Vico Therapeutics investment analysis

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.



Overview


Vico Therapeutics, a clinical-stage biotechnology company, focuses on the development of genetic medicine therapies for severe neurological disorders. The company's lead product candidate, VO659, an antisense oligonucleotide (ASO) therapy, is designed to address genetic anomalies causing polyglutamine diseases, including spinocerebellar ataxias types 3 and 1 (SCA3 and SCA1) and Huntington's disease (HD). These conditions, characterized by progressive motor control loss and other neurological functions, currently lack disease-modifying treatments.

VO659 has entered a multi-center Phase 1/2a clinical trial that began dosing in April 2023. The trial assesses the drug's safety and tolerability by administering multiple ascending doses intrathecally to patients with mild to moderate SCA3, SCA1, and early-manifest HD symptoms.

In January 2024, the company raised a $60 million (€54 million) Series B financing to support the trial and facilitate the expansion of Vico's therapeutic pipeline. The investment consortium includes lead investor Ackermans & van Haaren, Droia Ventures, EQT Life Sciences, Kurma Partners, and continued support from Polaris Partners, Pureos Bioventures, and Eurazeo.


Pipeline overview


Product nameModalityTargetIndicationDiscoveryPreclinicalPhase 1Phase 2Phase 3FDA submissionCommercial
VO659 Antisense oligonucleotide CAG repeat expansion Translation inhibitor Spinocerebellar Ataxia type 3







VO659 Antisense oligonucleotide CAG repeat expansion Translation inhibitor Huntington"s disease







VO659 Antisense oligonucleotide CAG repeat expansion Translation inhibitor Spinocerebellar Ataxia type 1







Rett syndrome Antisense oligonucleotide-mediated RNA editing Mecp2-R255X RNA editing Rett syndrome



Familial Alzheimer’s disease (FAD) Antisense oligonucleotide PSEN1 Degradation Familial Alzheimer’s disease (FAD)




Highlights and risks


Highlights

Targeting diseases with significant unmet need

Plausible scientific rationale of using ASOs targeting CAG repeats

Preclinical models support CAG repeat targeting and clinical success with ASOs for other neurological conditions supports ASO approach

Risks

Clinical development of Huntington's Disease therapies is challenging

Long-term efficacy and safety of ASO treatment for neurodegenerative diseases are not fully established

Achieving allele specificity, adequate blood-brain barrier penetration, and minimizing off-target effects are challenging


Valuation


We did not perform a valuation analysis due to the early-stage nature of the company.



Weekly analyses of biotech startups, generated by AI

Receive high quality, AI-generated analyses of biotech startups, public companies and scientific papers each week.


VO659


Scientific thesis


Spinocerebellar Ataxia type 3 (SCA3) and Huntington's disease (HD) as well as Spinocerebellar Ataxia type 1 (SCA1) are a group of hereditary neurodegenerative disorders each caused by a CAG repeat expansion mutation within the affected genes. These diseases share a mutation mechanism characterized by an abnormal elongation of a DNA sequence consisting of a triplet repeat of cytosine, adenine, and guanine (CAG) within their respective disease-causing genes. In SCA3/HD/SCA1, this CAG repeat encodes a polyglutamine (polyQ) tract in the respective proteins ataxin-3 (ATXN3), huntingtin (HTT), and ataxin-1 (ATXN1), leading to protein misfolding and aggregation, which is toxic to neurons.

The rationale behind developing allele-preferential antisense oligonucleotides (ASOs) to target these diseases is to selectively reduce the levels of the mutant proteins without affecting the normal counterparts. Here's the therapeutic rationale broken down:

  1. Allele-Preferential Targeting: Since these disorders are autosomal dominant, affected individuals carry both a normal and an expanded mutant allele. Selectively inhibiting the expression of the mutant allele while preserving the expression of the normal allele is crucial for maintaining the protein's natural function in the cell. Allele-preferential ASOs are designed to bind selectively to RNA transcribed from the mutant allele, recognizing the expanded CAG repeats or other sequence variations linked to the pathogenic allele.

  2. Antisense Oligonucleotides: ASOs are short, synthetic strands of nucleotides designed to bind to RNA transcripts through complementary base pairing. Once bound, they can modulate RNA function or processing. They can either promote the degradation of the RNA via RNase-H-mediated cleavage or block the translation of the RNA into protein, depending on their design and the RNase-H recruitment capability.

  3. CAG Repeat Expansion: The expanded CAG repeats create an elongated polyQ tract in the protein, which makes the protein prone to misfolding and aggregation. ASOs that selectively knock down transcripts containing the expanded CAG repeats can reduce the production of these toxic proteins.

  4. Disease Mechanism: By reducing the levels of the mutant protein, it is anticipated that the pathological features such as protein aggregation, neuronal toxicity, and neurodegeneration will be mitigated. This can lead to an amelioration of symptoms or a slowing of disease progression.

In summary, the therapeutic rationale for designing allele-preferential ASOs targeting CAG repeat expansions in diseases like SCA3, HD, and SCA1 is to reduce the toxic burden of the mutant protein on neurons while maintaining the expression of the functional protein from the normal allele, aiming to prevent or slow down the disease progression while minimizing potential side effects related to the suppression of the normal protein. The specificity of ASOs for the mutant allele and their ability to cross the blood-brain barrier make them promising therapeutic agents for these neurodegenerative diseases. Clinical trials are required to determine their safety and efficacy in human patients.

The scientific rationale for using allele-preferential antisense oligonucleotides (ASOs) to target CAG repeat expansions in conditions such as Spinocerebellar Ataxia type 3 (SCA3), Huntington's disease (HD), and Spinocerebellar Ataxia type 1 (SCA1) is based on a substantial body of preclinical evidence. However, as with any therapeutic strategy, especially in the complex field of neurodegeneration, there are still areas of uncertainty and ongoing debate.

Established Science:

  1. Genetics of CAG Repeat Disorders: The mechanism of CAG repeat expansion and its association with neurodegenerative diseases like SCA3, HD, and SCA1 is well established. It is known that these expanded repeats result in the production of toxic proteins that lead to cell dysfunction and death.

  2. Antisense Oligonucleotide Mechanism: The mechanism of ASOs is well understood. ASOs have been studied for several decades, and their ability to target specific RNA sequences to suppress gene expression has been well documented.

Areas of Uncertainty and Debate:

  1. Allele Specificity: While ASOs can be designed to target specific sequences, ensuring that they only target the mutant allele without affecting the normal allele is a significant challenge. Achieving high specificity requires a detailed understanding of the genetic differences between the normal and mutant alleles beyond the CAG repeat expansion, which might include single-nucleotide polymorphisms (SNPs) or other structural variations. The degree of specificity can vary and is a topic of current research.

  2. Delivery to the Central Nervous System (CNS): Effective delivery of ASOs to the CNS is a critical and non-trivial challenge due to the presence of the blood-brain barrier. While some ASOs are designed to cross this barrier, their distribution, uptake, and persistence in various brain regions are areas of active investigation.

  3. Long-term Efficacy and Safety: The long-term efficacy and safety of ASO treatments in humans, particularly for neurodegenerative diseases, are not yet fully established and are subject to ongoing clinical trials.

  4. Off-Target Effects: ASOs can have off-target effects, leading to unintended gene silencing or immune reactions. There is ongoing research into the optimization of ASOs to minimize these effects.

Preclinical studies using ASOs in models of SCA3, HD, and SCA1 have shown promising results, with evidence of decreased mutant protein levels and associated phenotypic improvements. However, the transition from preclinical models to effective treatments in human patients presents many challenges.

As for clinical evidence, there are a few ASOs that have been approved by regulatory agencies for other genetic conditions, which lends credibility to the approach. However, in the context of SCA3, HD, and SCA1, clinical trials are still underway to determine the efficacy and safety of ASOs targeting CAG repeat expansions. Current phase I and II trials are assessing various aspects such as dosing, safety, tolerability, and early signs of efficacy.

In conclusion, while there is a robust scientific foundation supporting the development of allele-preferential ASOs for CAG repeat disorders, their use as a therapeutic approach in clinical settings for these particular diseases remains in the investigational stage. There is high potential, but also significant work to be done before these treatments become standard clinical practice. The ongoing clinical trials will provide crucial data on efficacy, safety, and practical implementation of this therapeutic strategy.


CAG repeat expansions


CAG repeat expansions have been well-characterized in the literature as pathogenic mechanisms for several neurodegenerative disorders, including Spinocerebellar Ataxia type 3 (SCA3), Huntington's disease (HD), and Spinocerebellar Ataxia type 1 (SCA1). Below are some of the key findings from the literature that support the role of CAG repeat expansions in these diseases:

Spinocerebellar Ataxia Type 3 (SCA3):- The causative gene for SCA3 was identified as ATXN3, which encodes for the protein ataxin-3. The mutation responsible for SCA3 involves an expanded CAG trinucleotide repeat in the ATXN3 gene, resulting in an abnormally long polyglutamine (polyQ) tract. The reference for this discovery is: Kawaguchi Y, Okamoto T, Taniwaki M, et al. CAG expansions in a novel gene for Machado-Joseph disease at chromosome 14q32.1. Nat Genet. 1994;8(3):221-228. doi:10.1038/ng1194-221

Huntington's Disease (HD):- Huntington’s disease was one of the first disorders for which an expanded CAG repeat was identified as the cause. The expansion occurs in the HTT gene, and the length of the CAG repeat is inversely correlated with the age of onset of the disease. A foundational paper is: The Huntington's Disease Collaborative Research Group. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes. Cell. 1993;72(6):971-983. doi:10.1016/0092-8674(93)90585-e

Spinocerebellar Ataxia Type 1 (SCA1):- Similar to SCA3 and HD, SCA1 is caused by an expanded CAG repeat in the ATXN1 gene, which results in a neurotoxic polyglutamine expansion in the ataxin-1 protein. This discovery is detailed in: Banfi S, Servadio A, Chung MY, et al. Identification and characterization of the gene causing type 1 spinocerebellar ataxia. Nat Genet. 1994;7(4):513-520. doi:10.1038/ng0894-513

The literature consistently shows that CAG repeat expansions lead to the production of proteins with abnormally long polyQ tracts, which misfold and aggregate, leading to neuronal dysfunction and death. This aggregation is toxic to cells, particularly in the central nervous system, resulting in the progressive symptoms seen in these disorders.

It is important to note that the length of the CAG repeat is a critical factor in the pathogenesis of these diseases. Longer repeats tend to be associated with more severe symptoms and an earlier age of onset, a phenomenon known as anticipation. Clinical and molecular genetic studies have confirmed these observations across different populations and have provided a better understanding of these complex disorders.

Further research continues to uncover the precise mechanisms of toxicity, including effects on gene transcription, protein-protein interactions, and cellular homeostasis. Studies on therapeutic strategies, such as the use of antisense oligonucleotides, are ongoing to find treatments that can alter the course of these devastating diseases.

The therapeutic rationale for using allele-preferential antisense oligonucleotides (ASOs) to target CAG repeat expansions in neurodegenerative disorders such as Spinocerebellar Ataxia type 3 (SCA3), Huntington's disease (HD), and Spinocerebellar Ataxia type 1 (SCA1) has a varied evidence base with both strengths and weaknesses.

Strengths of the Evidence Base:

  1. Molecular Target Identification: The identification of the specific genetic mutations (CAG repeat expansions) that cause these diseases is strong and provides a clear molecular target for therapy.

  2. Mechanistic Understanding: The understanding of how CAG repeat expansion leads to the production of toxic proteins, and the subsequent cellular dysfunction is supported by a wide range of studies across molecular biology, genetics, and histopathology.

  3. Preclinical Studies: Preclinical studies in cell models and animal models of these diseases have shown that targeting CAG repeat expansions with ASOs can decrease levels of the toxic proteins and ameliorate disease symptoms.

  4. Clinical Precedent: Clinical successes with ASOs for other conditions, such as spinal muscular atrophy (SMA) and familial amyotrophic lateral sclerosis (ALS), lend support to the idea that ASOs can be effective drugs for neurodegenerative diseases.

Weaknesses of the Evidence Base:

  1. Translational Challenges: Although the preclinical studies are promising, many treatments that succeed in animal models do not translate to success in human trials. Differences in human vs. animal biology, disease progression, and ASO delivery can result in unanticipated outcomes.

  2. Allele Specificity: While allele-specific targeting is a goal, the potential for off-target effects or incomplete specificity is a concern. It's challenging to ensure that the ASO interacts exclusively with the mutant allele without affecting the normal allele.

  3. Long-term Effects: There is limited evidence about the possible long-term effects of downregulating genes in the human brain, as lifelong treatment is often needed for these chronic conditions.

  4. Clinical Trial Data: As of my last update, there were limited published clinical trial results specifically for ASOs targeting CAG repeat expansions in these conditions. Trials are ongoing, and more robust clinical data are needed to validate this approach.

  5. Clinical Endpoint Identification: Defining appropriate clinical endpoints to measure the efficacy of ASOs in neurodegenerative diseases can be complex, given the slow progression and variable presentation of these conditions.

  6. Technical Challenges: The efficient and targeted delivery of ASOs to the CNS across the blood-brain barrier remains difficult. Although newer ASO modifications improve CNS penetration, the optimal delivery method and dosing frequencies are still being investigated.

  7. Variability in Response: Genetic and environmental factors may lead to variability in patient responses to ASO treatment. Personalized approaches and precision medicine may be required to address this variability.

In conclusion, while a strong scientific rationale and promising preclinical data support the use of allele-preferential ASOs for treating SCA3, HD, and SCA1, the real test of their efficacy and safety will come from ongoing and future clinical trials. The continued development of ASOs for these diseases will be informed by this emerging clinical data, which will either strengthen the evidence base or reveal new challenges to overcome.


Clinical trial overview


Phase 1/2 study design

VO659 is being studied in a first-in-human trial for Spinocerebellar Ataxia Type 1 (SCA1), Spinocerebellar Ataxia Type 3 (SCA3), and Huntington's Disease (HD), assessing safety, tolerability, and pharmacokinetics (PK) of VO659, an antisense oligonucleotide, after intrathecal delivery. The trial is an open-label, phase 1/2a with multiple ascending doses over a 42-week period per participant.

Participants receive VO659 on four separate occasions (Day 1, 29, 57, and 85) followed by a 23-week monitoring phase. Initial dose cohorts consist exclusively of SCA3 patients, with later cohorts expanded to SCA1 and HD patients. Primary outcome measures include safety through adverse events tracking (treatment-related, serious, of special interest, or severe), vital signs, body weight, ECG, lab safety parameters for blood and CSF, and MRIs for brain structure, and screening for suicidal ideation or behavior via the C-SSRS. Secondary outcomes focus on pharmacokinetics—concentration of VO659 in CSF and plasma, along with plasma Cmax, Tmax, AUC0-t, and terminal half-life.

Critiques on the Study Design:

  1. Open-Label Bias: The lack of placebo controls and blinding could introduce bias in reporting outcomes and diminish the strength of the safety and efficacy conclusions.

  2. Cohort Composition: Starting with only SCA3 patients might limit the early safety and dosage data's applicability to SCA1 and HD, which may have different responses to treatment.

  3. Pharmacodynamics Measures: The description focuses on pharmacokinetics, with less emphasis on pharmacodynamics—how the drug actually affects the trajectory of the disease.

  4. Patient Population Limitation: By selecting "generally ambulatory participants with mild to moderate SCA1 or SCA3, or early manifest HD", the study might miss potential adverse effects in more advanced diseases stages and limit generalizability of the findings.

  5. Longitudinal Concerns: Since neurodegenerative diseases progress slowly, the follow-up period may not be long enough to observe significant pharmacodynamics outcomes, which may become more apparent over extended periods.

Operational/Technical Challenges:

  1. Intrathecal Administration: The lumbar intrathecal bolus injections require skilled healthcare providers and may cause discomfort or complications for patients, affecting compliance and retention.

  2. Sample Collection: Regular collection of CSF requires repeated lumbar punctures, which may lead to patient discomfort and dropout, as well as technical challenges in consistent sample collection.

  3. Participant Retention: The lengthy trial period may lead to patient dropout, skewing the results if the dropouts are non-random (e.g., only those experiencing side effects leave the study).

  4. Dose Escalation: Management of the dose-escalation in response to potential adverse effects may prove complex, particularly when balancing sufficient drug exposure with patient safety.

  5. Monitoring Complex PK: Monitoring multiple time points for VO659 concentration and plasma parameters requires precise timing and could be technically demanding, necessitating specialized laboratory requirements.

  6. Safety Monitoring: Frequent detailed assessments, such as ECGs and MRIs, are operationally intensive and require coordination across multiple centers and consistency in execution and interpretation.

This study holds the potential to provide proof-of-concept for the use of VO659 in treating Spinocerebellar Ataxia type 3 (SCA3) by using a rigorous methodology for assessing the treatment’s safety and pharmacokinetic profile. By ensuring that participants have genetically confirmed disease, the study is well-targeted at those most likely to be affected by the intervention. The use of well-established clinical scales, such as the Scale for Assessment and Rating of Ataxia (SARA), is appropriate for ensuring that participants have a degree of disease severity that is high enough to detect changes but not so advanced as to confound the results with other disease-related complications.

Appropriateness of Primary and Secondary Endpoints:

The primary endpoints selected are focused on safety and tolerability, which is appropriate for a first-in-human phase 1/2a trial. The comprehensive list of safety assessments covers a wide range of potential adverse effects and provides a good overview of the drug's impact on general health. Secondary endpoints related to the pharmacokinetic profile of VO659 are also critical parts of the proof-of-concept, as they will inform the understanding of how the drug is processed in the body, how it reaches target sites, and the relationship between administered dosage and drug concentration in relevant tissues.

Inclusion / Exclusion Criteria:

The criteria are suitably stringent to produce a homogenous participant group, which is necessary for reducing variability and increasing the reliability of the collected data. The age range (25-60 years) excludes potentially complicating factors related to pediatric or geriatric populations, and the requirement for genetic confirmation of disease ensures the target pathology is indeed present.

However, the need to have mild to moderate disease, as measured by SARA for SCA3 (and similar criteria for SCA1 and HD), may exclude patients with very mild or more severe symptoms who may also benefit from the drug. Furthermore, these criteria will make enrollment more challenging, potentially affecting the recruitment phase and thus delaying the study.

Reproducibility Challenges Posed by the Inclusion / Exclusion Criteria:

Taken together, while the strict inclusion and exclusion criteria are essential to control variables and ensure patient safety, they also pose certain challenges not only to the recruitment and retention of trial participants but also to the broader applicability and reproducibility of the study's findings. Subsequent trials will need to consider how to expand these criteria to ensure that the treatment is effective and safe in a wider pool of patients.


Market overview


Spinocerebellar Ataxia Type 3

Spinocerebellar Ataxia Type 3 (SCA3), also known as Machado-Joseph Disease, is a type of hereditary ataxia caused by a genetic mutation. It is one of the most common forms of autosomal dominant ataxias.

The hallmark of SCA3 is progressive ataxia, or lack of muscle coordination, which worsens over time. This primarily affects gait and limb coordination. Other symptoms can include eye movement abnormalities, dysarthria (difficulty in speaking), and dysphagia (difficulty in swallowing). As the disease progresses, patients may develop non-ataxia symptoms such as parkinsonism, spasticity, peripheral neuropathy, and cognitive impairment. The severity and range of symptoms can vary widely among individuals, even within the same family.

SCA3 is diagnosed through a combination of clinical assessment and genetic testing. The presence of a family history of similar symptoms is a strong indicator. Genetic testing identifies the CAG repeat expansion in the ATXN3 gene.

SCA3 is a progressive disease, meaning symptoms worsen over time. The rate of progression can vary. The onset of symptoms typically occurs in mid-adulthood, but can range from adolescence to late adulthood. Life expectancy can vary. Some individuals live for decades after the onset of symptoms, although they may require significant assistance in later stages.

To assess the market opportunity for VO659 in Spinocerebellar Ataxia Type 3 (SCA3), it is important to consider several factors, including the prevalence of the disease, the current standard of care, the competitive landscape, potential advantages of the new treatment, and the unmet needs in the therapeutic area.

  1. Prevalence: SCA3, also known as Machado-Joseph Disease, is the most common subtype of autosomal dominant cerebellar ataxias. Although considered a rare disease, it has a global presence with variable prevalence. Estimates suggest that its prevalence ranges from 0.3 to 2 per 100,000 in the general population. Despite its rarity, because it's the most common form of hereditary ataxia, there is a significant market for therapies.

  2. Current Standard of Care: As of early 2023, there is no cure for SCA3, and treatment is mainly supportive and symptomatic. Physical therapy, occupational therapy, and speech therapy are standard parts of patient care. Patients may also receive pharmacological treatment for symptoms like spasticity, pain, and sleep disturbances. Therefore, any drug that could modify the disease course or significantly reduce symptomatic burden could disrupt the market.

  3. Competitive Landscape: Given that SCA3 is a rare disease with no disease-modifying treatments available, the competitive landscape for VO659 may not be crowded. Prior to introducing VO659, it's essential to understand any other compounds in development and their mechanism of action, effectiveness, side effect profile, and where they are in the clinical pipeline. Success comes with first-mover advantage or demonstrating superior efficacy or safety.

  4. Other Successful Drugs in the Indication: There are no currently successful drugs in modifying the disease, which means a significant opportunity if VO659 can demonstrate efficacy in this area. However, drugs that manage symptoms or rehabilitative equipment/supplies (e.g., for gait abnormalities) may indirectly constitute the current "competition."

  5. Unmet Medical Need: There is a considerable unmet need in SCA3 for treatments that can slow, halt, or reverse disease progression. Patients and families also need therapies to better manage the diverse symptoms of the disease. Any drug that can tackle the cause of the disease at a molecular level, such as potential gene therapy or a drug targeting the underlying genetic mutation, would fulfill a significant unmet medical need.

  6. Other Considerations: Reimbursement, market access, and drug pricing are critical factors in defining the market opportunity. Rare disease treatments often carry high price tags but are supported by payers due to a lack of alternatives. Additionally, orphan drug status (if obtained) could offer benefits like market exclusivity, tax credits, and assistance with the regulatory application process.

Finally, VO659's market opportunity will also be influenced by its safety and tolerability profile. If it has fewer side effects compared to symptomatic treatments, this would be a strong competitive advantage. Also, demonstrating benefits that align with patients' goals such as improved quality of life, motor function, or increased independence can solidify its value proposition.

There are several therapeutic strategies in development for Spinocerebellar Ataxia Type 3 (SCA3) or actively being researched that could potentially compete with VO659. Here I'll outline a few of the promising avenues:

  1. Gene Therapy: Gene therapies that aim to correct the underlying genetic defect in SCA3 are being explored. These could involve strategies like gene silencing or editing to reduce the production of the mutant ataxin-3 protein that causes SCA3. Treatments that utilize CRISPR-Cas9 technology or antisense oligonucleotides (ASOs) to target the mutated gene could be direct competitors if in development.

  2. RNA Interference (RNAi): This approach uses small interfering RNAs (siRNAs) or ASOs to reduce the mutant ataxin-3 protein expression. Such treatments could slow or stop the progression of the disease and would be a major breakthrough. As of early 2023, CRISPR Therapeutics and Ionis Pharmaceuticals are examples of companies that have been involved in RNAi-based treatments for other diseases, which might also pursue treatments for SCA3.

  3. Small Molecule Inhibitors: These are molecules that can cross the blood-brain barrier and potentially alter disease progression by changing the protein folding or aggregation pathway, such as those targeting the proteostasis network. There might be small molecules in development that aim to modulate these pathways to provide a therapeutic benefit in SCA3.

  4. Stem Cell Therapy: Although likely in earlier stages of research, stem cell therapies aim to regenerate or repair the damaged neuronal cells in the cerebellum of patients with SCA3. If this technology becomes viable, it could revolutionize treatment for a range of neurodegenerative disorders, including SCA3.

  5. Symptomatic Treatments: Even though these do not alter the disease course, they could still compete indirectly with VO659 by improving the quality of life for patients. New drugs that offer better control of symptoms with fewer side effects could be preferred by patients and physicians.

  6. Repositioned Drugs: Occasionally, drugs approved for other indications show promise in treating different diseases. If a currently approved medication is identified as being effective for SCA3, it could be brought to market more quickly than VO659, especially if it is safe and well-tolerated.

Any company attempting to bring a treatment for SCA3 to market would need to conduct robust clinical trials to prove efficacy and safety. The speed at which competitors are able to progress through clinical stages and obtain regulatory approval will determine the level of competition VO659 might face. It will also be essential to understand how these potential treatments are different from VO659 in terms of their mechanism of action to fully evaluate the competitive landscape.

Continuous monitoring of emerging clinical trial data and the scientific literature is vital to have an up-to-date perspective on the treatments being developed for SCA3, including their clinical efficacy, safety, and the profile of companies backing these projects.As of my last update, there were no disease-modifying drugs specifically approved for the treatment of Spinocerebellar Ataxia type 3 (SCA3). Treatment for SCA3 primarily focused on managing symptoms and improving quality of life, as there had not been a therapy available that could alter the disease progression.

The drugs used in the symptomatic treatment of SCA3 typically include:

  1. Antispasmodics: For muscle cramps and spasticity, drugs like baclofen or tizanidine may be prescribed.

  2. Parkinsonian Medication: Levodopa and other Parkinson’s disease medications might be tried in some patients, particularly those with Parkinsonian features, although their effectiveness in SCA3 is not well-documented.

  3. Antidepressants and Antiepileptics: These may be used for neuropathic pain management. Drugs such as gabapentin or pregabalin are examples that have been used for neuropathic pain in various conditions.

  4. Sleep Aids: Medications could be prescribed to manage sleep disturbances, which are common in SCA3.

It should be noted that most treatments are used off-label, meaning they are not specifically approved for SCA3 by regulatory bodies like the FDA but have shown some efficacy in managing symptoms in these patients.

It is not possible to assess how VC659 might fit into the standard of care for SCA3 without data on how the drug impacts patients with the disease. Nevertheless, based on typical avenues of research and treatment options being explored for SCA3, let's consider how a hypothetical drug like VO659 might fit in if it were to be effective and approved:

  1. Disease-Modifying Potential: If VO659 proved to be disease-modifying, it could revolutionize the management of SCA3. Disease-modifying therapies could potentially slow disease progression or improve neurological function, which would be a paradigm shift from the current symptomatic management approach.

  2. Symptomatic Relief: If VO659 were aimed at managing symptoms of SCA3 — such as ataxia, muscle stiffness, or spasticity — it might be adopted alongside or in place of current symptomatic treatments, depending on its efficacy and safety profile relative to existing options.

In summary, the positioning of VO659 within the standard of care for SCA3 would largely depend on the detailed outcomes of clinical trials and the specifics of the drug's action and benefits. If VO659 demonstrated a clear advantage over current symptom management strategies in efficacy, safety, or both, it could become a significant part of the treatment paradigm for SCA3.

Huntington's disease


Huntington's disease is a progressive neurodegenerative disorder with a genetic basis. It significantly impacts both the nervous system and an individual's overall functional abilities.

Huntington's disease is caused by a genetic mutation in the HTT gene, leading to the abnormal expansion of a CAG trinucleotide repeat. This mutation results in an abnormal form of the protein huntingtin, which is toxic to neurons, particularly in the brain's basal ganglia and cerebral cortex. The mutant huntingtin protein forms aggregates within neurons, leading to cell death and brain atrophy, especially in areas involved in movement, cognition, and behavior.

Symptoms include:

Diagnosis is typically based on the combination of characteristic clinical features and a family history of Huntington's disease. Genetic testing is used to confirm the diagnosis, revealing the expanded CAG repeat in the HTT gene.

Huntington's disease is progressive and currently incurable. The onset of symptoms usually occurs in adulthood, typically in the 30s or 40s, although onset can range from childhood to old age. The progression of the disease varies but generally spans 15 to 20 years from the onset of symptoms. Late-stage disease is marked by severe motor and cognitive impairments.

  1. Prevalence: Huntington's disease is a rare, inherited neurodegenerative disorder characterized by motor dysfunction, psychiatric symptoms, and cognitive decline. The prevalence of HD in Western populations is about 5 to 10 per 100,000 people, indicating a relatively small but significant market opportunity for effective therapies.

  2. Current Standard of Care: Treatment for HD is primarily symptomatic, focusing on managing the different symptoms of the disease since there is no cure or treatment to slow the progression of the disease. Tetrabenazine is the only FDA-approved drug specifically for the treatment of chorea associated with Huntington's disease. Other medications often used include antipsychotics, antidepressants, mood stabilizers, and medications to control movement.

  3. Other Successful Drugs in the Indication: No disease-modifying drugs for HD had been successfully developed as of my last update, though several compounds have undergone clinical trials. For example, deutetrabenazine, a newer version of tetrabenazine with fewer side effects, has been approved for the treatment of HD chorea. Additionally, several strategies to lower the levels of mutant huntingtin protein (mHTT) have been pursued, such as antisense oligonucleotides, but none had led to a marketed product for disease modification.

  4. Unmet Medical Need: There is a significant unmet medical need in HD for treatments that can alter the disease course. The goal is to develop therapies that can delay onset, slow progression, or improve the symptoms that severely affect patients’ quality of life. Given the genetic basis of the disease, therapies that target the underlying cause or modify the disease process have the potential to fulfill this unmet need.

  5. Market Opportunity: Given the lack of disease-modifying treatments and the progressive nature of HD, the market opportunity for an effective treatment like VO659 could be substantial, irrespective of the disease's relative rarity. A new therapy that demonstrates safety and efficacy in altering disease progression or significantly improving functional outcomes or quality of life could command a high price point and market share, given the low number of alternative treatments available.

A successful new therapy in this space would address a mix of factors: efficacy in symptomatic relief or disease modification, improvement in the quality of life, a tolerable side effect profile, and ease of administration that aligns with the needs and capabilities of HD patients. Success in the market would also require seamless navigation through regulatory hurdles, a refined market access strategy, and a comprehensive understanding of the competitive landscape.

Furthermore, since HD profoundly impacts patients' ability to work and engage in daily activities, there is a strong argument from a health economics perspective for treatments that can extend the independent functioning of patients. Insurers and health systems can be receptive to these treatments despite the costs due to the long-term savings in healthcare costs and societal benefits.

In conclusion, a safe and effective treatment for HD, which addresses either symptomatic management or disease modification, represents a significant market opportunity due to high unmet clinical need, willingness to pay for effective therapies, and relative lack of competition. However, the successful commercialization of VO659 would depend on the demonstration of clinically meaningful benefits and achieving end-user acceptance (patients, clinicians, payers).

There are several promising avenues of research into treatments for Huntington's disease (HD):

  1. Antisense Oligonucleotides (ASOs): One of the most advanced approaches in treating HD involves the use of ASOs to reduce the production of the mutant huntingtin protein, which is believed to cause the neurodegeneration in HD. Roche's tominersen (formerly RG6042) had generated significant interest, although its late-stage clinical trial was halted due to a lack of efficacy. Despite this, the approach remains a key area of research.

  2. Small Molecule Inhibitors: These therapies target various pathways that may be dysregulated in HD, such as signaling pathways involved in neurodegeneration. For example, small molecules that modulate the activity of kinases or other enzymes involved in cellular homeostasis could provide symptomatic relief or potentially slow disease progression.

  3. Gene Editing: Techniques like CRISPR-Cas9 offer the long-term potential to correct the genetic mutation at the DNA level within neural cells. While still in the relatively early stages of research, gene editing represents a potentially curative approach but faces significant challenges in delivery, safety, and precision.

  4. RNA Interference (RNAi): Similar to ASOs, RNAi therapeutics aim to reduce levels of mutant huntingtin protein. RNAi-based drugs are designed to degrade mRNA before it can be translated into the problematic protein.

  5. Protein Homeostasis Modulators: These compounds aim to improve the cellular machinery's ability to deal with misfolded proteins. They can enhance the function of proteostasis networks which includes chaperones and protein degradation pathways.

  6. Neuroprotective Strategies: Drugs aimed at protecting neurons from damage include various compounds that reduce oxidative stress, improve mitochondrial function, or modulate excitotoxicity. Though they may not reverse or significantly slow the disease's progression, they could potentially improve or preserve neuronal function.

  7. Stem Cell Therapy: Investigations are ongoing into the potential of stem cells to provide neuroprotective effects or replace damaged neurons, though this area of research is still in early stages.

Any company developing treatments for HD must demonstrate that their drug can achieve significant clinical benefits in a patient population that is both in need and highly motivated for new therapeutic options. The ideal treatment would alter the disease progression through a disease-modifying mechanism, have a favorable safety and tolerability profile, and be convenient for patients to use.

For the latest updates on treatments for Huntington's disease, it would be crucial to monitor current clinical trials, publications in major neurology journals, and press releases from companies involved in the HD therapeutic market.

There are only a few notable pharmaceutical treatments for Huntington's disease (HD), and no disease-modifying treatments have been approved. The drugs available are primarily aimed at symptom management rather than altering the disease course:

  1. Tetrabenazine (Xenazine): This is one of the first drugs that was approved by the FDA for the specific treatment of chorea associated with Huntington's disease. Tetrabenazine reduces the amount of dopamine available at synapses and is therefore effective in treating hyperkinetic movement disorders.

  2. Deutetrabenazine (Austedo): Similar to tetrabenazine, deutetrabenazine is approved for the treatment of chorea associated with Huntington's disease and has the advantage of possibly causing fewer side effects due to its longer half-life, which allows for a lower dosage and less frequent dosing.

  3. Antipsychotics: Drugs like risperidone, olanzapine, and haloperidol are used off-label to manage some of the psychiatric symptoms of HD, such as chorea and psychosis. However, these can carry significant side effect risks, especially in the long term.

Aside from these, a range of other medications are used off-label to address various symptoms associated with HD. Antidepressants may be used for depression, mood stabilizers for mood swings and aggressive behavior, and other medications may be used for irritability and anxiety. Treatment is highly individualized and may require a combination of medications to manage the diverse symptoms experienced by patients with HD.

If VO659 indeed has a novel mechanism of action that targets the pathophysiology of HD or provides symptomatic relief more effectively than existing treatments, it might well define a new standard of care for HD. Conversely, if VO659 offers similar benefits to current therapies but with additional advantages, such as reduced dosing frequency or a better side effect profile, it may still find a niche in treatment protocols.

In summary, for VO659 to be integrated into the standard of care for HD, it would need to establish itself as a safe and effective option that addresses the unmet medical needs of this patient population. Factors such as how the drug is administered, patient quality of life outcomes, cost, payer coverage, and market access would also play a role in determining its potential adoption and utilization within the clinical setting. An understanding of how VO659 compares to other drugs in the pipeline is also critical for assessing its future place in the care regimen for HD.

Spinocerebellar Ataxia Type 1

Spinocerebellar Ataxia Type 1 (SCA1) is one of the several types of spinocerebellar ataxias, which are a group of hereditary, progressive, neurodegenerative disorders. Like other types of SCAs, SCA1 is characterized by impaired coordination of movement but has its own distinct features and progression pattern.

SCA1 is caused by a genetic mutation involving an abnormal expansion of a CAG trinucleotide repeat in the ATXN1 gene. This mutation leads to the production of an abnormal form of the protein ataxin-1. The mutant ataxin-1 accumulates in neurons, particularly in the cerebellum, brainstem, and spinal cord, causing neuronal dysfunction and degeneration. The pathology primarily affects the Purkinje cells in the cerebellum, but other areas of the central nervous system are also involved as the disease progresses.

The onset of symptoms usually occurs in adulthood, often between 30 and 40 years of age. Early symptoms typically include problems with coordination and balance (ataxia), which progressively worsen. Other symptoms can include dysarthria (difficulty speaking), dysphagia (difficulty swallowing), eye movement abnormalities, and cognitive impairment. As the disease progresses, symptoms such as spasticity, weakness in the limbs, and neuropathy may develop. Some individuals may also experience non-motor symptoms like depression and cognitive changes.

Diagnosis is based on the clinical presentation, family history, and genetic testing. Genetic testing can confirm the presence of the CAG repeat expansion in the ATXN1 gene. SCA1 is a progressive disorder, meaning that symptoms gradually worsen over time. The rate of progression can vary among individuals. Life expectancy varies, but individuals often live for several decades after the onset of symptoms. In later stages, complications such as pneumonia or other infections can be life-threatening.

There are no specific treatments for Spinocerebellar Ataxia Type 1 (SCA1), a progressive and degenerative genetic disorder. The market opportunity for a drug like VO659 in SCA1 would be similar to that in other forms of Spinocerebellar Ataxias such as SCA3, which I previously described.

  1. Prevalence: SCA1 is a rare condition, part of a group of similar genetic disorders known as SCAs. While precise prevalence can vary, SCAs together affect around 1 to 2 people per 100,000 globally, with SCA1 being one of the more common subtypes. The rarity of the condition suggests a small, specific target population, which may benefit from orphan drug status that can provide market exclusivity and other incentives.

  2. Current Standard of Care: The management of SCA1 is purely supportive and symptomatic. This includes physical therapy to enhance mobility and prevent complications, occupational therapy to assist with daily tasks, and speech therapy to help with communication. Pharmacological treatments may address symptoms such as spasticity, dystonia, and tremors but do not slow the disease's progression. This creates a strong opportunity for a drug like VO659 if it can show efficacy in modifying the disease course or substantially improving symptoms.

  3. Other Successful Drugs in the Indication: No drugs have been developed that specifically target the mechanisms that cause SCA1. Current pharmaceutical treatments are therefore not disease-specific and are used off-label to treat symptoms. Any drug that could offer a disease-modifying effect for SCA1 would be a first in its class and could become the market leader due to the high unmet need.

  4. Unmet Medical Need: The unmet need in SCA1 is significant, as no disease-modifying treatments are available, and symptoms invariably progress. Treatments that can slow or halt disease progression or offer considerable symptomatic relief would represent a major advance and could command a significant market presence. Beyond efficacy, other aspects contributing to unmet needs are the safety, quality of life improvement, and ease of drug administration.

  5. Market Opportunity: Given the lack of direct competition and the significant unmet medical need in SCA1, the market opportunity for a safe and effective treatment like VO659 could be quite favorable. The field is open for first-mover advantage, and the willingness of healthcare systems to pay for orphan drugs remains high, especially if those drugs can provide clear clinical benefits.

A successful entry into the market for VO659 would depend not only on its clinical efficacy and safety profile but also on factors such as manufacturing costs, pricing strategies, reimbursements policies, regulatory hurdles, market access, and acceptance by the clinical community. To capture the market opportunity, the drug's developer would need to engage with patients, advocacy groups, and neurologists to ensure that the full therapeutic potential of VO659 is understood and realized.

In summary, due to the lack of existing competitors and the possibility of securing orphan drug designation, VO659 could have a strong market opportunity in treating SCA1, provided that the drug shows favorable outcomes in clinical trials. Any new market entrant that offers a substantial therapeutic advantage would meet with significant interest from patients, healthcare providers, and payers alike.

Similar to SCA3, there are many promising avenues of research for SCA1 treatments, including, gene therapy, antisense oligonucleotides, RNAi, small molecules, protein homeostasis modulators, and stem cell therapy.

Currently, the condition is managed through symptomatic treatment, and patients are often prescribed a variety of medications that are used to manage symptoms associated with the disease, rather than the disease itself.

Common symptomatic treatments include:

  1. Antispasmodics: To manage muscle spasms and spasticity, patients may be prescribed medications such as baclofen or tizanidine.

  2. GABA Agonists: Gabapentin, which is sometimes used off-label for cerebellar ataxia, can help with pain and spasticity.

  3. Parkinsonian Medications: Some SCA1 patients may exhibit Parkinson's-like symptoms and may benefit from medications like levodopa.

  4. Beta-blockers: Sometimes used off-label for tremor management in cerebellar ataxias.

  5. Physical and Occupational Therapies: These are not drugs but are crucial components of the supportive treatment for SCA1, aiming to preserve motor function and independence for as long as possible.

There have been no newly approved branded drugs specifically for the treatment of SCA1 as of my most recent information. Research efforts in SCA1 and other SCAs focus largely on understanding the genetic and molecular underpinnings of these diseases to develop targeted therapies, including gene therapy, RNA-based therapies, and small molecules that may alter disease progression.

Given the unmet need in this area, a drug that could be approved for SCA1 would represent a major advance in the field and likely command significant attention and market share. The focus remains on seeking treatments that can either slow or halt the progression of the disease or considerably improve the patients' quality of life by addressing the specific mechanisms that underlie the neurodegeneration in SCA1.

Weekly analyses of biotech startups, generated by AI

Receive high quality, AI-generated analyses of biotech startups, public companies and scientific papers each week.


Platform overview

The scientific strategy of Vico Therapeutics involves the following aspects:

  1. RNA Modulation: Vico's approach centers on manipulating the RNA to modulate protein expression. RNA-targeted therapeutics can take various forms, including antisense oligonucleotides (ASOs), small interfering RNA (siRNA), and RNA editing. These techniques aim to modify the expression of genes implicated in neurological disorders either by reducing the production of harmful proteins or by correcting the RNA transcripts to produce the correct form of the protein.

  2. Focus on Rare Diseases: By targeting rare diseases like spinocerebellar ataxia and Huntington's disease, Vico may be able to address unmet medical needs where there are limited treatment options. Focusing on rare diseases also often comes with regulatory incentives like orphan drug designation that can expedite the development process.

  3. Precision Medicine: Vico's strategy likely incorporates precision medicine, tailoring treatments based on the genetic makeup of individual patients or subpopulations. This requires an understanding of the genetic mutations and pathways involved in each condition and designing therapeutics that specifically target those elements.

Similar approaches have been adopted by other companies working in the field of RNA-targeted therapeutics, such as Ionis Pharmaceuticals and Alnylam Pharmaceuticals. These companies have also developed ASOs and siRNA therapies for various genetic and rare diseases.

Risks and pitfalls that Vico might encounter include:

  1. Delivery Challenges: Effective delivery of RNA therapeutics to the CNS is a significant challenge. The blood-brain barrier (BBB) is a major obstacle, and getting sufficient amounts of the therapeutic agent into the brain without causing adverse effects requires sophisticated delivery systems.

  2. Off-target Effects: RNA-targeted therapies may have unintended interactions with other RNAs or proteins, leading to off-target effects. This can cause unintended side effects, which may be particularly concerning in the context of the CNS with its complex and sensitive functions.

  3. Immune Response: The introduction of synthetic oligonucleotides like ASOs and siRNAs can trigger immune responses, potentially leading to inflammation or other adverse reactions.

  4. Durability and Reversibility: Some RNA therapies may not provide long-lasting effects, requiring repeated administration. Additionally, the reversibility of the treatment is important, especially if adverse effects are observed.

  5. Technical and Regulatory Hurdles: Developing RNA-based therapies involves navigating through rigorous preclinical and clinical testing. Ensuring safety, efficacy, and meeting the regulatory requirements for approval is both time-consuming and costly.

  6. Market Risks: The commercial success of approved treatments for rare diseases is not guaranteed. The small patient populations and high costs of therapies might limit market penetration. Additionally, obtaining insurance coverage can be challenging due to the high price points often associated with orphan drugs.

In conclusion, the scientific strategy of Vico Therapeutics likely involves leveraging RNA modulation to target the underlying genetic causes of certain neurological disorders. While promising, this approach carries a set of risks and challenges that will need to be carefully managed through robust preclinical and clinical development, sophisticated drug delivery systems, and a nuanced understanding of regulatory and market landscapes.

Weekly analyses of biotech startups, generated by AI

Receive high quality, AI-generated analyses of biotech startups, public companies and scientific papers each week.









You may also like...

Biotech IPO tracker

The top biotech VCs

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