Tome Biosciences investment analysis

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.


Overview

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.



Highlights and risks


Highlights

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

Risks

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


Valuation


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.


Platform overview


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:


Advantages over other gene editing techniques


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.


Evaluating the technology


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.


Addressing the limitations of gene editing


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:


Potential of PGI to address limitations


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:


Delivery into Target Cells and Tissues


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:

Strengths of the evidence:

Weaknesses of the evidence:

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.


Immune Response


Evidence within the paper:

The paper mentions the consideration of immune responses to the components of the PASTE system, as outlined below:

Strengths of the evidence:

Weaknesses of the evidence:

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.


Genetic Complexity and Mosaicism


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:

Strengths of the evidence:

Weaknesses of the evidence:

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.


Long-term Effects and Stability


Evidence within the paper:

The paper provides some insights into the long-term effects and stability of the gene editing technology known as PASTE:

Strengths of the evidence:

Weaknesses of the evidence:

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.


Base Editing and Prime Editing Specific Challenges


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:






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