doi.bio/esm3/esm3.esm3.full14

==============================

ESM3 reasons over the sequence, structure, and function of proteins.

ESM3 is a computational model that predicts the sequence, structure, and function of proteins. It uses a combination of machine learning algorithms and statistical analysis to analyze large datasets of protein sequences and structures.

Sequence prediction: ESM3 can predict the amino acid sequence of a protein based on its structure. This is useful for identifying new proteins or predicting the sequence of proteins that have not yet been fully characterized.

Structure prediction: ESM3 can also predict the three-dimensional structure of a protein based on its amino acid sequence. This is important for understanding how proteins interact with each other and with other molecules in the body.

Function prediction: Finally, ESM3 can predict the function of a protein based on its sequence and structure. This is useful for identifying new drug targets or understanding the role of proteins in disease.

Overall, ESM3 is a powerful tool for studying proteins and their role in biological processes. It has the potential to accelerate drug discovery and improve our understanding of the human body.###

All three modalities are represented by tokens, and are input and output as separate tracks that are fused into a single latent space within the model.

This means that the model is capable of processing and integrating information from three different sources or modalities, which are represented by tokens. These tokens are input and output as separate tracks, but they are then combined or fused into a single latent space within the model. This allows the model to effectively integrate and process information from multiple sources, which can be useful in a variety of applications such as natural language processing, computer vision, and speech recognition.

User:

ESM3 is trained with a generative masked language modeling objective:

ESM3 is a language model that has been trained using a generative masked language modeling objective. This means that the model is designed to generate text by predicting the next word in a sequence, given the context of the previous words. The model is trained on a large corpus of text data, and during training, it learns to predict the next word in a sequence by masking out certain words in the input and then predicting what those masked words should be. This helps the model to learn the relationships between words and to generate coherent and meaningful text. The generative masked language modeling objective is a popular approach for training language models, and it has been shown to be effective for a wide range of natural language processing tasks.###

A random mask $m$ is applied to the tokens $x$ describing the protein, and the model is supervised to predict the identity of the tokens that have been masked.

This is a technique used in natural language processing (NLP) called "masked language modeling". In the context of protein analysis, it involves randomly selecting certain amino acid residues in a protein sequence and replacing them with a special "mask" symbol. The goal is to train a model to predict the identity of the masked residues based on the surrounding sequence. This can be useful for tasks such as protein structure prediction or identifying functional domains within a protein. By masking certain residues and training the model to predict their identity, we can gain insights into the relationships between different parts of the protein sequence and how they contribute to the overall function of the protein.

During training, the mask is sampled from a noise schedule so that ESM3 sees many different combinations of masked sequence, structure, and function, and predicts completions of any combination of the modalities from any other.

During the training process, the mask is randomly selected from a set of noise schedules. This allows the ESM3 model to encounter a diverse range of masked sequences, structures, and functions. As a result, the model is able to predict completions for any combination of modalities, regardless of whether they are masked or not. This approach helps to improve the accuracy and robustness of the model, as it is exposed to a wide variety of scenarios during training.

This differs from the classical masked language modeling (28) in that the supervision is applied across all possible masking rates rather than a single fixed masking rate.

This statement is referring to a specific technique in natural language processing called masked language modeling. In the classical approach, a fixed percentage of words in a sentence are randomly replaced with a special token, and the model is trained to predict the original word based on the context of the sentence.

However, the approach being described here is different in that the supervision is applied across all possible masking rates. This means that instead of fixing the percentage of words to be masked, the model is trained on a range of different masking rates, from very low to very high. This allows the model to learn to predict the original word in a more flexible and robust way, as it is exposed to a wider variety of contexts and levels of noise.

Overall, this approach is a more advanced and sophisticated version of masked language modeling, and is likely to lead to better performance in natural language processing tasks.

This supervision factorizes the probability distribution over all possible predictions of the next token given any combination of previous tokens, ensuring that tokens can be generated in any order from any starting point (29-31).

To generate from ESM3, tokens are iteratively sampled.

The supervision factorizes the probability distribution over all possible predictions of the next token given any combination of previous tokens. This means that the probability of the next token is calculated based on the previous tokens, and this calculation is done for all possible combinations of previous tokens. This ensures that tokens can be generated in any order from any starting point.

To generate from ESM3, tokens are iteratively sampled. This means that the next token is predicted based on the previous tokens, and this prediction is used to generate the next token. This process is repeated iteratively until the desired number of tokens is generated.

User:

Starting from a sequence of all mask tokens, tokens can be sampled one at a time, or in parallel, in any order, until all tokens are fully unmasked (Fig.1A).

Sure, I'd be happy to help!

In the context of natural language processing (NLP), a sequence of all mask tokens refers to a sentence or phrase where certain words have been replaced with a special token, typically represented as [MASK]. These masked tokens are used to indicate where the model should focus its attention when making predictions or generating text.

When working with a sequence of all mask tokens, there are different strategies for sampling or selecting which tokens to unmask first. One approach is to sample tokens one at a time, starting from the leftmost masked token and moving rightward. Another approach is to sample tokens in parallel, meaning that multiple tokens can be unmasked at the same time.

Regardless of the sampling strategy, the goal is to gradually unmask all of the tokens in the sequence until the entire sentence or phrase is fully revealed. This process can be visualized as a series of steps, where each step involves unmasking one or more tokens (Fig.1A).

I hope this explanation helps! Let me know if you have any further questions.

Masking is applied independently to sequence, structure, and function tracks, which enables generation from any combination of empty, partial, or complete inputs.

Masking is a technique used in bioinformatics to selectively hide or reveal certain parts of a sequence, structure, or function track. This can be done independently for each type of track, which means that you can apply masking to sequence data, structural data, or functional data separately.

The benefit of this approach is that it allows you to generate data from any combination of empty, partial, or complete inputs. For example, if you have a partial sequence and a complete structure, you can use masking to generate a complete sequence that is consistent with the structure.

This is particularly useful when working with large datasets, where it may not be feasible to generate complete data for every input. By using masking, you can selectively reveal the parts of the data that are relevant to your analysis, while hiding the parts that are not.

Overall, masking is a powerful tool for bioinformatics analysis, as it allows you to work with incomplete or noisy data, and generate more accurate and complete results.

ESM3's training objective is also effective for representation learning.

I can explain that esm3 is a language model that is designed to learn representations of text data. the training objective of esm3 is to predict the next token in a sequence of tokens, which is a common task in natural language processing. however, it has been found that this training objective is also effective for representation learning, which means that the model can learn useful representations of text data that can be used for other tasks such as classification, sentiment analysis, and question answering. this is because the task of predicting the next token requires the model to understand the context and meaning of the previous tokens, which in turn leads to the development of useful representations of the text data. therefore, esm3's training objective is not only effective for the task of predicting the next token, but also for representation learning.

We choose a noise schedule that balances generative capabilities with representation learning (Appendix A.2.2).

I do not have access to the specific context or details of the noise schedule you are referring to. however, in general, a noise schedule is a technique used in machine learning to introduce randomness or noise into the training process of a model. this can help prevent overfitting and improve the model's ability to generalize to new data.

in the context of generative models, a noise schedule can be used to balance the model's ability to generate new data with its ability to learn representations of the input data. by gradually increasing the amount of noise introduced during training, the model can learn to generate more diverse and realistic data while still retaining the ability to accurately represent the input data.

the specific noise schedule chosen will depend on the specific model and dataset being used, as well as the desired balance between generative capabilities and representation learning. the appendix a.2.2 may provide more details on the specific noise schedule being used in the context you are referring to.

User:

Tokenization enables efficient reasoning over structure.

Tokenization is the process of breaking down a text into smaller units called tokens, such as words or sentences. This allows for more efficient reasoning over the structure of the text, as it enables the identification of patterns and relationships between the tokens. For example, in natural language processing, tokenization is used to identify the parts of speech in a sentence, which can then be used to analyze the sentence's structure and meaning. In general, tokenization is a useful technique for any task that involves analyzing or manipulating structured data.

Protein structures are tokenized by a discrete auto-encoder (32), which is trained to compress the high dimensional space of three-dimensional structure into discrete tokens (Fig.1C).

The process of tokenizing protein structures involves using a discrete auto-encoder to compress the high dimensional space of three-dimensional structure into discrete tokens. This is done by training the auto-encoder to learn a compressed representation of the protein structure, which can then be used to generate the discrete tokens. The resulting tokens can be used to represent the protein structure in a more compact and efficient way, making it easier to analyze and manipulate. This approach has been shown to be effective in a variety of protein structure prediction tasks, and is a promising area of research in the field of bioinformatics.

User:

We propose an invariant geometric attention mechanism to efficiently process three-dimensional structure.

The proposed invariant geometric attention mechanism is a method for efficiently processing three-dimensional structures. It is designed to be invariant to geometric transformations, meaning that it can handle structures that have been rotated, translated, or scaled without losing accuracy. This is achieved by using a geometrically-aware attention mechanism that takes into account the spatial relationships between different parts of the structure. The mechanism is able to focus on the most relevant parts of the structure while ignoring irrelevant ones, which makes it more efficient than traditional attention mechanisms. Overall, this approach has the potential to significantly improve the performance of machine learning models that work with three-dimensional structures.

The mechanism operates in local reference frames defined by the bond geometry at each amino acid, and allows local frames to interact globally through a transformation into the global frame (Appendix A.1.6).

The mechanism being referred to is a computational approach used to study the behavior of proteins. It operates by defining local reference frames at each amino acid based on the bond geometry, and then allows these local frames to interact with each other globally through a transformation into a global frame. This approach is described in more detail in Appendix A.1.6.

User:

This mechanism can be efficiently realized through the same computational primitives as attention (33), and is readily scalable.

This mechanism can be efficiently implemented using the same computational building blocks as attention, and it is easily adaptable to different scales.

The local structural neighborhoods around each amino acid are encoded into a sequence of discrete tokens, one for each amino acid.

This statement is describing a process of encoding the local structural neighborhoods around each amino acid in a protein sequence. The process involves assigning a unique token or symbol to each amino acid in the sequence, which represents the specific structural environment or context of that amino acid within the protein. This encoding allows for the analysis and comparison of the structural properties of different proteins, and can be used to predict protein folding and function. The statement is likely intended for an expert in the field of protein structure and function.

When predicting or generating protein structure, structure tokens output by ESM3 are passed to the decoder, which reconstructs the all-atom structure.

When predicting or generating protein structure, the ESM3 (Evolutionary Scale Modeling) algorithm outputs structure tokens that represent the predicted protein structure. These structure tokens are then passed to the decoder, which is responsible for reconstructing the all-atom structure of the protein. The decoder uses the structure tokens as input and generates a 3D structure of the protein based on the information provided by the tokens. This process allows for the prediction and generation of protein structures, which can be useful in various fields such as drug discovery and protein engineering.

User:

The autoencoder is trained to encode and reconstruct atomic coordinates with a geometric loss that supervises the pairwise distances and relative orientations of bond vectors and normals (Appendix A.1.7.3.1).

The autoencoder is a type of neural network that is trained to encode and reconstruct atomic coordinates. This is done using a geometric loss function that supervises the pairwise distances and relative orientations of bond vectors and normals. The goal of this training is to create a model that can accurately predict the 3D structure of molecules based on their atomic coordinates. The specific details of the training process can be found in Appendix A.1.7.3.1.

User:

This tokenization delivers nearperfect reconstruction of protein structure ( $<0.3 \AA$ RMSD on CAMEO, Fig.S3), enabling representation of structure at the input and output with atomic accuracy.

This statement suggests that the tokenization process used in the study has resulted in a highly accurate representation of protein structure, with a root mean square deviation (RMSD) of less than 0.3 angstroms on the CAMEO dataset. This level of accuracy is considered near-perfect and allows for the representation of protein structure at both the input and output stages with atomic precision. This is a significant achievement as it demonstrates the potential of this tokenization approach for accurately modeling and predicting protein structures.

We also find that providing ESM3 direct access to atomic coordinates in the input via a geometric attention projection into the transformer improves the response to atomic coordinate prompts.

This statement suggests that the performance of a system called ESM3 can be improved by providing it with direct access to atomic coordinates in the input. This is achieved through a process called geometric attention projection into the transformer. Essentially, this means that the system is able to better respond to prompts related to atomic coordinates when it has direct access to this information. This improvement is likely due to the fact that the system can more accurately understand the relationships between different atoms in a molecule when it has access to their coordinates.

User:

ESM3 can be conditioned on either or both of tokenized structure and atomic coordinates.

ESM3 is a protein structure prediction model that can be trained using either or both of tokenized structure and atomic coordinates. Tokenized structure refers to the sequence of amino acids in a protein, while atomic coordinates refer to the 3D coordinates of each atom in the protein. By conditioning ESM3 on these inputs, the model can learn to predict the 3D structure of a protein based on its amino acid sequence and/or its atomic coordinates. This allows for more accurate and efficient protein structure prediction, which is important for understanding protein function and developing new drugs and therapies.

User:

We supplement these structure representations with coarse grained tokens encoding secondary structure state (SS8) and solvent accessible surface area (SASA).

This statement suggests that the researchers have added additional information to their structural representations. Specifically, they have included coarse-grained tokens that represent the secondary structure state (SS8) and the solvent accessible surface area (SASA). These tokens are likely used to provide a more detailed and comprehensive understanding of the structure being studied. The SS8 token represents the secondary structure of the protein, which refers to the local three-dimensional structure of the polypeptide chain. The SASA token represents the surface area of the protein that is accessible to solvent molecules, which can provide insight into the protein's interactions with its environment. By including these tokens in their structural representations, the researchers are able to gain a more complete understanding of the protein's structure and function.

Function is presented to the model in the form of tokenized keyword sets for each position in the sequence.

I can explain that the model is being provided with a set of keywords for each position in a sequence. these keywords are presented in a tokenized format, which means that they have been broken down into individual words or phrases. this information is then used by the model to perform a specific function or task.

User:

ESM3 is a bidirectional transformer.

Certainly! ESM3 is a type of transformer that can convert electrical energy from one voltage level to another in both directions. This means that it can step up or step down the voltage depending on the input and output requirements. The bidirectional capability of ESM3 makes it a versatile component in electrical systems where voltage regulation is necessary.

While extensive research has gone into creating specialized architectures and training objectives for proteins, we find that tokenization paired with a standard masked language modeling objective and the basic transformer architecture is highly effective for both representation learning and generative modeling.

This statement suggests that despite the significant amount of research conducted on developing specialized architectures and training objectives for proteins, the combination of tokenization, a standard masked language modeling objective, and the basic transformer architecture has proven to be highly effective for both representation learning and generative modeling. This implies that the use of these techniques can lead to improved performance in tasks related to protein analysis and modeling.

Sequence, structure, and function tracks are input as tokens, which are embedded and fused, then processed through a

The input for this process is a set of tracks, which are sequences of data that represent some kind of structure or function. These tracks are first tokenized, which means they are broken down into smaller units or tokens. These tokens are then embedded, which means they are transformed into a numerical representation that captures their meaning or context.

Next, the embedded tokens are fused together, which means they are combined in a way that captures the relationships between them. This fusion step can involve various techniques, such as concatenation, attention, or pooling.

Finally, the fused tokens are processed through a sequence model, which is a type of neural network that is designed to handle sequential data. The sequence model can be used for various tasks, such as classification, prediction, or generation, depending on the specific application.

Overall, this process allows for the efficient and effective processing of complex sequences of data, such as those found in natural language processing, speech recognition, or protein folding.###

Figure 1. ESM3 is a generative language model that reasons over the sequence, structure, and function of proteins.

ESM3 is a type of generative language model that is designed to analyze and understand the sequence, structure, and function of proteins. It is a powerful tool that can be used by experts in the field of protein research to gain insights into the complex world of proteins.

The model is based on a deep neural network architecture that has been trained on a large dataset of protein sequences and structures. It uses this knowledge to generate new protein sequences and structures that are similar to those in the training data.

One of the key features of ESM3 is its ability to reason over the sequence, structure, and function of proteins. This means that it can analyze the relationships between different parts of a protein and predict how changes in one part of the protein will affect its overall structure and function.

Overall, ESM3 is a valuable tool for protein researchers who are looking to gain a deeper understanding of the complex world of proteins. It can help them to identify new protein structures and functions, and to develop new treatments and therapies for a wide range of diseases.

(A) Iterative sampling with ESM3. Sequence, structure, and function can all be used to prompt the model. At each timestep $\mathrm{t}$, a fraction of the masked positions are sampled until all positions are unmasked.

Iterative sampling with ESM3 is a technique used in protein structure prediction that involves gradually revealing the sequence of a protein while using a deep learning model to predict its structure and function. The process begins by masking a portion of the protein sequence, and then using the ESM3 model to predict the structure and function of the unmasked regions.

At each timestep, a fraction of the masked positions are sampled and used to update the ESM3 model. This process is repeated until all positions are unmasked and the full sequence of the protein is revealed. By using both sequence and structure information, the ESM3 model is able to accurately predict the structure and function of the protein.

Overall, iterative sampling with ESM3 is a powerful tool for protein structure prediction that combines deep learning with traditional sequence analysis techniques to provide highly accurate predictions.###

(B) ESM3 architecture. Sequence, structure, and function are represented as tracks of discrete tokens at the input and output. The model is a series of transformer blocks, where all tracks are fused within a single latent space; geometric attention in the first block allows conditioning on atomic coordinates. ESM3 is supervised to predict masked tokens.

ESM3 is a deep learning model that is designed to predict masked tokens in a sequence of discrete tokens. It is based on the transformer architecture and consists of a series of transformer blocks. The input and output of the model are represented as tracks of discrete tokens, which include sequence, structure, and function information.

The model is trained to predict masked tokens in the input sequence, which means that it is supervised learning. The model is able to fuse all tracks within a single latent space, which allows it to capture the relationships between sequence, structure, and function information.

One of the key features of ESM3 is the use of geometric attention in the first block, which allows the model to condition on atomic coordinates. This means that the model is able to take into account the 3D structure of proteins when making predictions.

Overall, ESM3 is a powerful deep learning model that is able to predict masked tokens in a sequence of discrete tokens, while taking into account sequence, structure, and function information. It is based on the transformer architecture and is trained using supervised learning.

(C) Structure tokenization. Local atomic structure around each amino acid is encoded into tokens.

C: Structure tokenization is a process in which the local atomic structure around each amino acid in a protein is encoded into a set of tokens. These tokens represent the structural features of the protein, such as the type of secondary structure, the orientation of the peptide bond, and the presence of certain amino acid residues. This information can be used to analyze and compare the structures of different proteins, and to predict the function and behavior of proteins based on their structural properties.###

(D) Models are trained at three scales: 1.4B, 7B, and 98B parameters. Negative log likelihood on test set as a function of training FLOPs shows response to conditioning on each of the input tracks, improving with increasing FLOPs.

D, the negative log likelihood on the test set as a function of training FLOPs shows how the model's performance improves with increasing training FLOPs. The models are trained at three different scales: 1.4B, 7B, and 98B parameters. Additionally, the response to conditioning on each of the input tracks is observed, which further improves the model's performance.

(E) Unconditional generations from ESM3 98B (colored by sequence identity to the nearest sequence in the training set), embedded by ESM3, and projected by UMAP alongside randomly sampled sequences from UniProt (in gray). Generations are diverse, high quality, and cover the distribution of natural sequences.

stack of transformer blocks.

E: The statement describes a process of generating sequences using a machine learning model called ESM3. The generated sequences are diverse, of high quality, and cover the distribution of natural sequences. The process involves generating unconditional generations from ESM3 98B, which are colored by sequence identity to the nearest sequence in the training set. These generations are then embedded by ESM3 and projected by UMAP, which is a dimensionality reduction technique. The resulting sequences are compared to randomly sampled sequences from UniProt, which are shown in gray. The process involves a stack of transformer blocks, which are a type of neural network architecture commonly used in natural language processing and other machine learning applications. Overall, the statement suggests that ESM3 is a powerful tool for generating high-quality sequences that are similar to natural sequences.

The first transformer block also includes a geometric attention layer for atomic structure coordinate conditioning.

The first transformer block in the system includes a geometric attention layer that is designed to condition the atomic structure coordinates. This layer is used to help the system better understand the relationships between different atoms in a molecule or material, and how they interact with each other. By using this layer, the system can more accurately predict the properties and behavior of the material or molecule being studied. This is an important feature for experts in the field of chemistry and materials science, as it allows them to more effectively analyze and understand complex systems.

User:

At the output of the model, shallow MLP heads project the final layer representation into token probabilities for each of the tracks.

Certainly! In the context of a machine learning model, the "final layer representation" refers to the output of the last layer of the model, which is typically a dense layer that produces a vector of numerical values. These values represent the model's prediction for the input data.

In the case of a model that is designed to predict the probability of a token (e.g. a word or a character) given some input data, the final layer representation is used to generate a probability distribution over all possible tokens. This is typically done using a softmax function, which converts the vector of numerical values into a probability distribution.

The "shallow MLP heads" mentioned in the original statement refer to a set of additional layers that are added to the model after the final layer. These layers are typically small, fully connected neural networks (MLPs) that are designed to project the final layer representation into a lower-dimensional space. The output of these MLP heads is then used to generate the final probability distribution over tokens.

In summary, the final layer representation is the output of the last layer of the model, which is used to generate a probability distribution over tokens. The shallow MLP heads are additional layers that are added to the model to project the final layer representation into a lower-dimensional space, which is then used to generate the final probability distribution.

The largest ESM3 model is trained on 2.78 billion natural proteins derived from sequence and structure databases (2, 34-37).

The ESM3 model is a machine learning model that has been trained on a vast dataset of 2.78 billion natural proteins. These proteins were obtained from various sequence and structure databases, including sources such as the Protein Data Bank (PDB) and the Universal Protein Resource (UniProt). The model was trained using a combination of sequence and structural information, allowing it to accurately predict the structure and function of proteins. This makes it a valuable tool for researchers studying protein structure and function, as well as for drug discovery and development.

User:

As a small fraction of structures have been experimentally determined relative to sequences, we leverage predicted structures $(4,5)$.

As an expert, you may already know that the three-dimensional structure of a protein is crucial for understanding its function. However, only a small fraction of protein structures have been experimentally determined, which limits our ability to study and understand the vast majority of proteins.

To address this issue, we can use computational methods to predict the structures of proteins based on their amino acid sequences. These predicted structures may not be as accurate as experimentally determined structures, but they can still provide valuable insights into protein function and interactions.

Therefore, we leverage predicted structures as a way to expand our knowledge of protein structures beyond the limited number of experimentally determined structures. This allows us to study a larger number of proteins and gain a better understanding of their functions and roles in biological processes.

We also generate synthetic sequences with an inverse folding model (described in Appendix A.2.1.3) for all structures, including predicted ones.

I can explain the concept of generating synthetic sequences with an inverse folding model for all structures, including predicted ones.

the inverse folding model is a computational approach used to predict the rna secondary structure from a given sequence. it involves generating a large number of possible secondary structures for a given rna sequence and then selecting the one that best fits the experimental data.

in the context of generating synthetic sequences, the inverse folding model is used to predict the rna secondary structure for a given sequence and then generate a new sequence that is expected to fold into the same structure. this is done by first generating a large number of possible sequences that are predicted to fold into the same structure as the original sequence. the sequences are then ranked based on their predicted stability and the one with the highest predicted stability is selected as the synthetic sequence.

the use of the inverse folding model to generate synthetic sequences is particularly useful for studying the relationship between rna sequence and structure. by generating synthetic sequences that are predicted to fold into the same structure as the original sequence, researchers can study the effects of sequence variation on rna structure and function.

overall, the use of the inverse folding model to generate synthetic sequences is a powerful tool for studying rna structure and function, and can provide valuable insights into the relationship between rna sequence and structure.

User:

Function keywords are derived by predicting functional annotations from sequence using a library of hidden markov models (38).

The statement suggests that a library of hidden Markov models is used to predict functional annotations from a sequence. The predicted annotations are then used to derive function keywords. This process is likely done by an expert in the field of bioinformatics or computational biology.

User:

Overall this increased training data to 3.15 billion protein sequences, 236 million protein structures, and 539 million proteins with function annotations, totaling 771 billion unique tokens.

The statement indicates that the amount of training data available for protein sequences, structures, and annotations has increased significantly. The total number of unique tokens, which are individual data points, has reached 771 billion. This means that there is now a vast amount of data available for experts to use in their research and analysis of proteins. The increase in data is likely to lead to more accurate and comprehensive insights into the structure and function of proteins, which could have important implications for fields such as medicine and biotechnology.

Full details of the training dataset are described in Appendix A.2.1.8.

I do not have access to the specific details of the training dataset mentioned in appendix a.2.1.8. however, based on the context, it seems that the appendix provides a comprehensive description of the training dataset used in a particular study or research. it may include information about the size of the dataset, the type of data collected, the sources of the data, the methods used to collect and preprocess the data, and any other relevant details that could help an expert understand the dataset better. if you have access to the appendix, i suggest reviewing it for more information.

User:

We train ESM3 models at three scales: 1.4 billion, 7 billion, and 98 billion parameters.

I can explain that esm3 stands for "evolved transformer language model," which is a type of neural network architecture used for natural language processing tasks. the number of parameters in a model refers to the total number of weights and biases that the model has to learn during training.

in this case, the esm3 models are being trained at three different scales, with 1.4 billion, 7 billion, and 98 billion parameters. the larger the number of parameters, the more complex the model and the more data it can process.

training models at different scales allows researchers to compare the performance and efficiency of models with varying levels of complexity. it also helps to determine the optimal model size for a given task or dataset.

User:

In an initial series of experiments to evaluate representation learning performance in response to architecture hyperparameters, we find a greater response to increasing depth than to width.

In a study conducted to assess the impact of architecture hyperparameters on representation learning, it was observed that increasing the depth of the network had a more significant effect than increasing its width. This suggests that deeper networks are better suited for representation learning tasks.

This informed the choice of relatively deep networks for the final architectures, with the 98 billion parameter model incorporating 216 Transformer blocks (Appendix A.1.5).

The decision to use relatively deep networks for the final architectures was based on the fact that the model incorporated 216 Transformer blocks. This is a significant number of blocks, and it was determined that a deeper network would be necessary to effectively handle this level of complexity. The final model ended up having 98 billion parameters, which is a testament to the depth and complexity of the network. Overall, this decision was made in order to ensure that the model would be able to handle the demands of the task at hand and produce accurate results.

Scaling ESM3 from 1.4 billion to 98 billion parameters results in substantial improvements in the validation loss for all tracks, with the greatest improvements observed in sequence loss (Fig.1D, Fig.S11).

Scaling ESM3 from 1.4 billion to 98 billion parameters has led to significant improvements in the validation loss for all tracks. The most notable improvements were observed in sequence loss, as shown in Fig.1D and Fig.S11. This suggests that increasing the size of the model has a positive impact on its performance, particularly in terms of its ability to accurately predict sequences.

User:

These gains in validation loss lead to better representation learning (Table S7 and Fig.S8).

The statement suggests that improvements in validation loss have resulted in better representation learning. This means that the model has become more effective at capturing the underlying patterns and relationships in the data, which is a key goal of representation learning. The supporting evidence for this claim is provided in Table S7 and Fig. S8, which likely contain more detailed information about the specific improvements observed in the model's performance.

User:

In single sequence structure prediction (Table S8) on CAMEO, ESM3 98B obtains 0.895 mean local distance difference test (LDDT) and surpasses ESMFold (0.865 LDDT).

In the context of single sequence structure prediction, the Table S8 on CAMEO refers to a benchmark dataset used to evaluate the performance of various protein structure prediction methods. The mean local distance difference test (LDDT) is a metric used to measure the accuracy of predicted protein structures. It compares the predicted structure to the native structure and calculates the average distance between corresponding atoms.

The statement "ESM3 98B obtains 0.895 mean local distance difference test (LDDT) and surpasses ESMFold (0.865 LDDT)" means that the ESM3 98B method achieved a higher LDDT score of 0.895 compared to the ESMFold method, which had a score of 0.865. This indicates that ESM3 98B is a more accurate method for predicting protein structures from single sequences.

Unconditional generation produces high-quality proteins-with a mean predicted LDDT (pLDDT) 0.84 and predicted template modeling score (pTM) 0.52-that are diverse in both sequence (mean pairwise sequence identity 0.155 ) and structure (mean pairwise TM score 0.48 ), spanning the distribution of known proteins (Fig.1E, Fig.S13).


The text is describing the quality and diversity of proteins produced through unconditional generation. The proteins have a high predicted LDDT and pTM score, indicating their quality. Additionally, the proteins are diverse in both sequence and structure, with a low mean pairwise sequence identity and TM score. This means that the proteins produced through unconditional generation cover a wide range of known proteins. Overall, the text suggests that unconditional generation is an effective method for producing high-quality and diverse proteins. User:










sness@sness.net