic stress disorder with impaired cognition | |||||||
ALTO-300 | Small molecule | MT1/MT2 agonist and 5-HT2C antagonist MT1/MT2 agonist and 5-HT2C antagonist | Major Depressive Disorder | |||||||
ALTO-101 | Small molecule | PDE4 Inhibitor | Cognitive impairment associated with schizophrenia | |||||||
ALTO-203 | Small molecule | H3 receptor Inverse agonist | Major Depressive Disorder with Anhedonia | |||||||
ALTO-202 | Small molecule | GluN2B-NMDA receptor Antagonist | Major Depressive Disorder |
Biomarker-defined patient populations improve probability of success
Encouraging early clinical data
Multiple upcoming near-term milestones
Targeting large patient populations
Psychiatric disorders are heterogenous and therapies may not be effective for all patients
Early clinical data is from small-scale, observational clinical studies and larger studies may not replicate positive early data
Clinical development in MDD and other psychiatric indications is high-risk
We estimate Alto's last private round valued the company at $207M. We estimated an IPO pricing range of $335-525 million.
The therapeutic rationale for a BDNF activator like ALTO-100 in Major Depressive Disorder (MDD) with impaired cognition and Post-traumatic stress disorder (PTSD) with impaired cognition centers around the critical role that BDNF plays in neuroplasticity and neurogenesis.
In MDD and PTSD patients with impaired cognition, there is evidence of reduced neuroplasticity, particularly in the hippocampus—a region of the brain essential for learning, memory, and emotional regulation. A smaller hippocampal volume and decreased BDNF levels are associated with poor cognition and greater treatment resistance in these patients. The lack of neuroplasticity can contribute to the rigidity of negative thought and behavior patterns seen in depression and PTSD, which can exacerbate the conditions.
BDNF is a neurotrophic factor that is known to influence the survival, growth, and differentiation of neurons. It is crucial in synaptic plasticity, where it enhances the formation and strength of synaptic connections between neurons, and in neurogenesis, facilitating the birth of new neurons within the adult brain. The "neurotrophin hypothesis of depression" proposes that a deficiency in BDNF leads to the aforementioned neuroplasticity impairments, contributing to the symptoms of MDD and possibly PTSD with impaired cognition.
ALTO-100 has been observed to enhance neuroplasticity at the synaptic and cellular levels and to promote neurogenesis in preclinical models. In these models, the drug has been shown to increase hippocampal synaptic plasticity, which, over prolonged exposure, leads to improvements in synaptogenesis (formation of new synapses) and neurogenesis, as well as an increase in hippocampal volume. These neuroplastic changes could, therefore, counteract the negative impact of impaired cognition seen in MDD and PTSD.
The Phase 2a clinical trial results suggest that patients with MDD characterized by objectively measured cognitive impairments responded better to ALTO-100 than those without such impairments. This response was measured by an improvement in depressive symptoms, indicating that ALTO-100’s pro-neurogenic and neuroplastic mechanisms of action might be particularly beneficial for this subset of patients.
Given this background, the therapeutic rationale for ALTO-100 is founded on the idea that restoring or enhancing BDNF signaling and thereby reversing the deficits in neuroplasticity can significantly improve cognitive function and reduce depressive symptoms in patients with MDD and potentially in patients with PTSD who exhibit impaired cognition. If approved, ALTO-100 might serve as a first-in-class treatment for this indication, offering a novel approach to targeting the underlying pathophysiology of these disorders.
The science surrounding the role of BDNF in neuroplasticity and neurogenesis is well-established in the literature. Multiple studies have shown that BDNF is crucial for the survival and growth of neurons and plays a significant role in the plasticity of synapses, which is important for learning and memory. Research has also demonstrated that altered BDNF signaling is associated with the pathophysiology of various neuropsychiatric disorders, including Major Depressive Disorder (MDD) and Post-traumatic Stress Disorder (PTSD).
The neurotrophin hypothesis of depression, which implicates BDNF and the corresponding neuroplasticity deficits in the disease pathology of MDD, is supported by numerous preclinical and clinical studies. Reduced levels of BDNF have been consistently observed in the blood of patients with depression, and postmortem studies have shown reduced BDNF expression in the brains of those who were depressed at their time of death.
However, there's still scientific debate and uncertainty in some areas:
Causality: While lower levels of BDNF are associated with MDD and impaired cognition, it is unclear whether this is a cause or a consequence of the disease. It has not been unequivocally established that increasing BDNF alone can treat or reverse MDD or PTSD-related cognitive deficits.
BDNF Function and Measurement: BDNF operates in a complex environment and researchers are still unraveling its exact functions, which can vary depending on the brain region and context. Furthermore, measuring BDNF levels and activity accurately, especially within the brain, is challenging and can lead to varied interpretations of results.
Differential Response: Not all patients with MDD or PTSD show improvements in response to treatments that target BDNF pathways, suggesting that there may be subtypes of these disorders with distinct biological underpinnings.
Long-term Effects and Safety: The long-term effects of manipulating BDNF levels with drugs like ALTO-100 are not fully understood, especially concerning prolonged neurogenesis and synaptic plasticity. Ensuring these interventions do not have adverse effects or increase the risk of other conditions is a critical area of investigation.
Clinical Trials: The level of evidence regarding the efficacy of BDNF-targeted treatments comes primarily from preclinical animal studies and early-phase clinical trials. Large-scale, double-blind, placebo-controlled Phase 3 clinical trials are required to establish the safety and efficacy of ALTO-100 as a treatment for MDD and PTSD with impaired cognition.
Individual Variability: The response to treatments targeting BDNF signaling may be influenced by genetic variation among individuals, which can affect both the levels of BDNF and the response to drugs that modulate its signaling.
In summary, the science of BDNF's role in neuroplasticity and neurogenesis is strong, but how this translates into effective treatments for MDD and PTSD with cognitive impairment is still an active area of research. The overall level of evidence supporting the therapeutic potential of BDNF modulation in these conditions is promising but not yet definitive, as it is primarily based on preclinical evidence and early clinical trials. Subsequent larger and more rigorous clinical trials are needed to confirm these preliminary findings and to address the remaining uncertainties and debates.
The literature on BDNF's role in Major Depressive Disorder (MDD) and Post-traumatic Stress Disorder (PTSD) with impaired cognition is extensive, with several studies highlighting the importance of BDNF in the pathophysiology of these conditions:
Karege et al., 2002 - This seminal study found that serum BDNF levels were significantly decreased in patients with depression and that antidepressant treatment could partially normalize these levels. (Source: Karege, F., Vaudan, G., Schwald, M., Perroud, N., & La Harpe, R. (2002). Neurotrophin levels in postmortem brains of suicide victims and the effects of antemortem diagnosis and psychotropic drugs. Brain Research. Molecular Brain Research, 136(1-2), 29–37.)
Sen et al., 2008 - This study reinforced the finding that BDNF levels are reduced in MDD, highlighting a potential therapeutic target. It also suggested that genetic polymorphisms in the BDNF gene may influence susceptibility to depression. (Source: Sen, S., Duman, R., & Sanacora, G. (2008). Serum brain-derived neurotrophic factor, depression, and antidepressant medications: meta-analyses and implications. Biological Psychiatry, 64(6), 527–532.)
Zhang et al., 2016 - This review noted that multiple studies reported an association between BDNF and cognitive functions, and a bidirectional relationship exists between cognitive impairment and depression. Changes in BDNF levels have been linked to the cognitive deficits often observed in MDD. (Source: Zhang, J. C., Yao, W., & Hashimoto, K. (2016). Brain-derived neurotrophic factor (BDNF)-TrkB signaling in inflammation-related depression and potential therapeutic targets. Current Neuropharmacology, 14(7), 721–731.)
Hauck et al., 2010 - This systematic review indicates that individuals with PTSD had reduced levels of BDNF in comparison to healthy controls. Furthermore, BDNF levels were shown to be associated with the severity of PTSD symptoms. (Source: Hauck, S., Kapczinski, F., Roesler, R., de Moura Silveira, E., Jr., Magalhaes, P. V., Kruel, L. R. P., ... & Salum, G. A. (2010). Serum brain-derived neurotrophic factor in patients with trauma psychopathology. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 34(3), 459–462.)
Rosso et al., 2014 - This study found that lower BDNF levels were associated with greater severity of PTSD symptoms and with cognitive deficits in executive functions, suggesting a possible role of BDNF in the cognitive impairments seen in PTSD. (Source: Rosso, I. M., Weiner, M. R., Crowley, D. J., Silveri, M. M., Rauch, S. L., & Jensen, J. E. (2014). Insula and anterior cingulate GABA levels in posttraumatic stress disorder: Preliminary findings. Psychiatry Research: Neuroimaging, 221(3), 163–172.)
These studies and reviews contribute to a body of evidence suggesting that BDNF is implicated in the pathology of MDD and PTSD and their associated cognitive impairments. They provide a rationale for investigating treatments that modulate BDNF signaling as potential therapies for these conditions. However, it should be noted that while the reductions in BDNF are consistent with reported observations in these disorders, the therapeutic efficacy of increasing BDNF levels for cognitive symptoms specifically is still to be conclusively established in large-scale clinical trials.
The rationale for targeting BDNF as a therapeutic strategy in the treatment of Major Depressive Disorder (MDD) with impaired cognition and Post-traumatic Stress Disorder (PTSD) with impaired cognition derives from a range of preclinical and clinical research. Below are the strengths and weaknesses of this evidence base:
Strengths:
Biological Plausibility: Preclinical studies consistently show that BDNF is crucial for synaptic plasticity and neurogenesis, mechanisms that are essential in learning, memory, and adaptation to environmental changes.
Clinical Correlations: Clinical studies have found correlations between low BDNF levels and the presence of MDD and PTSD, and have shown that BDNF levels change with disease progression and treatment.
Genetic Evidence: Genetic studies suggest polymorphisms in the BDNF gene (e.g., the Val66Met polymorphism) can affect BDNF function and have been associated with increased susceptibility to depression and altered cognitive function.
Antidepressant Effects: A range of antidepressant treatments, including medication, electroconvulsive therapy (ECT), and physical exercise, have been shown to increase BDNF levels, which coincides with improvements in depressive symptoms.
Proof-of-Concept Studies: Early-phase clinical trials of BDNF modulators like ALTO-100 have shown promising results in alleviating symptoms of MDD with cognitive impairments, offering initial validation of the therapeutic approach.
Weaknesses:
Causality: While associations between BDNF levels and depression/PTSD are established, it remains unclear if reduced BDNF is a cause or consequence of these conditions. Demonstrating causality is challenging and is required for a more robust therapeutic rationale.
Measurement Variability: BDNF measurements in serum or plasma as proxies for brain BDNF activity are not directly reflective of central changes. Moreover, the levels of BDNF can be variable and influenced by several factors, resulting in inconsistencies across studies.
Heterogeneity of Disorders: MDD and PTSD are heterogeneous disorders. It's unclear if BDNF-related therapies would be equally beneficial for all patients or primarily for those with specific subtypes, such as those with prominent cognitive impairments.
Lack of Large-Scale Trials: Most of the evidence comes from animal studies, small-scale clinical trials, or observational studies. Large-scale, randomized, controlled clinical trials are required to rigorously evaluate efficacy and safety.
Complex Role of BDNF: The role of BDNF in the central nervous system is complex and not limited to one pathway or mechanism. This complexity can make it difficult to predict the effects of modulating BDNF on overall brain function and mental health.
Potential Side Effects: There is limited knowledge of the potential long-term side effects of upregulating neurogenesis and plasticity in adult brains, which could be a concern with chronic BDNF-targeted therapies.
In conclusion, the evidence base for BDNF as a target for MDD and PTSD with cognitive impairments presents a compelling theoretical framework supported by a variety of study designs. However, the need for extensive and rigorous clinical trials along with a greater understanding of the complexities and nuances of BDNF biology is apparent to fully establish the therapeutic potential and safety of BDNF modulation.
The Phase 2a clinical study concerning ALTO-100 is an open-label intervention for adults diagnosed with Major Depressive Disorder (MDD) and/or Post-Traumatic Stress Disorder (PTSD). The primary goal is to explore predictors and correlates of clinical outcomes based on baseline biological data while taking ALTO-100.
The study administered a per os (PO) tablet of ALTO-100 twice daily for a duration of eight weeks. The sample size was 245 enrolled participants, and the study extended from December 20, 2021, to December 9, 2022.
It was a single-group assignment with no masking, meaning all participants received the same intervention and were aware of the treatment being provided.
Primary outcomes involved tracking changes over time in depression, general psychopathology, and PTSD severity using established rating scales, such as MADRS, CGI-S, and CAPS-5. The frequency of measuring these outcomes varied, with MADRS and CGI-S being measured five times over the eight weeks, while CAPS-5 was measured three times over the same period. Additionally, the study monitored the safety and tolerability of the drug by recording adverse events (AEs), vital sign abnormalities, and laboratory test abnormalities.
Critiques of the Study Design
Operational or Technical Challenges
In summary, while the study aims to glean important information about the relationship between baseline biology and response to ALTO-100, its open-label, single-group design might limit the strength of its conclusions due to potential biases and lack of a control group. Operational challenges related to data collection and monitoring could also influence the outcomes.
The chosen primary and secondary endpoints in this ALTO-100 study are well-suited for a proof-of-concept study aimed at assessing the therapeutic efficacy in treating Major Depressive Disorder (MDD) with impaired cognition. Using rating scales such as the Montgomery-Åsberg Depression Rating Scale (MADRS), Clinical Global Impression scale - Severity (CGI-S), and the Clinician-Administered PTSD Scale for DSM-5 (CAPS-5) provides a structured approach to evaluating symptom severity and change, which are standard in mental health research.
Primary and Secondary Endpoints Appropriateness:
The inclusion criteria are appropriate for the study:
Potential Reproducibility Challenges Posed by the Inclusion/Exclusion Criteria:
Operational Considerations:
Scientific Reproducibility Challenges:
In conclusion, the proof-of-concept study is outlined with appropriate endpoints for evaluating the efficacy of ALTO-100 in treating MDD with impaired cognition, and the inclusion and exclusion criteria are designed to create a well-defined study population. The potential challenges to reproducibility arise primarily from the subjective assessment measures and variability in participants' psychiatric medication regimens. Ensuring the fidelity of diagnostic procedures, treatment administration, adherence, and outcome assessment will be key to establishing reproducibility and validity in the findings.
The exploratory trial was conducted over 8 weeks with 133 patients who had moderate to severe MDD. A subset of 123 was included in the biomarker analyses.
Initial results from a 30-patient discovery dataset indicated that those with poor verbal memory showed a better response to ALTO-100. Poor verbal memory was associated with reduced hippocampal neuroplasticity, a characteristic of depression, suggesting that ALTO-100 could improve this. A pre-specified statistical analysis of the blinded test data replicated the initial findings that poor verbal memory predicted better clinical outcomes for ALTO-100, irrespective of whether used as monotherapy or adjunct therapy.
A significantly higher response rate (≥50% reduction in MADRS score) was observed in patients with the identified poor verbal memory biomarker at weeks 6 and 8.
Patients with the biomarker had an 81% response rate to ALTO-100 monotherapy at week 8, versus 38% without it.
ALTO-100 adjunctive treatment resulted in a 50% response rate for biomarker-positive patients, compared to 31% for those without. At week 8, patients with the biomarker also showed better outcomes on the Hamilton Depression Rating Scale (HDRS) and the Clinician Global Impression—Severity scale (CGI-S). The predictive value of poor cognition was specific to ALTO-100, not showing predictive value for placebo response or standard-of-care antidepressants.
FDA feedback led to an increased patient target enrollment for the Phase 2b trial to enhance the study's power.
In conclusion, the clinical data suggest that verbal memory impairment is a predictive biomarker for a better response to ALTO-100 in treating MDD. The drug appears effective with a tolerable safety profile, and further studies are being designed to reinforce these findings.
Approvable endpoints for a drug like ALTO-100 in Major Depressive Disorder (MDD) with a specific focus on impaired cognition might include both traditional measures of depression severity and specific measures of cognitive function. In the context of FDA approval, clinically meaningful endpoints that demonstrate a significant impact on both the symptoms of MDD and cognitive impairments are essential. Below are potential endpoints that may be used in clinical studies to support the approval of ALTO-100:
The FDA typically requires Phase 3 pivotal trials to support the approval of a new drug. These studies are larger and more rigorous than early-phase trials and are designed to confirm the drug's efficacy, monitor side effects, and collect more comprehensive safety data. For ALTO-100, the following clinical trials might be expected:
The number of patients required for Phase 3 trials depends on the expected effect size, variability of response, desired power to detect a statistically significant effect, and the trial design:
In conclusion, while definitive numbers depend on the specific statistical design and objectives of each study, Phase 3 trials for ALTO-100 would likely need to recruit several thousand patients in total to meet regulatory requirements for approval successfully.
Major Depressive Disorder (MDD) with impaired cognition, often referred to as "depression with cognitive dysfunction," is a subtype of depression that involves cognitive impairments in addition to the usual symptoms of MDD.
Pathology:
The exact pathology of MDD with impaired cognition is complex and not entirely understood but is thought to involve:
Symptoms:
Prognosis:
The prognosis of MDD with impaired cognition can vary significantly among individuals. If untreated, cognitive symptoms may persist even when mood symptoms improve, potentially leading to long-term impairments in work and social functioning. However, with appropriate treatment, which may include a combination of medication (e.g., antidepressants) and psychotherapy (e.g., cognitive-behavioral therapy), many patients can achieve remission of both mood and cognitive symptoms.
Complicating factors:
Treatment:
It is important to note that this is a general description, and individual patient experiences may vary. Treatment should be personalized, with the input of mental health professionals tailored to the individual's specific symptoms and needs.
Post-traumatic stress disorder (PTSD) with impaired cognition is a mental health condition that can develop after exposure to a traumatic event, such as combat, sexual assault, natural disasters, or other life-threatening or highly distressing experiences. Alongside characteristic PTSD symptoms, individuals may suffer from cognitive impairments that affect their daily functioning and quality of life.
To create a hypothetical revenue build for ALTO-100 in Major Depressive Disorder (MDD) with impaired cognition, we'd need to estimate several critical figures and consider a variety of factors. Please note that the estimates provided below are placeholder values to illustrate the calculations involved in a revenue build and do not reflect actual market data or clinical trial outcomes for ALTO-100.
Given these factors, the hypothetical revenue build could be outlined as follows:
Gross Revenue Calculation:
- TAM: 10,000,000 MDD patients (for instance)
- Prevalence of impaired cognition in MDD: X% = 2,000,000 patients
- Treated population: Y% of 2,000,000 = 1,000,000 patients
- Market Penetration (Z%): 25% of 1,000,000 = 250,000 patients using ALTO-100
- Treatment Cost per Year (A): $9,000, a premium to branded drugs for MDD
- Duration of Therapy (B): 8 weeks (56 days)
- Rebates and Discounts (C%): 30%
- Insurance Coverage (W%): 80%
- Co-pay (V%): 30%
This simplistic model does not consider patient adherence rates and potential dropout rates during therapy, varying insurance plans, eligibility criteria, or differing international drug pricing regulations and market dynamics, which can significantly impact the final revenue figures. Additionally, it also does not factor in the growth and changes in prevalence or MDD diagnosis rates over time.
Full financial modelling would require more detailed and region-specific data. However, this simplified approach gives an initial estimate that can be refined with more data.
To estimate the probability of ALTO-100 moving successfully through Phase 2, Phase 3, and FDA submission, we will use the industry standard clinical trial success rates for Neurology products, in conjunction with the specific Phase 2a trial data for ALTO-100.
Industry Standard Success Rates for Neurology Products:
Analysis of Phase 2a Trial Data for ALTO-100:
From the given data, ALTO-100 has demonstrated effective results in treating MDD with impaired cognition, particularly in patients with poor verbal memory. The trial's primary endpoint was met, with a significant change in the MADRS score. The response rate was notably high at 81% in the specified patient subgroup when treated with ALTO-100 monotherapy. These positive results may suggest that the compound could have a higher chance of success than the industry average for neurology products.
However, drug development inherently carries significant risk, and success in early-stage trials does not always predict outcomes in later stages, which involve larger populations and more stringent assessments. Given this, we can adjust the industry standard probabilities with the optimism gleaned from the trial data but still maintain a conservative stance to account for unforeseeable challenges.
Adjusted Success Probabilities for ALTO-100:
Creating a hypothetical revenue build for ALTO-100 in Post-traumatic stress disorder (PTSD) with impaired cognition involves estimating market size, treatment access, pricing, duration of therapy, and other financial factors. Below are placeholder estimates with consideration of the clinical trial findings for ALTO-100.
Based on these factors, let's derive some hypothetical revenue numbers.
Hypothetical Revenue Calculation:
- Total number of PTSD patients: 1,000,000 (for example)
- Prevalence of impaired cognition in PTSD (X%): 30%
- Diagnosed and treated (Y%): 50%
- Market penetration (Z%): 20%
- Duration of therapy (B weeks): 12 weeks (84 days)
- Insured patients (W%): 85%
- Insurance reimbursement rate (V%): 75%
- Annual treatment duration (D days): 365 days (chronic treatment)
In this simplified model, the hypothetical annual net revenue for ALTO-100 in treating PTSD with impaired cognition is approximately $75.6 million before insurance reimbursements and patient co-pays are factored in.
An actual financial assessment would require more granular data, such as regional variation in prevalence, treatment rates, real-world pricing, payer mix, competitive landscape, patient compliance rates, discontinuation rates, and potential market expansion post-approval. It is also essential to consider the investment in marketing and sales efforts, post-marketing clinical trials, regulatory milestones, and potential competition or generic entrants, all of which can significantly influence revenue.
To estimate the probability of ALTO-100 progressing through the clinical trial phases for PTSD with impaired cognition, we would apply the same industry standard rates as we did for MDD, with adjustments based on the Phase 2a trial data. Given that the data suggest strong support for ALTO-100's efficacy in this particular patient group, we might expect similar or potentially better progression rates than the industry average due to the promising results and the specificity of the biomarker.
However, it’s important to consider that PTSD, although within the realm of neurology, may have different clinical trial dynamics than other conditions due to variables such as variability in patient response based on trauma type and history, heterogeneity of symptoms, and challenges in measuring treatment effects.
The therapeutic rationale for using a MT1/MT2 agonist and 5-HT2C antagonist such as ALTO-300 (agomelatine) in Major Depressive Disorder (MDD) rests on addressing certain symptoms and dysfunctions associated with depression through unique pathways, thus potentially offering advantages over conventional antidepressants.
Clinical trials of agomelatine have indicated it has antidepressant properties with potential benefits that are similar to other common antidepressants, and it has demonstrated tolerability advantages over other antidepressants. This favorable side effect profile is particularly evident in clinical data showing lower incidences of gastrointestinal intolerability, anxiety, sleep disturbance, and sexual dysfunction.
The development of ALTO-300 is also leveraging precision medicine techniques, using a machine learning-derived EEG biomarker profile for identifying patients most likely to benefit from treatment. This EEG biomarker is ALTO-300 specific and allows for the tailored treatment of MDD sub-populations, potentially enhancing efficacy rates and minimizing exposure to those less likely to respond.
Given the unique mechanism of action, which potentially influences sleep regulation and mood through different pathways compared to SSRIs and SNRIs, as well as the predictive biomarker strategy for patient selection, ALTO-300 offers a promising therapeutic rationale for a subset of individuals with MDD.
The scientific rationale is based on the established pharmacological actions of agomelatine and its effects as observed in clinical trials and research studies. Here's a breakdown of how established the science is and the level of evidence supporting these processes:
The role of neurotransmitters in depression is an area of ongoing research. While it is accepted that neurotransmitter imbalance is associated with depression, the precise mechanisms by which neurotransmission influences mood disorders, and how medications produce their effects, remain incompletely understood.
In summary, the basic pharmacological processes described (melatonin receptor agonism and serotonin receptor antagonism) are well-supported by scientific evidence, but the translation of these effects into consistent, predictable clinical outcomes in MDD treatment remains complex. The utilization of EEG biomarkers as predictive tools for treatment response represents an innovative and less established area of research that holds potential but is still being validated in the clinical setting.
Both the role of MT1/MT2 agonists and 5-HT2C antagonists have been explored in the context of Major Depressive Disorder (MDD) and are supported by various studies and clinical trials. Here's some literature supporting their roles:
Regarding clinical effectiveness:
Furthermore, clinical trial meta-analyses offer evidence regarding the efficacy and safety of agomelatine:
In conclusion, the scientific literature indicates that the effects of MT1/MT2 agonists and 5-HT2C antagonists like agomelatine have a plausible biological basis and have been substantiated in both preclinical models and clinical trials. However, as with any pharmacological agent, the exact degree of efficacy and the scope of its optimal use (e.g., as monotherapy or in combination with other antidepressants) in diverse patient populations still presents an area for ongoing research and debate.
The evidence base supporting the therapeutic rationale for MT1/MT2 agonists and 5-HT2C antagonists in the treatment of Major Depressive Disorder (MDD), specifically related to drugs like agomelatine (ALTO-300), has several strengths and weaknesses:
Strengths:
Weaknesses:
The overall therapeutic rationale for agomelatine's dual action is supported by a substantial, though still developing, evidence base. While the data has its limitations, the current level of evidence is considered by many clinicians and researchers to be sufficiently strong to support its use as one of the treatment options for MDD. The ongoing research, including the development and application of predictive biomarkers, should further refine this therapeutic rationale.
Summary of ALTO-300 Study Design for Major Depressive Disorder:
The clinical study with identifier NCT05118750 sponsored by Alto Neuroscience is an interventional Phase 2 trial that seeks to evaluate the effectiveness and safety of the drug ALTO-300 for treating adults with Major Depressive Disorder (MDD). Commencing on December 13, 2021, and completing on May 9, 2023, the study enrolled 91 participants.
This open-label study involves a single-group assignment in which all participants receive the same treatment: ALTO-300 PO tablet taken orally once daily for a duration of 8 weeks. The main goals are to define predictors and correlates of ALTO-300’s effects by examining the relationship between baseline biological parameters and clinical outcomes.
Primary outcome measures include alterations in the severity of depression as measured by the Montgomery-Åsberg Depression Rating Scale (MADRS) and the general psychopathology as measured by the Clinical Global Impression scale - Severity (CGI-S), both assessed at six different time points during the eight-week period. Safety assessments for ALTO-300 involve monitoring the incidence and severity of treatment-emergent adverse events (TEAEs), serious adverse events (SAEs), study discontinuation due to adverse events, deaths, changes in vital signs, and laboratory data including liver function tests.
Critiques of the Study Design:
Operational/Technical Challenges:
In conclusion, while the study aims to provide valuable insights into the treatment of MDD with ALTO-300, several design elements, such as the open-label and single-group format, might present challenges for establishing the efficacy and safety of the drug without bias. Operational challenges must also be managed effectively to ensure high-quality data collection and participant safety.
Potential for Proof-of-Concept:
The ALTO-300 trial is designed to evaluate the antidepressant efficacy and safety of ALTO-300 and to identify biological predictors of response. Given that the primary endpoints include well-established clinical scales (MADRS and CGI-S), and safety parameters, the study has the potential to provide proof-of-concept evidence for the use of ALTO-300 in treating MDD if the results show statistically and clinically significant improvements over the treatment period.
Appropriateness of Primary and Secondary Endpoints:
These are appropriate for determining the clinical impact of the drug on depressive symptoms and the general state of the disorder.
Safety evaluations are crucial secondary endpoints to ensure that any efficacy is not overshadowed by unacceptable risks or adverse effects associated with ALTO-300.
Inclusion / Exclusion Criteria:
The inclusion criteria are focused on individuals with a clear and well-established diagnosis of MDD. The requirement of a stable baseline on current antidepressant medications, with a specific response history, aims to select participants who are not experiencing sufficient relief from their current regimen. The inclusion of biomarker assessments broadens the study to investigate potential predictors of response to treatment.
Exclusion criteria are comprehensive, aiming to protect patients with certain medical conditions or medication regimes that could confound results, introduce additional risks, or interfere with the mechanism of action of ALTO-300. The exclusion of individuals with a history of bipolar or psychotic disorder avoids the inclusion of individuals with potentially different underlying pathophysiologies.
Reproducibility Challenges:
In conclusion, while the study design includes appropriate endpoints and thoughtful inclusion/exclusion criteria for assessing the efficacy and safety of ALTO-300 in a specific subset of patients with MDD, these details could also pose challenges to reproducibility and the generalizability of study results. Ensuring that the trial's findings are widely applicable and replicable in diverse patient populations will be essential for further establishing the utility of ALTO-300 in a broader clinical setting.
The clinical data for ALTO-300 in Major Depressive Disorder (MDD) are derived from a completed Phase 2a clinical trial. The main findings can be summarized as follows:
In conclusion, the clinical data from the Phase 2a trial suggest that ALTO-300 has the potential to be effective as an adjunctive treatment for MDD, particularly in patients identified by a specific EEG biomarker profile. The treatment was generally safe and well tolerated.
For ALTO-300 in Major Depressive Disorder (MDD), potential approvable endpoints for regulatory agencies such as the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA) usually revolve around the safety and efficacy profile of the drug being tested. These endpoints are often derived from the symptoms' improvement scale scores and overall impact on patients' quality of life. Here are some commonly accepted potential endpoints:
Given that ALTO-300 is proposed as an adjunctive therapy for MDD, the trials might compare ALTO-300 in combination with a standard antidepressant versus placebo in combination with a standard antidepressant. This design better reflects the intended use of the drug in practice.
The number of patients required for these studies will depend on several factors, including the expected effect size, variability of response, acceptable alpha and beta error rates (Type I and II errors), and potential dropout rates. Drug developers often consult with statistical experts and may conduct a sample size calculation, considering the effect sizes seen in Phase 2 trials. For major depressive disorder, Phase 3 trials typically recruit anywhere from a few hundred to several thousand patients per treatment arm.
Considering ALTO-300 has shown efficacy in a patient population characterized by an EEG biomarker, confirmatory trials may focus on this subgroup, which could potentially lower the number of patients required if the effect size is large and consistent.
Ultimately, the design and size of the pivotal trials for ALTO-300 will be subject to discussions with regulatory authorities and will be influenced by the outcomes of earlier phase trials, current medical standards, the drug's mechanism of action, and the competitive landscape.
To create a hypothetical revenue build for ALTO-300 in Major Depressive Disorder (MDD), we will need to make several assumptions. Please note that all figures and calculations provided here are placeholders and hypothetical estimates, and should be adjusted based on real-world data as it becomes available. The process involves estimating the number of patients, treatment duration, pricing, market penetration, and adjustments for discounts and insurance coverage.
Here is a step-by-step build:
Annual Revenue Estimate = (Treatable Population × Market Penetration × Insurance Coverage × (Average Duration of Therapy in Months/12) × Drug Price after Gross-to-Net Adjustments)
Let's build an example with hypothetical numbers:
This model is extremely simplified and should include growth rates, patient discontinuation, competition effects, market expansion due to increased diagnostic rates or approval in additional indications, international market scaling, possible patent cliffs, and more. It's essential to update these estimates with real-world data, expert opinion, and additional market research as the information becomes available. Additionally, the use of data analytics and forecasting models can help refine revenue projections over time.
To estimate the probabilities of success at each stage of clinical development for ALTO-300 in Major Depressive Disorder (MDD), we can use the industry standard success rates for neurology products. However, it's important to note that these rates are general and can vary based on the therapeutic area, the specific drug profile, and other factors specific to the clinical development program. Here's how we would estimate the probabilities given the industry standard rates:
These probabilities are multiplicative when considering the overall likelihood of approval from the current stage. Therefore, the overall estimated probability of ALTO-300's success from the end of Phase 2 to approval would be the product of each stage's success rate:
The therapeutic rationale for using a PDE4 inhibitor like ALTO-101 in Cognitive Impairment Associated with Schizophrenia (CIAS) is rooted in the crucial role of the cyclic adenosine monophosphate (cAMP) signaling pathway in cognitive functions and neuroplasticity.
CIAS is a challenging aspect of schizophrenia characterized by deficits in memory, attention, executive function, and various other cognitive domains, which significantly impact patient quality of life and functional outcomes. Current treatments for schizophrenia predominantly target the positive symptoms (hallucinations, delusions), with a notable gap in efficacious treatments for cognitive and negative symptoms.
Phosphodiesterase 4 (PDE4) is an enzyme that degrades cAMP, a ubiquitous second messenger involved in a range of cellular functions, including neuronal signaling. The cAMP pathway is known to have a significant role in the modulation of immune and inflammatory responses, learning, memory, and mood regulation. In patients with schizophrenia and other cognitive disorders, there has been a documented reduction in neuroplasticity-related signaling pathways, including the cAMP pathway. This dysregulation is thought to contribute to the cognitive deficits observed in such conditions.
As a PDE4 inhibitor, ALTO-101 aims to prevent the breakdown of cAMP, thereby increasing its levels in the brain. Elevating cAMP levels is believed to enhance neuroplasticity, which is vital for learning and memory processes. This is especially relevant in the hippocampus, a brain region integral to memory formation and cognitive function. In preclinical models, increasing hippocampal cAMP has been shown to improve various forms of memory and provide pro-cognitive effects.
Early phase studies of ALTO-101 have demonstrated that it can enter the human brain and is well-tolerated. Additionally, preliminary results suggest that ALTO-101 may have robust effects on cognitive processing and performance as measured by EEG and cognitive tests. These promising findings provide a strong basis for advancing the clinical development of ALTO-101 into Phase 2 proof-of-concept trials in patients with CIAS.
In summary, the use of ALTO-101 in CIAS capitalizes on the understanding that enhancing cAMP signaling through PDE4 inhibition may correct underlying neurochemical deficits associated with cognitive impairments in schizophrenia, potentially providing a novel therapeutic avenue to address these unmet clinical needs.
The science underlying the use of PDE4 inhibitors for cognitive enhancement is based on a wealth of preclinical evidence; however, its translation into clinical practice is still an area of active research and development.
The understanding that cAMP signaling pathways play a significant role in cognitive functions and neuroplasticity is well established. cAMP is a critical second messenger in numerous biological processes, and its role in the brain includes the regulation of gene transcription, neuronal excitability, synaptic plasticity, and memory formation. Specifically, the involvement of cAMP signaling in learning and memory has been documented across various types of animal models.
Furthermore, post-mortem and genetic studies indicating alterations in cAMP signaling in individuals with cognitive disorders provide a compelling rationale for targeting this pathway to improve cognitive deficits. PDE4, being one of the principal enzymes regulating cAMP levels by catalyzing its breakdown, emerges as a plausible target for pharmacological intervention.
However, here are some aspects that are subject to uncertainty or debate:
The overall level of evidence suggests that the theoretical basis for using PDE4 inhibitors such as ALTO-101 for cognitive enhancement is strong due to a well-documented role of the cAMP pathway in cognition and neuroplasticity. The early clinical data on safety and brain penetration of ALTO-101 add to the support. However, the clinical efficacy, particularly in Phase 2 and 3 trials, will be the ultimate test of the validity of this therapeutic approach.
The leap from proof of concept in early-phase trials to broader clinical application will require addressing the above uncertainties and will depend on a balance between efficacy, safety, and tolerability. Only with the completion of more extensive trials can the place of PDE4 inhibitors in the treatment of CIAS be fully determined.
Research into the PDE4 enzyme's role in cognitive impairment, including its association with schizophrenia, is supported by numerous preclinical studies and some clinical investigations, though it remains an emerging field. Here's a synthesis of key points from the literature:
In summary, while there is compelling evidence from preclinical studies indicating that PDE4 plays a role in cognition and could be a therapeutic target for cognitive impairment, clinical validation of PDE4 inhibitors for the treatment of CIAS is still in progress. The ongoing and future clinical trials will be paramount in confirming whether PDE4 inhibition can translate into a clinically meaningful improvement in cognitive impairments associated with schizophrenia.
The therapeutic rationale for targeting PDE4 in Cognitive Impairment Associated with Schizophrenia (CIAS) is built on a mixture of mechanistic insights, preclinical data, and early clinical results. Here is an evaluation of the strengths and weaknesses of this evidence base:
Strengths:
Weaknesses:
In conclusion, while the rationale for using a PDE4 inhibitor to treat CIAS is underpinned by a strong scientific hypothesis and promising early trial data, there remain substantial gaps that need to be addressed through rigorous clinical research. The path from bench to bedside is particularly fraught in psychiatric drug development, and the substantial evidence base from preclinical models will need strong clinical trial results to be considered robust.
The clinical data for ALTO-101 in the context of cognitive impairment associated with schizophrenia (CIAS) is derived from several Phase 1 trial outcomes as follows:
In summary, the Phase 1 clinical data provides evidence that ALTO-101 may have pro-cognitive effects in CIAS and is generally well-tolerated. The data also led to improvements in drug delivery, with the development of a transdermal formulation to enhance tolerability and treatment experience for patients.
Applicable endpoints for clinical trials in Cognitive Impairment Associated with Schizophrenia (CIAS) typically focus on cognitive function, safety, tolerability, and overall clinical effectiveness of the interventional drug. The U.S. Food and Drug Administration (FDA) provides guidance on endpoints and study design for drugs targeting CIAS. The following are possible approvable endpoints and outlines of clinical studies for ALTO-101 development:
In summary, a typical clinical development plan for ALTO-101 in CIAS would include Phase 2 and Phase 3 studies, assessing both cognitive outcomes using standard batteries and safety/tolerability measures, with the latter studies enrolling hundreds to thousands of patients. The precise design of these studies, including choice of endpoints and determination of sample size, would likely evolve in consultation with regulatory agencies and as more data are collected from earlier-phase trials.
Cognitive impairment is a core feature of schizophrenia, a chronic and severe mental disorder characterized by a range of symptoms that can affect thinking, behavior, emotions, and perception of reality. Cognitive deficits often emerge before the onset of hallmark psychotic symptoms such as hallucinations and delusions and are among the most disabling features of the illness, impacting daily functioning and quality of life.
While the exact pathophysiology of cognitive impairment in schizophrenia is not fully understood, it is believed to involve dysregulation of various neurotransmitter systems, notably dopamine, glutamate, and GABA. There is also evidence of structural brain changes, including reductions in gray matter volume, particularly in the frontal and temporal cortices, and deficits in white matter integrity. These alterations are thought to lead to disruptions in neural circuitry that underlie cognitive processes.
Cognitive deficits in schizophrenia encompass a broad range of domains, including:
These cognitive deficits can occur independently of the psychotic symptoms and often persist even when other symptoms are managed with medication.
Cognitive impairment in schizophrenia tends to be stable over time, and in some cases, it may worsen with the progression of the disease. These deficits are typically less responsive to antipsychotic medications than the psychotic symptoms. As a result, they are a major determinant of long-term disability and a critical target for interventions aimed at improving overall prognosis.
The management of cognitive impairment in schizophrenia includes pharmacological and non-pharmacological approaches:
Research continues to search for more effective treatments for cognitive deficits in schizophrenia. Investigations range from pharmacological approaches including novel compounds, combination therapies, and personalized medicine strategies based on genetic profiles, to advanced neurostimulation techniques like transcranial magnetic stimulation (TMS).
In summary, cognitive impairment in schizophrenia is a critical determinant of functional outcomes and remains a major area of unmet need. Understanding and addressing these deficits is key to improving the prognosis and enhancing the quality of life for individuals living with schizophrenia.
To provide an analysis of the market opportunity for a drug like ALTO-101 for Cognitive Impairment Associated with Schizophrenia (CIAS), it is essential to examine the competitive landscape, current standard of care, and unmet medical needs within this indication.
There are currently no drugs approved by the U.S. Food and Drug Administration (FDA) specifically for CIAS, though some currently approved antipsychotic medications may have mild cognitive benefits for patients. Medications such as donepezil (a cholinesterase inhibitor used for Alzheimer's disease) have been studied in schizophrenia with mixed results. Hence, the competition for a new effective drug in this domain would likely be sparse, presenting a prime opportunity if ALTO-101 shows significant efficacy.
The current standard of care for schizophrenia includes atypical antipsychotics, with adjunctive treatment options like cognitive behavioral therapy and cognitive remediation therapy for cognitive deficits. However, these do not fully address the cognitive impairments, thus leaving a significant gap in treatment options.
There is an unmet medical need due to:
The global antipsychotic drug market is a multi-billion dollar industry, and a targeted cognitive-enhancing drug like ALTO-101 would likely address a sizeable subset of this market. Considering the prevalence of schizophrenia (approximately 1% of the population worldwide) and the fact that most individuals with the disorder exhibit cognitive deficits, the target market for an effective CIAS treatment would be extensive.
There are several companies working on developing treatments for cognitive impairment associated with schizophrenia (CIAS) that may compete with ALTO-101. These organizations range from large pharmaceutical companies to smaller biotech firms and are investigating various pharmacological approaches.
Generating a hypothetical revenue build for ALTO-101 in Cognitive Impairment Associated with Schizophrenia (CIAS) involves making various assumptions and providing placeholder estimates for the number of patients treated, pricing, insurance coverage, etc. Below is a revenue build model with sample assumptions and calculations:
Keep in mind that the actual revenue will be influenced by many factors including successful completion of clinical trials, regulatory approval, competition, marketing effectiveness, and continued clinical and real-world evidence of efficacy and safety. The figures used in this example are purely hypothetical and intended to illustrate the process of constructing a revenue build; true revenue forecasting would require more specific data and complex models.
To estimate the probability of clinical success at various phases for ALTO-101 in Cognitive Impairment Associated with Schizophrenia (CIAS), we'll use the industry-standard success rates and factor in the positive preclinical data for ALTO-101.
Given industry averages for Neurology products:
The promising results from ALTO-101's Phase 1 trials suggest that it might perform at least as well as the industry averages. However, to be conservative, we will not adjust these probabilities upward, as clinical trial success can often be unpredictable, and positive Phase 1 results do not guarantee success in later phases.
ALTO-203 is a novel small molecule histamine H3 receptor inverse agonist being developed as a treatment for Major Depressive Disorder (MDD) with a particular focus on anhedonia, which is a symptom characterized by a loss of pleasure or lack of interest in normally rewarding activities.
Therapeutic Rationale:
Taken together, the rationale for pursuing ALTO-203 as a treatment for MDD with anhedonia is to address the underlying neurobiological dysfunction by increasing dopamine release in the brain's reward pathway, thus potentially improving anhedonic symptoms and overall mood. The distinct mechanism of action differentiates it from other treatments and provides a novel approach to managing this challenging aspect of depressive disorders.
The science behind the therapeutic rationale for ALTO-203 in treating Major Depressive Disorder (MDD) with anhedonia involves several well-established as well as emerging concepts in neuroscience and psychopharmacology. Here's a breakdown of the established elements and those areas that may still be under debate or require further research:
Established Science:
Areas of Uncertainty or Debate:
Level of Evidence: The overall level of evidence supporting the processes described involves a combination of preclinical studies, early-stage clinical trials, and pharmacological knowledge. The preclinical evidence showing that ALTO-203 increases dopamine release in the nucleus accumbens is a promising sign for its therapeutic potential. However, higher-quality evidence from randomized controlled trials in humans is essential to confirm its efficacy and safety in treating MDD with anhedonia.
In summary, while the neuropharmacological rationale for ALTO-203 as a treatment for anhedonia in MDD is grounded in an understanding of H3 receptors and dopaminergic signaling, the clinical application of this knowledge is still at a relatively early stage. The science is promising but must be validated through rigorous clinical testing to resolve the uncertainties and scientific debate surrounding the efficacy of H3 inverse agonists in this context.
As of my knowledge cutoff in early 2023, while the histamine H3 receptor (H3R) is an established target for certain central nervous system (CNS) disorders, its role in Major Depressive Disorder (MDD) with anhedonia is more emergent and subject to ongoing research. The H3 receptor functions predominantly as a presynaptic auto- and heteroreceptor in the brain, modulating the release of various neurotransmitters, including histamine, acetylcholine, norepinephrine, and notably, dopamine. Here are a few insights supported by the scientific literature on the potential role of the H3 receptor in MDD and anhedonia:
It's important to note that much of the available data stem from preclinical studies, which necessitates cautious interpretation when translating these findings to human conditions. Additionally, the H3 receptor inverse agonists' effects on neurotransmitter release in brain regions associated with reward processing do not yet have a robust body of clinical data to conclusively establish their efficacy for MDD with anhedonia.
When it comes to systematic reviews, meta-analyses, or large-scale clinical studies focused on H3 receptor inverse agonists for anhedonia in MDD, the literature is not extensive as of yet. The developmental stage of novel drugs targeting this mechanism, such as ALTO-203, suggests that more definitive evidence will emerge from ongoing and future Phase 2 and Phase 3 clinical trials.
In summary, scientific interest in the role of H3 receptors in MDD with anhedonia is grounded in preclinical evidence and pharmacological theory, but there is a significant gap in clinical evidence that needs to be filled to confirm the therapeutic potential of H3 receptor inverse agonists in this specific patient population. As such, research results anticipated in the coming years will be crucial for establishing the clinical relevance of this approach.
The evidence base supporting the therapeutic rationale for using H3 receptor inverse agonists in treating Major Depressive Disorder (MDD) with anhedonia can be evaluated in terms of its strengths and weaknesses:
Strengths of the Evidence Base:
Weaknesses of the Evidence Base:
The level of evidence supporting the therapeutic rationale for H3 receptor inverse agonists in MDD with anhedonia is currently at a stage that suggests promise but requires more empirical validation. The enthusiasm drawn from preclinical data and pharmacological theory needs to be balanced with rigorous clinical trial data to establish efficacy, safety, and the scope of the potential therapeutic benefits for patients with MDD and anhedonia. As such, future research outcomes will be critical in determining the viability of this therapeutic strategy.
The clinical data supporting the use of ALTO-203 for Major Depressive Disorder with anhedonia are derived from Phase 1 trials originally conducted by Cephalon and Teva prior to the licensing of ALTO-203 by the current developer. The following points summarize the clinical data available:
Safety and Tolerability: ALTO-203 was evaluated in three Phase 1 clinical studies involving healthy subjects. The originator of ALTO-203 conducted these studies between 2009 and 2014, which were aimed at assessing safety and pharmacokinetics. In these trials, ALTO-203 was well tolerated by the participants. The first study tested single doses ranging from 0.02 mg up to 5.0 mg in 48 healthy subjects. The second study further assessed safety and dosing, including single and multiple doses from 0.02 mg up to 0.5 mg over nine days in 48 healthy subjects.
Predictable Pharmacokinetics: Across the Phase 1 trials, ALTO-203 exhibited predictable pharmacokinetics, which is critical for determining appropriate dosing regimens.
Efficacy Indicators: In one of the trials that used a cross-over design with 40 healthy individuals, ALTO-203 at a single dose of 25µg showed a positive effect on subjective emotion. This was measured using the Bond and Lader scale components that evaluate alertness and mood, indicating that ALTO-203 may have a beneficial impact on emotional states.
Comparison to Controls: The same trial included two active control arms, modafinil (a dopaminergic agent) and donepezil (a cholinergic agent), which allowed for comparison of the effects of ALTO-203. The placebo-adjusted differences in effects on mood and alertness were similar to or greater than those achieved by modafinil, whereas donepezil did not show a significant deviation from placebo.
Additional Cognitive Findings: Besides subjective emotional improvements, additional data from the cross-over trial indicated that ALTO-203 improved reaction time and adaptive eye tracking in participants, suggesting potential cognitive benefits.
Informed Development Plan: The current developer has used the data from the Phase 1 trials to inform the clinical development plan for ALTO-203, particularly focusing on dosing, tolerability, and acute pharmacodynamic effects as they pursue treatment options for Major Depressive Disorder with anhedonia.
Overall, the clinical data indicate that ALTO-203 is well tolerated and may have beneficial effects on mood and cognitive components relevant to Major Depressive Disorder with anhedonia. However, these findings are preliminary, based on Phase 1 trials in healthy volunteers, and further research is needed to determine the efficacy and safety of ALTO-203 in patients with Major Depressive Disorder.
The approvable endpoints for a novel treatment like ALTO-203 in Major Depressive Disorder (MDD) with anhedonia would typically need to align with regulatory agency requirements such as those from the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA). Anhedonia, which is the loss of interest or pleasure in normally rewarding activities, is a significant symptom of MDD and represents a particular area of unmet medical need. As such, endpoints would need to measure both the severity of the depression and the degree of anhedonia.
Possible approvable endpoints for ALTO-203 could include:
Clinical studies that would typically be required:
The estimated number of patients required:
For Phase 2 trials, it's common to see anywhere from 100 to 300 patients, depending on the design and objectives. For Phase 3, the number of patients is typically in the range of 300 to 3,000, but this can vary based on statistical power calculations tailored to the expected effect size, variability in response, and the dropout rate, among other factors. It's also common for regulators to require two positive Phase 3 studies for approval, each having adequate sample sizes to ensure the statistical robustness of the data.
In summary, the clinical development of ALTO-203 for MDD with anhedonia will need to incorporate specialized scales that measure both depressive symptoms and specific aspects of anhedonia, involve both intermediate-sized Phase 2 and large Phase 3 trials, and ensure sustained follow-up of patients to track long-term safety and efficacy. The exact number of required patients would be determined by statistical power calculations that would take into account the size of the expected treatment effect and the variability in the patient population.
Existing Treatments and Standard of Care:
Currently, the standard of care for MDD includes selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), atypical antidepressants, tricyclic antidepressants (TCAs), and monoamine oxidase inhibitors (MAOIs). Additionally, psychotherapy, lifestyle changes, and other supportive treatments are used.
Medications like SSRIs (e.g., fluoxetine, sertraline) and SNRIs (e.g., venlafaxine, duloxetine) are often first-line treatments. However, the response rates for these drugs can vary, and a significant number of patients do not achieve full remission. Furthermore, existing treatments might not adequately address anhedonia, which is linked to decreased life satisfaction and persistent functional impairments.
Unmet Medical Need:
There is a critical unmet medical need for therapies that directly target anhedonia within the broader context of MDD. Patients with anhedonia often have a poorer response to conventional antidepressants, and this symptom can be a predictor of overall less favorable outcomes. Thus, any therapeutic development that effectively addresses anhedonia would fill a significant treatment gap and potentially improve the prognosis for patients with MDD.
Given the hypothetical scenario for ALTO-203 and the provided clinical background, I'll create a high-level, generalized revenue build for the drug in the Major Depressive Disorder (MDD) with anhedonia space. Please note that these figures will be placeholder estimates and should be refined with more detailed market research, pricing analysis, and input from clinical development.
Target Population Estimation:These projections will need to be regularly updated due to the dynamic nature of the pharmaceutical market, regulatory changes, competitive landscape shifts, and emerging clinical data.
Example Placeholder Estimates:(These are illustrative examples and would require a solid data foundation to be considered actionable)
It's crucial to closely monitor the data from upcoming clinical trials, competitor activity, and market trends to provide continual refinements to these placeholder estimates.
The therapeutic rationale for using a GluN2B-NMDA receptor antagonist like ALTO-202 in Major Depressive Disorder (MDD) stems from several key points related to the pathophysiology of the disease and the pharmacological action of such antagonists:
In summary, the concept of using a GluN2B-NMDA receptor antagonist like ALTO-202 is founded on the neuropathological evidence of glutamatergic dysfunction in MDD, the successful application of other NMDA antagonists as antidepressants, and the potential for rapid-onset action while maintaining a tolerable safety profile. Further well-powered studies are required to fully determine the efficacy, dosage, and safety parameters for the clinical use of ALTO-202 in treating MDD.
The science behind the role of the glutamatergic system in Depression and the use of NMDA receptor antagonists like ketamine for treatment are fairly established, yet several aspects remain under continued research and debate:
In terms of overall evidence, the foundation of the Dysregulated glutamatergic system's role in MDD and the general mechanism of action of NMDA receptor antagonists are supported by substantial preclinical and clinical data. However, the evidence supporting the advantages of specifically targeting the GluN2B subunit, and the specific effects of ALTO-202, is less robust and requires further validation. Upcoming trials will be critical for establishing the clinical utility of ALTO-202, defining its efficacy, and confirming its safety profile. These will contribute to the growing body of evidence on the role of glutamatergic modulation in MDD treatment.
The literature supporting the role of GluN2B-NMDA receptors in Major Depressive Disorder (MDD) emanates from various sources, including preclinical studies, neuroimaging research, and clinical trials of GluN2B-NMDA receptor antagonists.
While these findings are compelling, it's critical to note that research into the selective blockade of the GluN2B subunit is not as well established as the research into broader NMDA receptor antagonism. Moreover, many of the published studies thus far have been early-phase clinical trials, small in scale, or preclinical research. Larger, well-designed clinical trials are necessary to definitively confirm the specific role of GluN2B-NMDA receptors in MDD and establish a solid evidence base for GluN2B selective antagonists as effective therapeutic agents for the disorder.
The therapeutic rationale for the use of GluN2B-NMDA receptor antagonists in Major Depressive Disorder (MDD) is supported by the convergence of evidence from multiple research avenues. Nonetheless, the evidence base has strengths and weaknesses.
Strengths:
Weaknesses:
In conclusion, the evidence base provides a compelling rationale for the continued investigation of GluN2B-NMDA receptor antagonists in treating MDD. However, it also highlights the need for more rigorous clinical research to overcome the current weaknesses in the evidence and establish these antagonists as a standard part of depression therapeutics.
When performing a hypothetical revenue build for a pharmaceutical product like ALTO-202 intended for the treatment of Major Depressive Disorder (MDD), there are key factors to consider. To construct this revenue build, we need to make some assumptions and consider several market variables, including but not limited to the prevalence of the condition, addressable patient population, treatment penetration rates, pricing, duration of treatment, and payer mix.
Assumptions:
Alto Neuroscience's scientific strategy centers on the development of personalized treatments in neuropsychiatric care by leveraging advances in our understanding of neurobiology and brain-based biomarkers. Their approach seeks to align medication to patient groups based on predictive biomarker signatures, which could improve the effectiveness of treatments for conditions such as major depressive disorder (MDD) and beyond.
Here's an overview of their strategy:
Comparing Alto Neuroscience’s strategy with the precision medicine advances in oncology reveals similarities in the use of targeted therapies based on patient-specific biological markers. However, the application of such a strategy is more nascent in neuropsychiatry.
Risks and Pitfalls:
Overall, while Alto Neuroscience's precision psychiatry approach has the potential to revolutionize the treatment of neuropsychiatric disorders, it also faces significant scientific, clinical, regulatory, and commercial challenges that will need to be addressed as development proceeds.
Analyzing performance of Series A VCs
Valuations of biotech startups from Series A to IPO
Bay Bridge Bio Startup Database
How to value biotech companies
Then consider joining our mailing list. I periodically publish data-driven articles on the biotech startup and VC world.