Metagenomi Technologies IPO investment analysis

January 8, 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


Metagenomi is a company specializing in genetic medicine, focusing on developing treatments for genetic diseases using a unique genome editing toolbox derived from metagenomics. This approach involves analyzing genetic material from various natural environments to discover and utilize microbial genetic diversity.

The platform includes an array of tools such as programmable nucleases (including type II and type V Cas nucleases and SMall Arginine-Rich sysTems or SMART nucleases), base editors, RNA and DNA-mediated integration systems, prime editing systems, and CRISPR-associated transposases (CASTs). The platform leverages environmental DNA from diverse ecosystems, which is deeply sequenced to identify novel proteins and CRISPR systems. This has led to the discovery of over 20,000 new genome editing systems. The company employs high-throughput screening, artificial intelligence, and proprietary algorithms to mine through billions of novel proteins for creating genome editing tools.

Metagenomi's tools are designed for site-specific genome editing. This includes programmable nucleases for precise DNA cutting, base editors for single nucleotide changes, and systems for both small and large genomic integrations. The tools have been extensively evaluated in various models, including human primary cells and nonhuman primates, demonstrating efficacy and addressing limitations like off-target effects. The platform's tools are compatible with a range of delivery methods, both viral and nonviral, due to the varied sizes and biochemistry of the nucleases and guide RNAs.

Metagenomi filed to go public in January 2024. This note includes an analysis of the potential IPO valuation.


Pipeline overview


Product nameModalityTargetIndicationDiscoveryPreclinicalPhase 1Phase 2Phase 3FDA submissionCommercial
Hemophilia A program Gene editing FVIII knock in Hemophilia A



Primary Hyperoxaluria Type 1 program Gene editing HAO1 knock down Primary Hyperoxaluria Type 1



Transthyretin Amyloidosis program Gene editing TTR knock down Transthyretin Amyloidosis



Cardiovascular Disease program Gene editing AGT knock down Cardiovascular Disease

Alpha 1 Antitrypsin Deficiency program Gene editing A1AT RIGS/Base editing Alpha 1 Antitrypsin Deficiency

Wilson's Disease program Gene editing ATP7B RIGS Wilson's Disease

Familial ALS program Gene editing SOD1 Knock down Familial ALS

Spontaneous ALS program Gene editing ATXN2 Knock down Spontaneous ALS

Charcot-Marie-Tooth Type 1A program Gene editing PMP22 Knock down Charcot-Marie-Tooth Type 1A

Duchenne Muscular Dystrophy program Gene editing DMD Exon skipping/CAST Duchenne Muscular Dystrophy

Cystic Fibrosis program Gene editing CFTR CAST Cystic Fibrosis

Immuno-oncology cell therapy programs Cell therapy Multiple Oncology



Autoimmune CAR-T program Cell therapy Multiple Autoimmune disease


Highlights and risks


Highlights

Broad platform of gene editors including base editors, prime editors and systems for large-scale integration

Potential best-in-class editors, including some of the smallest base editors to date

Platform of over 20,000 new genome editing systems provides potential for differentiated, unique systems

Large pipeline of a variety of gene editing programs targeting validated indications

Risks

Early-stage pipeline with no programs in clinical trials

Targeting validated, but competitive indications creates a high bar for competitive differentiation

Development of broad pipeline will require significant capital and partnerships, and prioritizing asset development will be critical


Valuation



Metagenomi filed to go public in January 2024. The company has riased over $350 million to date. We estimate the post-money valuation of the last Series B-1 round at $806 million.

Based on comparable IPOs, we estimate an IPO valuation range of $1.1 billion to $1.8 billion, on a fully diluted post-money basis. We estimate that the company could raise $200-300 million in the IPO.

Due to the early-stage of the pipeline, we did not conduct a DCF analysis. We anticipate that the company could potentially become an M&A target after demonstrating clinical proof-of-concept, but due to the early-stage nature of the gene editing field, there have not been any large M&A transactions.

Based on recent IPOs, we estimate the following investors could be interested in Metagenomi's IPO:




Weekly analyses of biotech startups, generated by AI

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


Major Translational Programs


Hemophilia A Program

This program focuses on a novel, durable, knock-in method to enable the expression of the clotting Factor VIII (FVIII). Instead of providing the FVIII gene episomally, which could be unstable, Metagenomi's approach is to insert a FVIII DNA cassette into a 'safe harbor location' within the albumin gene. This allows for the expression of FVIII driven by the strong albumin promoter.

Feasibility has been shown in mice, and ongoing studies in nonhuman primates (NHP) have shown promising results for up to 4.5 months post-treatment. The goal is to establish long-term FVIII expression.

Next steps include comparison of different FVIII donor DNA cassettes in NHP studies to select a development candidate by Q2 2024, and preparation for IND-enabling studies.

Primary Hyperoxaluria, Type 1 (PH1) Program

The program aims for a durable knockdown of HAO1 gene expression to reduce the substrate in PH1, a metabolic disorder leading to kidney failure. Optimal nuclease and gRNA combinations have been screened, and preclinical proof-of-concept has been achieved in a mouse model.

Next steps include completion of candidate confirmation for NHP studies, with NHP data expected in 2024 to support final development candidate selection.

Transthyretin Amyloidosis Program

The goal is to knock down TTR gene expression in patients with this disease, where misfolded proteins aggregate and cause organ dysfunction. More than 90% knockdown of human TTR protein has been achieved after a single dose in a humanized TTR mouse model.

Next steps include advanced nuclease and guide selection, with plans to move into NHP studies in 2024.

Further Areas of Focus

These programs are all in the preclinical stage, with no approved products yet. The goal of these programs is to leverage Metagenomi's genome editing toolbox to develop therapies that could offer long-term solutions to these diseases, supported by validating preclinical and clinical data, with a focus on meeting significant clinical needs.


Platform overview


Metagenomi's platform includes a variety of gene editing components:

Their approach offers several advantages:

Metagenomi's platform represents a comprehensive and advanced system for genome editing, with the potential to address complex genetic diseases through a variety of sophisticated and precise tools.


Overview of Gene Editing


Gene editing is a revolutionary technology that enables precise, directed changes to genomic DNA. At its core, it involves the use of engineered nucleases, or "molecular scissors," to make targeted cuts in the DNA strand. This allows for the deletion, insertion, or alteration of specific DNA sequences. Key technologies in this field include CRISPR-Cas9, TALENs (Transcription Activator-Like Effector Nucleases), and ZFNs (Zinc Finger Nucleases). CRISPR-Cas9, in particular, has garnered widespread attention due to its ease of use, efficiency, and versatility.

Some notable gene editing products and their applications include CRISPR Therapeutics' CTX001 for the treatment of beta thalassemia and sickle cell disease, and Editas Medicine's EDIT-101 for Leber Congenital Amaurosis, a rare genetic eye disease. These products represent the forefront of gene editing technologies being applied in clinical settings.

Compared to traditional therapeutic approaches, gene editing offers a high degree of specificity and the potential for a one-time curative treatment. This approach can directly target the root cause of genetic diseases, rather than just treating symptoms. It's particularly advantageous in diseases caused by a single gene defect. Gene editing also reduces the long-term costs and complications associated with ongoing treatments like enzyme replacement therapies.

The potential of gene editing extends beyond monogenic diseases. It holds promise for the treatment of more complex conditions like cancer, HIV, and neurodegenerative diseases. In oncology, gene editing could be used to engineer immune cells to better target and destroy cancer cells. The technology is also being explored in the development of antiviral strategies, particularly for diseases like HIV, where the virus integrates into the host genome. Furthermore, ongoing research is investigating the use of gene editing for regenerative medicine, such as repairing damaged tissues and organs.

While the full potential of gene editing is yet to be realized, its capability to provide precise, personalized, and potentially permanent treatments for a range of diseases marks it as a transformative advance in the field of medicine.


Limitations of Gene Editing Technologies


Various challenges are faced by traditional CRISPR-based approaches, as well as other methods like NHEJ, HDR, base editing, prime editing, serine integrases, PASTE technology, and more. Here's a summary:

Beyond these general limitations, specific gene editing techniques have unique challenges:

Metagenomi's technology can potentially address the previously mentioned limitations:

In summary, Metagenomi's platform presents a promising approach to addressing several limitations of current gene editing technologies, especially in terms of diversity and specificity of tools, and targeting flexibility. However, there are inherent challenges in the field of gene therapy, such as delivery, immune response, and HDR efficiency, that are not fully overcome by any current technology, including Metagenomi's. Continued research and clinical trials will be crucial to assess the efficacy and safety of their platform.


Platform in depth


Programmable nucleases


Metagenomi's approach to identifying programmable nucleases is multifaceted and innovative, leveraging the diversity of microbial life to find novel genome editing tools. Here's how they do it:

Metagenomi uses a proprietary database built from environmental samples to discover a vast array of novel CRISPR systems. This database is expected to contain novel sequences that have evolved in various microorganisms, providing a rich source of potential new genome editing enzymes.

Their attention is on type II and type V CRISPR systems due to their simplicity and programmability. These systems use RNA-guided nucleases that can target specific DNA sequences and, in some cases, RNA sequences. They have found several new types of ultra-small nucleases, which they refer to as SMART, along with novel type V sub-groups. These are considerably smaller than the commonly used SpCas9, making them potentially more suitable for certain delivery vectors.

To validate the activity of these newly discovered nucleases, Metagenomi performs high-throughput in vitro testing. This process involves bioinformatic predictions followed by experimental validation in cell-free systems and mammalian cells. Their screening process emphasizes finding natural, un-engineered nucleases that show high activity and specificity. They aim to identify systems that perform as well or better than SpCas9 with fewer off-target effects. They evaluate the targeting density of their nucleases, which refers to the frequency of targetable sites within the human genome. A higher targeting density increases the chances of finding an effective nuclease-guide RNA combination for any given genomic target.

Beyond simple genome editing, the programmable nucleases they discover can serve as platforms for developing more specialized tools like base editors and RNA-mediated integration systems (RIGS). They also use a modular approach to create chimeric systems that combine the high activity of top-performing nucleases with the unique targeting domains from other systems. This allows them to tailor their tools to specific genomic targets.

Metagenomi's technology has the potential to overcome some limitations of current gene editing technologies. Their smaller, potentially more specific nucleases could address delivery size constraints and target sequence limitations. The high-throughput screening process and focus on specificity could help minimize off-target effects. However, challenges such as in vivo delivery, immune responses, and the efficiency of homology-directed repair (HDR) are not specifically addressed and remain areas where Metagenomi's approach will need to demonstrate efficacy. Additionally, the translation of these discoveries into clinical applications is a complex process that will require further development and validation.

Metagenomi's approach to gene editing using a diverse array of programmable nucleases from their metagenomics platform offers several potential advantages over established nucleotide editing approaches:

These advantages suggest that Metagenomi's approach could complement and potentially surpass the capabilities of existing gene editing technologies in terms of precision, versatility, and applicability. However, these benefits are theoretical until proven in clinical settings, where real-world challenges such as delivery efficiency, immune responses, and long-term safety need to be thoroughly evaluated.

Weekly analyses of biotech startups, generated by AI

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

The evidence presented by Metagenomi indicates that their nucleases have the potential to address several limitations of current gene editing technologies. However, while the data appears robust and promising, the translation from preclinical models to successful clinical applications will be the ultimate test of these advantages. It will be important for Metagenomi to demonstrate that the nucleases work effectively in humans, with sustained efficacy and without adverse effects over the long term. Additionally, while the in vitro and in vivo data are encouraging, issues such as delivery to specific tissues, immune responses, and long-term durability of the edits in the context of a living organism are complex challenges that still need to be navigated as development progresses.


Potential advantages of Metagenomi's Nucleases


Nucleases for gene editing face several limitations, some of which are inherent to the nature of the technology and some due to biological complexity. Here is an overview of these challenges and an assessment of how well Metagenomi's nucleases might overcome them:

Delivery to Specific Tissues Effective delivery to target tissues besides the liver, such as the brain or muscle, remains a challenge. Metagenomi's discovery of ultra-small nucleases could facilitate the use of delivery vectors with broader tissue tropism, potentially addressing this limitation.

Immunogenicity The human immune system can recognize and mount a response against the bacterial-derived Cas proteins used in gene editing, which may limit their effectiveness. Metagenomi's approach to identifying a diverse array of nucleases may uncover enzymes that are less immunogenic or amenable to modifications that reduce immunogenicity.

Durability of Editing Ensuring that gene edits are stable over the long term and do not revert or cause late-onset off-target effects is important. The specificity and high efficiency of Metagenomi's nucleases suggest they could create durable edits, but this needs to be confirmed over time in living organisms.

Efficiency in Different Cell Types Some nucleases work well in certain cell types but not others. Metagenomi's nucleases have shown high activity across a range of primary human cells, which is promising for their versatility.

Complex and Large-Scale Genomic Edits Achieving large-scale genomic insertions or complex rearrangements is difficult with current technology. While Metagenomi's platform shows promise for a variety of edits, the ability to perform large or complex rearrangements has not been specifically addressed.

In Vivo Re-administration The potential for immune responses against viral vectors can limit the ability to re-administer gene-editing components when necessary. Metagenomi's platform does not directly address this challenge, as it is more related to the delivery system than to the nuclease itself.

Ethical and Regulatory Hurdles Gene editing technology raises significant ethical and regulatory considerations, especially concerning germline editing. These are policy and public perception issues that are outside the technical scope of Metagenomi's platform.

Cost-Effectiveness The development, manufacturing, and delivery of gene-editing therapies can be expensive. While the use of Metagenomi's high-throughput screening and AI algorithms could streamline some aspects of the development process, it remains to be seen how this will impact the overall cost of therapies developed using their nucleases.

Scalability of Manufacturing Producing gene-editing components at a scale sufficient for widespread clinical use while maintaining quality is challenging. Metagenomi's methods may simplify the discovery process, but manufacturing scalability is a separate issue that is not directly addressed by their technology.

Cas9-Induced P53 Response There's evidence that the DNA damage response from Cas9 editing can activate the p53 pathway, which may select for p53-deficient cells, potentially leading to oncogenic outcomes. The specificity of Metagenomi's nucleases may reduce DNA damage and hence p53 activation, but this would need specific investigation.

In conclusion, Metagenomi's nucleases show potential to overcome several but not all of the limitations faced by current gene editing tools. Some challenges, particularly those related to delivery, immune response, and long-term safety and efficacy in humans, are areas where additional research and clinical data will be necessary to fully assess the capabilities and limitations of their technology.


Metagenomi's Base Editors


Metagenomi's approach to identifying base editors is rooted in the integration of their proprietary programmable nucleases with their vast metagenomics discovery platform. Here's how they are developing these base editors and how the evidence supports the potential advantages of their technology:

Metagenomi has created both Adenine Base Editors (ABEs) and Cytosine Base Editors (CBEs), which have been validated in mammalian cells and in vivo. These base editors consist of a targeting component (a modified nuclease) and a deaminase that chemically alters the specific bases. ABEs and CBEs offer the advantage of precise editing without inducing double-stranded breaks, a significant advancement over traditional CRISPR/Cas9 editing which often relies on creating such breaks.

The base editors are designed using modified nickases, programmed to make a single-strand cut in the DNA, allowing the deaminase to chemically modify the targeted bases. This avoids the potential complications associated with double-stranded DNA breaks, such as unwanted insertions or deletions.

Evidence of high editing efficiency is presented, showing that their ABEs and CBEs can achieve significant levels of editing. This includes up to 60% editing efficiency in primary hepatocytes and 35% in vivo in mouse livers using a lead ABE construct.

The described ABEs and CBEs can target a significant proportion of the adenine and cytosine bases in the human genome. This extensive targetability is demonstrated by the theoretical coverage of their base editors, which significantly exceeds that of traditional SpCas9-based editors.

Metagenomi has discovered and engineered some of the smallest base editors to date, which provides substantial opportunities for vector optimization. These editors, derived from their SMART nuclease platform, are within the size range compatible with AAV delivery methods.

The provided data suggests that Metagenomi's ABE system is capable of efficient editing in primary cells and in vivo, supporting its potential for therapeutic applications. The editing efficiency in mouse liver suggests that a significant proportion of the target cells were successfully edited.

The small size of the SMART base editors, particularly those under 1,000 amino acids, suggests that they could be efficiently packaged into AAV vectors, which are commonly used for in vivo delivery but have size limitations.

Summary: The evidence provided suggests that Metagenomi's base editors have the potential to address some of the limitations of current gene editing technologies, particularly in terms of precision and delivery. The data supporting their claims include in vitro and in vivo studies, showing high editing efficiency and specificity. Their approach could allow for the correction of point mutations associated with a wide range of diseases and the efficient knockdown of genes without the risks associated with double-stranded DNA breaks.

However, while the in vitro and in vivo data seem promising, the true test of these potential advantages will come with further development and clinical trials. The efficiency and specificity of these base editors in humans, their long-term stability and safety, and the ability to scale up for therapeutic applications are all areas that will require additional investigation.


Advantages of Metagenomi's Base Editing Approach


Metagenomi's approach to base editing, as outlined and based on the evidence provided, suggests several potential advantages over established base editing approaches:

Smaller Size for AAV Delivery One of the primary advantages of Metagenomi's base editors is their small size, particularly those derived from the SMART nuclease platform. Smaller nucleases can be more easily packaged into AAV vectors, which have strict size limitations. This can enable the delivery of the entire base editing construct within a single AAV vector, as opposed to splitting it across two vectors, which is often necessary with larger SpCas9-based systems.

Higher Targeting Flexibility Metagenomi's base editors are designed to have extensive genome targetability, potentially allowing for the targeting of a broader range of genomic sites. This could provide the ability to correct a wider array of point mutations across the genome compared to traditional base editors.

Reduced Off-Target Effects The approach emphasizes the selection of highly specific nucleases, which may lead to fewer off-target effects. While off-target effects are a concern for all CRISPR-based editing tools, Metagenomi's meticulous screening for specificity could provide an advantage in terms of safety.

Efficiency Without Double-Stranded Breaks By using engineered nickases that induce single-strand breaks instead of double-stranded breaks, Metagenomi's base editors aim to maintain high editing efficiency while reducing the risks associated with double-stranded DNA breaks, such as translocations or large deletions.

Potential for Multiplexing The presented data suggests that their base editors could be used for multiplexing in primary human T cells with no impact on cell viability. This could be particularly advantageous for therapeutic applications where multiple genes need to be edited simultaneously.

Rapid Optimization Metagenomi's modular, chimeric nuclease platform allows for the rapid optimization of base editors capable of editing various target sites. This can speed up the development of new therapeutic applications.

Novel Deaminases The company has mined a vast library of over three million deaminases, allowing for the identification of novel enzymes that may require minimal engineering to achieve high editing efficiencies.

Versatility for Therapeutic Development The versatility of the platform, as shown by effective editing in primary cells and in vivo, indicates potential use in broad therapeutic applications, including both ex vivo cell therapies and direct in vivo treatments.

While these advantages are promising, it is important to recognize that moving from preclinical success to clinical efficacy and safety is a significant hurdle. The actual therapeutic benefit of these base editors will need to be demonstrated in clinical trials, and issues such as long-term safety, delivery to the correct tissues, and avoidance of immune responses will be critical factors to consider.


Metagenomi's RIGS


Metagenomi's approach to developing RNA-mediated Integration Systems (RIGS), which encompasses both prime editing (Little RIGS) and systems for large genomic integrations (Big RIGS), is detailed and innovative. Here’s how their approach works and how the evidence supports their strategy:

The approach and evidence suggest that Metagenomi's RIGS could potentially provide several advantages over existing gene editing methods:

While these potential advantages are promising, the ultimate test will be the clinical application of these systems. The translation of benchtop innovations to bedside treatments will require thorough validation in clinical trials, ensuring that the edits are safe, stable, and efficacious in the long term. Additionally, while the approach may simplify some aspects of delivery and increase the range of edits, challenges such as immune responses, in vivo efficiency, and tissue-specific targeting still need to be addressed.

Metagenomi's approach to RNA-mediated Integration Systems (RIGS), including Little RIGS for prime editing and Big RIGS for large genomic integrations, could offer several potential advantages over other approaches:

The potential advantages outlined by Metagenomi's approach align with the ongoing evolution of gene editing technologies towards higher precision, safety, and versatility. However, the transition from promising preclinical results to clinical success is complex, and these technologies will need to demonstrate safety, efficacy, and deliverability in a clinical setting. The potential for off-target effects, immune responses, and challenges associated with in vivo delivery and long-term expression stability will be crucial factors to consider as Metagenomi's RIGS technology progresses toward therapeutic applications.


Metagenomi's Systems for Large Genomic Integration


Metagenomi's approach to developing systems for large genomic integration, such as CRISPR-associated transposases (CASTs), appears to focus on overcoming limitations of current gene therapy methods, especially for diseases that require the introduction of large segments of DNA. Here’s a summary of their strategy:

The evidence presented suggests that Metagenomi’s novel CAST systems have demonstrated the capability for targeted integration of transgenes into human cells, which is a significant step forward in the genome editing field. If these results can be replicated and scaled up, they could indeed provide a new class of genome editing therapeutics capable of addressing complex genetic diseases with large genomic integrations. However, the efficiency, specificity, long-term stability, and safety of these integrations in vivo, as well as the potential immune responses to such interventions, will need to be thoroughly investigated in further preclinical and clinical studies.


Metagenomi's CAST integration systems


Metagenomi's approach to large genomic integrations using CRISPR-associated transposases (CAST) and other DNA-templated systems offers several potential advantages compared to other gene therapy approaches:

Despite these potential advantages, it is important to note that clinical translation involves many challenges. These include ensuring that the integrations are stable and do not lead to long-term adverse effects, confirming that the integrated genes are expressed at therapeutic levels without disrupting the expression of other important genes, and avoiding immune responses to the integrated sequences or the proteins they encode. Additionally, manufacturing and regulatory hurdles for new genetic medicines can be substantial. As such, while the preclinical evidence is promising, comprehensive clinical studies will be necessary to validate these potential advantages in a therapeutic setting.

Potential for Therapeutic Development

The successful demonstration of targeted integration lays the groundwork for therapeutic-driven optimization. The next steps would likely focus on increasing efficiency, ensuring long-term expression and safety, and possibly developing single vector systems.

The evidence supports the potential advantages of Metagenomi's CAST technology for integrating large DNA cargos with specificity. However, the relatively low integration efficiency highlights the need for further research and optimization to reach the levels required for clinical application. Additionally, this is preclinical proof-of-capability, and translating these findings into safe and effective therapies will involve overcoming challenges related to immune responses, long-term stability, and the control of gene expression levels post-integration. The continued discovery and development of novel genome editing systems through Metagenomi's metagenomics platform suggest a promising trajectory for advancing the field of genome editing.

Weekly analyses of biotech startups, generated by AI

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


Market Overview in Hemophilia A


Hemophilia A is a genetic disorder caused by missing or defective Factor VIII, a clotting protein. The condition manifests in an inability to form blood clots effectively, leading to prolonged bleeding after injury or surgery and spontaneous bleeding, especially into joints and muscles. Severe cases may experience frequent joint bleeds, leading to chronic joint disease and pain. With appropriate treatment, individuals with Hemophilia A can lead relatively normal lives. However, without management, the condition can result in chronic joint damage, life-threatening hemorrhages, and significantly reduced quality of life.

Hemophilia A affects around 1 in 5,000 to 10,000 male births worldwide. An estimated 20,000 individuals in the United States live with Hemophilia A. Globally, the number is estimated at over 400,000.

Symptoms include

Standard of care includes:

Gene therapies like valoctocogene roxaparvovec aim to introduce a functional FVIII gene to produce the clotting factor endogenously. These therapies have shown promise in maintaining therapeutic levels of FVIII and reducing bleeding events. Durability of expression and immune responses are key areas of ongoing research. Novel therapies exploring RNA-based treatments or CRISPR-based gene editing are also being developed for a more durable and potentially curative option:

While replacement therapy is effective, it is not a cure and requires lifelong treatment. The risk of inhibitor development and the high cost of therapy remain significant challenges, along with the need for effective treatments for children and those with inhibitors. Gene therapy offers potential but faces issues such as potential waning effects, immune responses, and re-treatment limitations. Below are some notable aspects of the market:

The Hemophilia A market is at a transformative stage, with potential shifts from conventional therapies to longer-acting and potentially curative treatments such as gene therapies and gene editing. These advances offer the hope of reducing the treatment burden and improving the quality of life for patients with Hemophilia A. However, these therapies come with high costs and complex manufacturing processes, which may pose challenges for widespread adoption. The successful commercialization of these products will depend on their long-term efficacy, safety profiles, and cost-effectiveness compared to existing treatments.


Gene Therapies for Hemophilia A


Gene therapies for hemophilia A aim to provide a long-term solution to the underlying cause of the disease: the lack of functional Factor VIII (FVIII) protein. By introducing a functional FVIII gene into the patient's own cells, the body can potentially produce its own FVIII, mitigating or even eliminating the need for regular factor infusions. Here are several gene therapy approaches that have been in development:

The gene therapy market for hemophilia A is poised to grow as these therapies progress through clinical development and receive regulatory approvals. Their success will depend on overcoming challenges related to durability of expression, immunogenicity, and accessibility. Each new therapy will need to demonstrate a favorable risk-benefit profile to gain acceptance.

There are several safety concerns associated with gene therapies for hemophilia A that researchers, clinicians, and regulatory agencies monitor closely:

Clinical trials for gene therapies in hemophilia A are designed to carefully monitor patients for these and other potential safety issues. Long-term follow-up studies are also a key part of understanding and managing the risks associated with gene therapies.


Metagenomi's Hemophilia A approach



Primary Hyperoxaluria Type 1 (PH1)


Primary Hyperoxaluria Type 1 (PH1) is a rare genetic disorder that affects the metabolism, leading to excessive production and accumulation of oxalate. It is caused by a deficiency of the liver enzyme alanine:glyoxylate aminotransferase (AGT), due to mutations in the AGXT gene. This enzyme deficiency results in the overproduction of oxalate, a substance that, when present in high amounts, combines with calcium to form calcium oxalate crystals that can lead to kidney stones and kidney damage.

The prognosis for individuals with PH1 varies depending on the severity of the condition and when treatment is initiated. If left untreated, PH1 can lead to end-stage renal disease (ESRD) usually in childhood or young adulthood. Systemic oxalosis, where oxalate crystals accumulate in other organs, can occur once the kidneys fail, leading to bone disease, anemia, skin ulcers, and heart and eye problems, further complicating the condition.

PH1 is estimated to affect 1 to 3 people per million, with higher prevalence in certain regions due to founder effects or consanguinity. It accounts for a significant proportion of pediatric ESRD cases.

The standard of care includes aggressive hydration to reduce oxalate concentration in urine, use of citrate to prevent stone formation, pyridoxine (Vitamin B6) in responsive patients, and dialysis in cases of ESRD. Liver transplantation has been a treatment option, as the liver is the site of the defective enzyme; dual liver-kidney transplantation may be considered for those with significant kidney damage.

Despite advancements, there remains an unmet medical need for effective treatments that can prevent the overproduction of oxalate and protect kidney function without the need for lifelong treatments or invasive procedures like organ transplantation.

Emerging therapies include gene therapy approaches that aim to correct the underlying genetic defect and prevent the overproduction of oxalate. Lumasiran, an RNA interference (RNAi) therapy, targets the HAO1 gene to decrease the production of glycolate oxidase, thus reducing oxalate production. Nedosiran, another RNAi therapeutic, inhibits lactate dehydrogenase, which is also involved in oxalate production. These therapies aim to reduce the oxalate burden significantly, potentially changing the disease's natural history. However, issues such as long-term efficacy, accessibility, cost, and the need for ongoing treatment persist, highlighting the ongoing need for novel therapeutic approaches.


Therapeutic Rationale for Gene Editing in Primary Hyperoxaluria Type 1


The therapeutic rationale for using gene editing to durably knock down HAO1 in Primary Hyperoxaluria Type 1 (PH1) is rooted in addressing the underlying metabolic defect causing the disease. PH1 is characterized by mutations in the AGXT gene, leading to dysfunctional alanine glyoxylate aminotransferase and subsequent accumulation of glyoxylate. This excess glyoxylate is converted to oxalate, forming insoluble calcium oxalate crystals in the kidneys.

Metagenomi is partnered with Moderna for the PH1 program. A summary of Metagenomi's approach is as follows:

Safety and Efficacy

Advantages Over Current Treatments

Overall, the gene editing approach to knock down HAO1 in PH1 represents a novel and potentially transformative therapy. It targets the metabolic basis of the disease with the prospect of a durable, once-off treatment, offering significant advantages over current management strategies.


Risks in Gene Editing Approach for PH1


The gene editing approach to durably knock down HAO1 in Primary Hyperoxaluria Type 1 (PH1) is promising, but it comes with potential scientific and clinical risks crucial for evaluating the treatment's safety and efficacy.


Transthyretin Amyloidosis (ATTR)


Transthyretin Amyloidosis (ATTR) is a progressive and often fatal disease characterized by the accumulation of amyloid fibrils, primarily composed of transthyretin (TTR) protein, in various tissues and organs. It significantly impacts clinical outcomes, particularly affecting the heart and peripheral nerves. ATTR leads to progressive organ dysfunction, affecting cardiac and neurological functions, with a life expectancy of typically 3-5 years after diagnosis for cardiac manifestations.

There are two primary types of ATTR:

Approximately 50,000 patients globally with ATTRv and 300,000-500,000 with ATTRwt, more common in older adults and certain populations.

Standard of care includes:

Despite treatments, ATTR remains progressive with significant morbidity and mortality. Existing therapies require lifelong treatment with limited efficacy, especially in advanced stages. Challenges include treatment accessibility, need for earlier diagnosis, and impact on cardiac and neurological functions.


Gene Editing in Transthyretin Amyloidosis

The therapeutic rationale for using a gene editing approach to knock down the expression of the TTR gene in Transthyretin Amyloidosis (ATTR) focuses on addressing the root cause of the disease – the misfolding and aggregation of transthyretin protein. Metagenomi is partnered with Ionis in ATTR.

In ATTR, either variant (ATTRv) or wild-type (ATTRwt) TTR proteins misfold and aggregate, forming amyloid deposits in tissues, particularly in the heart and peripheral nerves. The TTR protein is synthesized primarily in the liver.

Current treatments, like tafamidis and similar therapies stabilize the TTR tetramer but do not stop its production. Antisense oligonucleotides and siRNA therapies aim to reduce TTR levels but require ongoing treatment and may not completely halt disease progression.

The benefits to a gene editing approach include:

While gene editing is a promising, potentially longer-lasting therapy, it comes with additional risks:

The gene editing approach for TTR knockdown in ATTR is innovative and potentially durable, but it carries risks and challenges, particularly in safety and delivery. Compared to existing therapies, it offers sustained benefits but lacks established safety profiles and reversible treatment options. Careful clinical development and safety monitoring are vital for mitigating these risks.


Key license agreements


Moderna Agreement



Affini-T agreement



Ionis Agreement



Competition


Metagenomi faces competition from various entities based on their technological focus and corporate status. Here is an overview:

In summary, Metagenomi operates in a highly competitive field with various companies employing different gene editing technologies. These include both public and private companies engaged in innovative research and development across several areas, including CRISPR/Cas9, base editing, prime editing, older genome editing technologies, and gene therapy approaches.









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