December 13, 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.
TOME Biosciences has developed a technology platform for Programmable Genomic Integration (PGI), enabling the precise insertion of any size DNA sequence into specific genomic locations. This platform, licensed from MIT, combines CRISPR/Cas9's site-specificity with enzymes for inserting DNA, including entire genes, without double-strand DNA breaks.
The key component of this technology is the integrase-mediated PGI (I-PGI), which uses proprietary integrases for high-precision DNA insertion, akin to text-pasting in a word processor. I-PGI has successfully inserted over 30kb of genetic code into various cell types and can be used for complex cell engineering.
This technology facilitates more controlled gene therapy, allowing natural promoters in the human genome to regulate gene expression. Tome initially focuses on therapies for monogenic liver diseases and autoimmune diseases.
The team, led by Dr. Rahul Kakkar, brings extensive experience in pharma, biotech, and gene therapy. Having raised $213M from prominent biotech investors like Andreessen Horowitz and ARCH, Tome's platform represents a significant advancement in genomic medicine with potential widespread therapeutic applications.
Ability to insert large DNA sequences at specific sites is differentiated vs. existing technologies
Experienced management team and well-funded balance sheet
Technology has potential to develop therapies in many indications
Technology is early-stage and the risks and advantages are not fully understood
Cell therapy is expensive and logistically challenging, and cell therapy for autoimmune disease is still an emerging area
In vivo gene editing is still limited by delivery challenges
Given the early stage of the company and limited information about its programs, we did not conduct a valuation analysis. The company has raised $213M to date and is thus likely valued between $300-500M.
Tome Bioscience's PGI platform builds on CRISPR technology and is characterized by several innovative components and functions. The platform is based on technology licensed from MIT, coined "Programmable Addition via Site-specific Targeting Elements" (PASTE).
The platform enables the integration of large pieces of DNA into any part of the genome through a combination of targeted CRISPR-Cas9 technology and serine integrase-mediated recombination. Here’s how the system works to achieve this:
PASTE does not rely on creating DSBs, which minimizes unwanted DNA damage such as large deletions, translocations, and potential chromosomal rearrangements.
Unlike HDR (homology-directed repair) methods, PASTE is efficient in non-dividing cells, expanding its potential for therapeutic use in a wider range of cell types, including those predominantly non-dividing in the human body.
PASTE displays fewer off-target effects compared to other gene insertion methods like HITI (homology-independent targeted integration) due to the specificity provided by CRISPR-guided targeting and the use of circular DNA templates that limit random integration.
It has demonstrated high specificity and insertion purity, which are critical for therapeutic applications where accuracy and safety are paramount.
Prime editing allows for the introduction of small DNA edits (up to around 44 base pairs) without double-stranded DNA breaks. It is highly precise for small-scale edits but is not designed for integrating large DNA sequences. Base editors enable the conversion of one DNA base into another without making double-stranded DNA breaks, useful for correcting point mutations. However, they cannot be used to insert larger DNA sequences.
The combination of CRISPR-based targeting and serine integrase-mediated recombination in this system overcomes a limitation of both prime editing and base editing by enabling the insertion of much larger DNA sequences than what is possible with these methods. This capability is particularly significant for therapeutic applications where a functional gene or large DNA sequence needs to be integrated into the genome. It also potentially simplifies the integration process by using a single delivery mechanism to introduce both the integrase landing site and the DNA cargo.
The company makes several assertions about the capability of the technology. Based on the publication provided by the company, here is the evidence provided for each assertion.
The technology can insert DNA sequences of any size into designated genomic locations.
Evidence within the paper supporting this assertion includes:
It is important to be cautious in claiming that sequences of any size can be inserted, as the study demonstrates successful integration up to around 36 kb but does not empirically validate beyond that size. While the results are promising, they represent a subset of the potential sizes of DNA sequences.
Strengths of the Evidence:
Weaknesses of the Evidence:
In conclusion, while the evidence supports the assertion for sequences up to 36 kb, the claim that PASTE can insert sequences of any size into genomic locations requires cautious interpretation and further empirical support, especially for sequences larger than 36 kb. The technology is promising, but the biological complexities that come with genetic manipulation necessitate thorough understanding and careful claims.
The technology can insert DNA into any part of the genome
Evidence within the paper supporting the assertion
Strengths of the Evidence:
Weaknesses of the Evidence:
In summary, while the technology has been shown to integrate DNA at various sites within the genome, the assertion that it can insert DNA into any part of the genome is overly broad and requires additional evidence. There remain technical, biological, and efficiency challenges associated with CRISPR-based genome editing that can affect targeting flexibility. It's essential to clarify these limitations when asserting the capabilities of PASTE to ensure accurate communication of the technology's potential.
Enables the insertion of DNA sequences without the formation of double-strand breaks (DSBs)
This assertion is consistent with various portions of the text:
Strengths of the Evidence:
Weaknesses of the Evidence:
Concluding, the evidence strongly supports the assertion that PASTE can insert DNA sequences without the need for double-strand breaks, which is a pivotal improvement over existing genome editing technologies that often rely on induction of DSBs. This evidence is a strength of the technology and represents an area where PASTE has a clear advantage. However, it is essential for future studies to explore the limits and contexts in which this technology functions most effectively without DSBs to fully harness its potential.
Unprecedented precision compared to existing tech including base editing and prime editing
Evidence within the paper supporting the assertion:
Strengths of the Evidence:
Weaknesses of the Evidence:
Based on the evidence within the publication, PASTE shows promising precision for large DNA insertions with potentially fewer off-target effects than some traditional genome editing technologies. However, the provided data do not sufficiently compare PASTE with the precision of base editing and prime editing for it to be declared unprecedentedly precise across the board. While PASTE may offer advantages for inserting large DNA sequences, base and prime editing's precision for their designed applications—smaller nucleotide changes without double-strand breaks—sets a high bar which PASTE does not directly challenge. In summary, while the assertion of high precision is supported concerning certain editing approaches, the term "unprecedented" should be used cautiously until further comparative data are available.
The technology can be multiplexed, placing multiple sequences simultaneously
Strengths of the Evidence:
Weaknesses of the Evidence:
In conclusion, the publication provides solid evidence that PASTE can be multiplexed to place multiple sequences simultaneously at distinct genomic locations, showcasing one of its potential strengths. Multiplexed editing demonstrated in the paper represents an advance in gene insertion technology. However, more data would be beneficial to define the range and limitations of multiplexing capability more precisely and to ensure robust performance across varying conditions.
The technology can edit dividing and non-dividing cells
Strengths of the Evidence:
Weaknesses of the Evidence:
In summary, the evidence provided clearly demonstrates that PASTE can be used to edit both dividing and non-dividing cells, supporting the assertion. This capability broadens the potential applications of this technology, especially in the context of tissue and cell types that do not proliferate regularly. However, further research to optimize efficiency and validate the technology's efficacy across various non-dividing cell types and in vivo settings would be beneficial.
The technology has fewer off-target edits than other approaches
Evidence within the paper supporting the assertion:
Evidence not addressed within the paper:
Strengths of the Evidence:
Weaknesses of the Evidence:
In conclusion, while the paper demonstrates that PASTE has a specificity profile that may be more favorable than methods using DSBs like HITI, the assertion that it has fewer off-target edits compared to base editing or prime editing is not directly supported by the provided evidence. Comparative, comprehensive analyses would be required to fairly assess the precision of PASTE relative to these other highly precise genome editing technologies.
Gene editing technologies, including traditional CRISPR-based approaches and newer methods like base editing and prime editing, have transformed biomedical research and hold immense potential for treating genetic disorders. However, they also come with several limitations and challenges:
Below is discussion of the evidence supporting PGI's ability to overcome the limitations associated with gene editing. Some of these have already been discussed above, so I'll focus on limitations that haven't already been covered.
Drawing on evidence provided in the paper describing Tome's technology, I will look for evidence within the paper regarding PASTE's potential to overcome the limitation of off-target effects, common to CRISPR and other gene editing approaches:
Evidence within the paper:
The technology developed by Tome Biosciences, as detailed in the paper, addresses the challenge of delivering gene editing tools into target cells and tissues with the following evidence:
PASTE was demonstrated to function in both dividing and non-dividing cells, including primary human hepatocytes and T cells.
The paper mentions the delivery of PASTE components using adenoviral (AdV) vectors, which are noted to be capable of packaging large cargos, thus accommodating the size requirements of PASTE components for efficient delivery.
The use of programmable genome insertion without DNA repair pathways suggests that PASTE could be active across a range of cell types, potentially translating to various tissues in vivo.
In the paper, the researchers showed successful in vivo programmable gene insertion in the liver using PASTE components delivered via adenoviral vectors in mouse models.
There is mention of compatibility with different viral delivery methods, including adeno-associated virus (AAV) and adenovirus (AdV), to transport DNA integration templates, broadening the delivery options.
Strengths of the evidence:
Efficacy in a wide range of cells, including non-dividing primary human hepatocytes and T cells, indicates a broad potential for reaching different tissues.
Adenoviral delivery is a well-established method for in vivo gene therapy and the efficient transduction achieved is a strong point of evidence.
The successful demonstration of potential in vivo gene insertion in the liver of a mouse model raises confidence in the versatility and translatability of the PASTE delivery system.
Weaknesses of the evidence:
While the data indicate that PASTE components can be delivered via viral vectors in vitro and in mouse models, the safety, efficiency, and potential immunogenicity of such vectors in humans are not addressed in the paper.
Data on tissue-specific delivery in vivo are limited to the liver, and a broader range of tissues needs to be assessed to support the claim of wide applicability.
AAV vectors, although mentioned, are known for their limited packaging capacity, which could pose a constraint for delivering larger gene editing tools.
The long-term expression and potential integration of viral vector genomes into the host genome, which is a concern for AdVs, are not discussed.
Potential of the technology to overcome the limitation:
PASTE shows promise in overcoming the challenge of delivering gene editing tools into target cells and tissues, primarily due to its adaptability with viral vectors known for efficient delivery. The successful liver-targeted gene insertion in vivo signals potential applicability for therapeutic gene editing. However, additional research into alternate delivery systems, like non-viral vectors or physical methods (e.g., electroporation), could further augment delivery options and address limitations related to safety and specificity.
Evidence within the paper:
The paper mentions the consideration of immune responses to the components of the PASTE system, as outlined below:
The use of adenoviral (AdV) vectors and adeno-associated virus (AAV) vectors for delivery: While AdVs are known to elicit an immune response, AAVs are generally characterized by lower immunogenicity. The publication discusses the compatibility of PASTE with these vectors, suggesting that AAVs could be a less immunogenic option for delivering PASTE components.
Transcriptome analysis of cellular responses to PASTE: The paper states that transcriptome-wide analysis was conducted to assess off-target effects of expressing PASTE components in HEK293FT cells, with very few changes observed in gene expression, potentially indicating a limited cellular stress response.
PASTE in vivo testing: The in vivo application of PASTE in liver-humanized mouse models (specifically the FRG KO mouse model) is mentioned, which can provide preliminary data related to immune responses in a living organism, although the focus is on integration efficiencies and not the immune reaction per se.
Strengths of the evidence:
The consideration of different viral vectors for PASTE component delivery expands the range of options, potentially allowing for tailoring the delivery method to reduce immunogenicity.
The observation of minimal transcriptome changes when expressing PASTE components suggests a reduced stress or immune response to the system within the context of the tested cell type.
Weaknesses of the evidence:
While AAVs generally have a lower immunogenic profile, the response can be different in humans, and the long-term immunogenicity of PASTE components in clinical settings remains unknown.
Transcriptome analysis, although providing initial insights into cellular responses, does not directly address adaptive immune responses that could be triggered by the introduction of PASTE components into humans.
Data from in vivo mouse model experiments, while essential, may not fully represent the immune responses in humans, and further testing in more relevant models would be necessary.
There is no detailed discussion about whether PASTE components themselves may be immunogenic or if they could alter cell surface markers and thereby impact immune recognition.
Potential of the technology to overcome the limitation:
The PASTE technology has the potential to mitigate immune responses, especially if compatible with AAV vectors known for low immunogenicity, and assuming that the PASTE components themselves do not provoke significant immune reactions. However, this potential is primarily speculative at this stage and requires in-depth investigation.
Evidence within the paper:
The PASTE technology potentially enables the integration of large sequences at targeted genomic locations, which might be beneficial in addressing genetic complexity by allowing the insertion or replacement of multigene segments. The key aspects from the paper concerning this are:
PASTE's ability to integrate sequences as large as ~36 kb at multiple genomic loci across cell types suggests it can be utilized to deliver complex genetic cassettes required to address multigenic disorders.
The precision of PASTE, facilitated by fusion with serine integrases, can potentially prevent mosaic outcomes, as it promotes site-specific integration rather than random insertion.
PASTE was shown to perform multiplexed gene integration by introducing three genes at three separate loci, indicating its capacity for complex genetic manipulations.
The successful use of PASTE in non-dividing primary human hepatocytes indicates that it might overcome issues related to cell division, which can contribute to mosaicism in gene-edited populations.
Strengths of the evidence:
The ability to deliver large genetic sequences implies that PASTE could be utilized for complex genetic engineering scenarios, including multigene disorders.
The demonstrated multiplexing capability is a significant advantage when tackling diseases caused by mutations in multiple genes or those that require the introduction of several genetic elements.
Weaknesses of the evidence:
While the paper describes the introduction of large and multiplex edits, the functional outcomes of these edits and their potential to address complex genetic diseases are not characterized.
The extent to which PASTE avoids mosaicism has not been thoroughly demonstrated. While site-specific insertions might increase homogeneity, comprehensive analysis of editing outcomes at the cellular population level was not presented.
There is no discussion on the regulation of the inserted genes, which is crucial for diseases with complex regulatory networks that cannot be addressed by gene insertion alone.
Potential of the technology to overcome the limitation:
PASTE's potential to accommodate larger genetic payloads and enable multiplex editing suggests it could be used to address genetic complexity in diseases. Moreover, its site-specific nature might reduce mosaicism. However, the actual performance of PASTE in tackling complex genetic diseases and the prevention of mosaicism need to be proven in disease-relevant model systems, particularly for polygenic conditions.
Evidence within the paper:
The paper provides some insights into the long-term effects and stability of the gene editing technology known as PASTE:
PASTE does not rely on DSBs or HDR, suggesting that the edits introduced should, in theory, be more stable due to the avoidance of error-prone repair pathways.
Multiplexed gene integration capability is shown, which—if stable—could demonstrate PASTE’s potential for long-lasting genetic modification.
For evaluating the stability of the integration, the authors present data where they test the targeting of genes with larger cargos (up to ~36 kb) and data showing in vivo experiments in liver-humanized mice.
PASTE is said to have fewer detectable off-target events, which could point to less genomic instability overall post-editing.
Strengths of the evidence:
The technology's ability to integrate large DNA fragments without relying on error-prone DNA repair mechanisms enhances the prospect of achieving stable genetic modifications.
The paper provides initial in vivo validation in liver-humanized mice, which indicates the potential for long-term stability of the edits.
Weaknesses of the evidence:
The publication primarily focuses on the efficiency and precision of the PASTE technology, with less emphasis on the long-term implications of genomic alterations.
There is limited information on the consistency of edit stability through multiple rounds of cell division, particularly in tissues with high cellular turnover.
Findings from the liver-humanized mice cannot be directly extrapolated to humans or to other tissues and cell types.
The evidence does not explicitly address the persistence of edits in a clinical context, nor does it mention the impact on epigenetics or the possible genotoxic effects of the integrated DNA sequences over an extended period.
Potential of the technology to overcome the limitation:
PASTE potentially provides a stable alternative for gene editing, especially for permanent correction of genetic disorders. Its design minimizes reliance on cellular DNA repair pathways, which can increase the fidelity and stability of edits. However, the long-term effects, particularly regarding safety, stability across cell divisions, and in different tissues, require further validation.
Evidence within the paper:
The PASTE technology described in the paper addresses some of the limitations associated with base and prime editing, as detailed below:
Large Payload Integration: PASTE demonstrates its capability to integrate large sequences (~36 kb) with efficiencies up to ~50-60% in cell lines, which surpasses the current capabilities of base and prime editors that are restricted to small nucleotide changes and short insertions or deletions.
Editing Versatility: The paper outlines PASTE’s ability to efficiently target multiple loci for integration, suggesting a potential advantage over base editing’s limited editing window. PASTE's use of integrases with various recognized attachment sites also presents an opportunity to target distinct loci within the genome.
Multiplexed Editing: The paper shows PASTE performing multiplexed integration by simultaneously introducing three genes at three separate loci, which is beyond the current scope of base or prime editing that typically targets single loci.
Strengths of the evidence:
The multi-kb insertions capability of PASTE is a significant advance over base and prime editing's limits in terms of the size and complexity of genetic material that can be added to the genome.
PASTE’s versatility in targeting and efficiency points towards substantial improvements over prime editing in these respects.
Weaknesses of the evidence:
The paper doesn’t compare PASTE directly with base editors or prime editors, so there’s no direct evidence that PASTE is more effective in scenarios where base or prime editing would traditionally be used (e.g., single or few base pair edits).
While prime editing is known to be less efficient, PASTE does not elucidate its efficiency rates in comparison to prime editing at a granular level.
The complexity of PASTE’s delivery is acknowledged, involving multiple components (Cas9 nickase, reverse transcriptase, integrase). This added complexity could be comparable to prime editing’s more intricate system, but the paper does not discuss this aspect in detail.
Potential of the technology to overcome the limitation:
PASTE appears to have the capacity to overcome some of the challenges specific to base and prime editing, particularly those related to the size of the editable genetic material and the ability to perform multiplexed edits. Its approach, which circumvents DSBs, also stands to potentially offer a more precise and less-damaging method of genome modification.
View our growing database of research articles on innovative biotech startups. The analysis is conducted by AI.
Tome says its initial therapeutic focus is on gene therapy for monogenic liver disease and cell therapy for autoimmune disease.
The rationale for using gene therapy and gene editing in treating liver diseases is based on several key factors. The liver has a remarkable ability to regenerate, making it a resilient target for gene therapy. Modified cells can proliferate, enhancing the effectiveness of the treatment. The liver plays a crucial role in metabolism, including protein synthesis and detoxification. Correcting genetic defects in liver cells can restore normal function and prevent systemic complications. The liver is readily accessible for gene delivery, especially through the bloodstream. This makes it easier to deliver genetic material or gene-editing tools directly to liver cells. The liver has a unique immune-tolerant environment, which can reduce the likelihood of immune reactions against the introduced genetic material, making gene therapy safer and more effective.
TOME Biosciences' Programmable Genomic Integration (PGI) technology, particularly advantageous for treating monogenic liver diseases, could excel in addressing conditions that are challenging for existing gene editing technologies like CRISPR/Cas9, base editing, and prime editing. The key advantages of PGI, such as the ability to insert large DNA sequences with high precision and avoid double-strand DNA breaks, position it uniquely for certain diseases. Here are a few monogenic liver diseases where Tome's technology might have a particular advantage:
Disease Name | Description | Associated Gene(s) | Standard of Care | Prognosis | Unmet Need |
---|---|---|---|---|---|
Alpha-1 Antitrypsin Deficiency (AATD) | A genetic disorder causing liver and lung disease due to alpha-1 antitrypsin protein deficiency. | SERPINA1 | Augmentation therapy, bronchodilators, and pulmonary rehabilitation for lung issues; liver transplantation in severe cases. | Variable; lung disease can be severe; liver disease may lead to cirrhosis; not all patients develop liver disease. | Effective treatments for lung and liver aspects; gene therapy to correct underlying genetic defect. |
Wilson's Disease | Copper accumulation in liver, brain, and other organs, leading to liver disease and neurological symptoms. | ATP7B | Chelating agents to remove excess copper, zinc therapy, and in severe cases, liver transplantation. | Good with early treatment; poor if untreated leading to liver failure or severe neurological damage. | Therapies with fewer side effects; treatments addressing neurological symptoms more effectively; earlier diagnosis. |
Hereditary Hemochromatosis | Excessive iron absorption causing liver damage, heart problems, and diabetes. | HFE, HJV, HAMP, TFR2 | Phlebotomy, iron chelation, and management of complications like liver disease and diabetes. | Generally good with early diagnosis and treatment; can be poor if complications like cirrhosis develop. | Non-invasive diagnostic methods; broader screening; therapies to prevent organ damage. |
Citrullinemia Type I | Ammonia accumulation due to urea cycle deficiency. | ASS1 | Low-protein diet, medications to remove ammonia, liver transplantation in severe cases. | Variable; can be severe with neurological damage if untreated; better with early intervention. | Effective long-term treatments; gene therapies to address the underlying cause. |
Progressive Familial Intrahepatic Cholestasis (PFIC) | Rare liver disorders disrupting bile flow in children, leading to liver damage. | ATP8B1, ABCB11, ABCB4 | Medications to reduce itching and cholestasis, vitamin supplementation, liver transplantation in severe cases. | Variable; can progress to liver failure requiring transplantation. | Treatments to prevent progression; better management of symptoms. |
Glycogen Storage Diseases | Disorders affecting sugar use and glycogen storage, causing liver enlargement and metabolic problems. | Various, including G6PC, SLC37A4, AGL, GBE1, GAA, PYGL, PFKM | Dietary management, supplements, medications to control symptoms, liver transplantation for specific types. | Depends on type; can range from relatively mild to life-threatening. | Therapies to correct the underlying metabolic defects; treatments to improve quality of life; gene therapies. |
B cells play a crucial role in the immune system, including the production of antibodies and presentation of antigens. In autoimmune diseases, these cells can become dysregulated, attacking the body's own tissues. Cell therapy can be designed to selectively target and modulate these aberrant B cell functions, restoring normal immune system balance.
Traditional treatments for autoimmune diseases often involve broad-spectrum immunosuppressants, which can have significant side effects and increase infection risk. Cell therapies could offer a more targeted approach, potentially reducing the need for long-term systemic immunosuppression.
Cell therapies, especially those that are genetically edited, can be engineered to target specific B cell populations or pathways involved in the autoimmune process. This specificity has the potential to increase the efficacy of the treatment compared to broader immunosuppressive therapies.
However, there are several limitations and challenges with the current approaches to cell therapy for B cell autoimmune diseases:
Tome Biosciences' Programmable Genomic Integration (PGI) technology could potentially overcome several limitations of current approaches in treating B cell autoimmune diseases:
In conclusion, Tome's PGI technology addresses key challenges in treating B cell autoimmune diseases, including precision, safety, efficiency, and the potential for curative therapies. It represents a significant advancement in the field of cell and gene therapy, particularly for complex conditions like autoimmune diseases.
View our growing database of research articles on innovative biotech startups. The analysis is conducted by AI.
Analyzing performance of Series A VCs
Valuations of biotech startups from Series A to IPO
Bay Bridge Bio Startup Database
How to value biotech companies
Then consider joining our mailing list. I periodically publish data-driven articles on the biotech startup and VC world.