Gate Bioscience investment analysis

November 6, 2023


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 a relationship with the company.



Overview



Gate Bioscience is a preclinical-stage biotech company specialized in developing "Molecular Gates", a new class of small molecule therapeutics designed to selectively block disease-causing extracellular proteins from being secreted by cells. Founded in 2021 and emerging with $60 million Series A funding from Versant Ventures, a16z Bio + Health, ARCH Venture Partners, and GV, the company's approach targets the secretory translocon—a cellular channel crucial for protein secretion.

With more than 1,000 extracellular proteins implicated in various diseases, including cancer, neurodegeneration, fibrosis and other disorders, the commercial potential of Gate's platform is vast.

As small molecules, molecular gates can potentially be administered orally and cross the blood-brain barrier. This distinguishes Gate Bioscience’s potential treatments from biologics and other larger molecule therapeutics, which typically face challenges in oral bioavailability and central nervous system penetration.

The company's next steps likely include preclinical validation of its therapeutic candidates, optimizing their drug-like properties, and conducting the necessary studies to progress to clinical trials. Given the early stage of the company, the focus will be on establishing proof-of-concept and safety profiles for their lead compounds.


Highlights and risks


Highlights

Small molecule Molecular Gates potentially provide functional benefits of biologics without their challenges in oral bioavailability and central nervous system penetration

Potential to impact a wide variety of diseases including neurodegeneration, cancer, fibrosis and many other diseases

Potential to develop leading platform for a new class of therapeutics

Experienced management team and advisors with expertise in secretory translocon structure and function

Risks

Significant technical risk involved in developing new modality, including specificity/selectivity, pharmacology, and manufacturing

Inappropriate blockade of secretory translocon could have undesirable effects

Platform risk: if significant technical issues emerge related the platform or modality, company's value will be negatively impacted

Significant spend on platform development in advance of identifying and developing products can create valuation overhang


Valuation


Given the early stage of the company and limited information about its programs, we did not conduct a valuation analysis.


Scientific approach


While specifics about Gate's programs are not available, we can speculate on how their approach works:

The approach resembles that of targeted protein degradation strategies like PROTACs (proteolysis-targeting chimeras), but with a distinct focus on intercepting and degrading proteins during their synthesis and secretion pathway. The innovation here lies in the interception at an earlier, pre-secretory stage, which could have significant advantages in terms of preventing extracellular accumulation of pathogenic proteins, a key factor in many diseases.


The secretory translocon


The secretory translocon, primarily composed of the Sec61 complex in eukaryotic cells, is a fundamental component of the endoplasmic reticulum (ER) membrane that facilitates the translocation of newly synthesized polypeptides into the ER lumen or their insertion into the ER membrane. The ER is the entry point for proteins destined for secretion or residence in the endomembrane system and plasma membrane.

The Sec61 complex is the core of the translocon. It is formed by multiple subunits:

The Sec61 complex associates with additional components depending on the context, such as:

The translocon works as a highly selective gate, ensuring that only proteins with the appropriate signals are translocated or integrated. It can open and close in response to these signals, preventing ions and other small molecules from leaking across the ER membrane. The intricate workings of the translocon involve complex mechanisms that are not fully understood, such as how it distinguishes between proteins to be integrated into the membrane and those to be secreted into the ER lumen. The translocon has multiple functions:

The specificity and efficiency of the secretory translocon make it a compelling target for therapeutic intervention, as drugs that modulate its function could potentially influence the fate of numerous proteins, including those implicated in various diseases.


Gate's expertise in the secretory translocon


The involvement of scientific founders and advisors such as Pat Sharp, Rebecca Voorhees, Ramanujan Hegde, and Ville Paavilainen, who have demonstrated expertise in the mechanics of protein secretion and the function of the secretory translocon, particularly Sec61, provides deeper insight into the potential mechanisms that could be at play in Gate Bioscience's Molecular Gates technology.

Sec61 is a key component of the translocon complex at the endoplasmic reticulum (ER) membrane, acting as a channel through which nascent proteins are translocated into the ER lumen or integrated into the ER membrane. Here's how their expertise might inform and enhance the Molecular Gates technology:

Analyze your company with our AI tools

Get a report like this one for your company or portfolio. Includes excel files with valuation analysis.

Contact us to learn more.


Technical and scientific risks


Developing therapies based on a mechanism like the Molecular Gates presents a range of challenges and limitations. Here are some key considerations:

To manage these challenges, sophisticated pharmacological models would likely be necessary to predict how molecular gates behave in different tissues and under various physiological conditions. Extensive preclinical and clinical testing would be required to establish safe and effective dosing regimens. This may include the development of biomarkers to monitor the effects of the molecular gate and guide dose adjustments for individual patients.


Therapeutic applications


The strategy to selectively eliminate disease-causing extracellular proteins by targeting them inside the cell before they can be secreted and cause harm has the potential to impact a wide range of diseases. Moreover, because these are small molecules, they may have advantages in terms of tissue distribution, oral bioavailability, and the ability to cross the blood-brain barrier, which is particularly important for treating central nervous system diseases.

According to the company, there are over 1,000 extracellular proteins that are implicated in diseases, and theoretically all of these proteins can be targeted with Molecular Gates. The following diseases and conditions could be potential targets for their molecular gate therapies:

However, it's important to note that the specific targeting of disease-associated proteins without affecting the normal function of other proteins that go through the secretory pathway will be crucial for the safety and effectiveness of such therapies. The development of these drugs will likely require a deep understanding of the structure and function of both the translocon and the target proteins, as well as robust screening and testing to ensure specificity and efficacy.

More specifically, here are some diseases that could be good candidates for molecular glues:

Disease Therapeutic Rationale for Molecular Gates Existing Antibody/Other Therapies Advantages of Molecular Gates Over Existing Therapies Unmet Need Challenges Using Molecular Gates Potential Protein Targets
Rheumatoid Arthritis Molecular gates could block pro-inflammatory cytokines directly at the source within cells. TNF inhibitors (e.g., Humira, Enbrel) Oral administration, potential for fewer side effects, and deeper tissue penetration. Non-injectable treatments with fewer side effects. Specificity in blocking only disease-related cytokines without disrupting normal immune function. TNF-α, IL-6, IL-1β
Alzheimer's Disease Prevent the secretion and subsequent aggregation of beta-amyloid plaques. Monoclonal antibodies (e.g., Aduhelm) Small molecules may cross the blood-brain barrier more effectively and modulate protein secretion. Treatments that can halt or reverse disease progression. Targeting the correct stage of amyloid protein for effective intervention; crossing the blood-brain barrier; molecular gates would not help clear existing plaques Beta-amyloid precursor protein (APP)
HER2-positive Breast Cancer Inhibit the secretion of the HER2 receptor tyrosine kinase. Trastuzumab (Herceptin) and similar agents Oral bioavailability, ease of administration, and potentially reduced immunogenicity. Oral chemotherapy alternatives. Achieving selective inhibition of HER2 without affecting other necessary receptor pathways; ensuring that the normal function of related receptors and cellular processes is not adversely affected. Human Epidermal growth factor Receptor 2 (HER2)
Idiopathic Pulmonary Fibrosis Reduce fibrotic extracellular matrix protein deposition. Antifibrotic agents (e.g., Ofev, Esbriet) Directly targeting the secretory pathway could prevent the secretion of fibrotic proteins. More effective anti-fibrotic therapies with better outcomes. Identifying and targeting the specific proteins responsible for fibrosis without impacting normal tissue repair; targeting a single protein may not suffice due to the multifactorial nature of the fibrotic process Collagen, Fibronectin, TGF-β
Atherosclerosis Control the levels of lipoproteins and clotting factors, or targeting inflammation in atherosclerosis. Statins, PCSK9 inhibitors Potential for more specific targeting of proteins involved in plaque formation. Non-statin therapies for patients with hyperlipidemia intolerant to statins. Balancing the reduction of harmful proteins while maintaining levels required for normal physiological processes. Apolipoprotein B, PCSK9, Clotting Factors (e.g., Factor VIII)
Cystic Fibrosis May help correct protein folding and trafficking of the CFTR protein. CFTR modulators (e.g., Trikafta) Small molecules that can potentially address proteins that do not respond to current CFTR modulators. Treatments for CF patients who do not respond to current therapies. Ensuring the correct folding and function of the CFTR protein while avoiding off-target effects; current CFTR modulators are very effective for certain mutations so new therapies would likely address non-responders. Cystic Fibrosis Transmembrane Conductance Regulator (CFTR)
Systemic Lupus Erythematosus Reduce the secretion of autoantibodies and immune complexes. Biologics (e.g., Benlysta) and immunosuppressants Oral administration and targeted suppression Many patients have refractory disease despite current treatments, and long-term use of steroids and immunosuppressants leads to serious side effects. Identifying specific extracellular targets without affecting the normal immune response could be challenging; target specificity is exceptionally challenging due to the diversity of autoantibodies involved in SLE. Autoantibodies, BLyS (B lymphocyte stimulator)
Chronic Viral Infections Preventing the secretion and assembly of viral components, potentially reducing viral load and disease progression. Antiviral drugs, which may include direct-acting antivirals and host-targeted agents. Could offer a new mechanism of action to overcome resistance to current antivirals, with potentially broad applicability across different viruses. Resistance to current therapies, chronic infections require long-term management, and not all patients respond to existing therapies. Targeting viral proteins without affecting host protein secretion, risk of resistance development, and viral diversity; not all viral proteins are secreted via the classical secretory pathway, so molecular gate technology may have limitations depending on the virus and its biology Secreted viral proteins specific to each virus (e.g., hepatitis C virus (HCV) core protein, HIV gp120).

Potential first indication: rheumatoid arthritis


Of the above indications, rheumatoid Arthritis (RA) may be a compelling initial target for Gate Bioscience for several reasons:

Therefore, targeting RA as an initial focus could provide a strategic balance between scientific viability, clinical need, market potential, and regulatory clarity—all of which are crucial for a startup looking to establish itself in the biotechnology space.

Analyze your company with our AI tools

Get a report like this one for your company or portfolio. Includes excel files with valuation analysis.

Contact us to learn more.









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