Skip to main content
Bioinformatics and Biology Insights logoLink to Bioinformatics and Biology Insights
. 2025 Sep 2;19:11779322251358314. doi: 10.1177/11779322251358314

Language Modelling Techniques for Analysing the Impact of Human Genetic Variation

Megha Hegde 1,, Jean-Christophe Nebel 1, Farzana Rahman 1
PMCID: PMC12409042  PMID: 40917099

Abstract

Interpreting the effects of variants within the human genome and proteome is essential for analysing disease risk, predicting medication response, and developing personalised health interventions. Due to the intrinsic similarities between the structure of natural languages and genetic sequences, natural language processing techniques have demonstrated great applicability in computational variant effect prediction. In particular, the advent of the Transformer has led to significant advancements in the field. However, transformer-based models are not without their limitations, and a number of extensions and alternatives have been developed to improve results and enhance computational efficiency. This systematic review investigates over 50 different language modelling approaches to computational variant effect prediction over the past decade, analysing the main architectures, and identifying key trends and future directions. Benchmarking of the reviewed models remains unachievable at present, primarily due to the lack of shared evaluation frameworks and data sets.

Keywords: Variant effect prediction, large language models, small language models, evolution of language models, genomics

Introduction

Understanding the impact of genetic variants is crucial for unravelling gene regulation mechanisms and disease causality. As we enter the era of personalised medicine, it has become of great interest to understand how an individual’s genetic makeup can impact their risk of developing a particular disease or their response to a specific treatment or medication.1,2

Any change in a coding region can directly affect the function of the associated protein; hence, certain gene mutations can be linked with specific diseases. While Mendelian (monogenic) diseases, such as cystic fibrosis and haemophilia, are caused by mutations in a single gene,3,4 polygenic diseases, including many cancers,5,6 result from combinations of mutations.7,8 Variation in the non-coding region of the genome is more challenging to interpret than that in the coding region, as variants impact disease-related genes by altering processes such as transcription, chromatin folding, or histone modification.9,10

Indeed, linguistic metaphors, from alphabets to grammars, have been readily used to describe the molecular world since the discovery of the structure of DNA in the 1950s.11,12 For instance, as genetic sequences are comprised of nucleotides or amino acids represented as letters, the sequences themselves can be represented as strings of letters, and processed in a way that is analogous to human language.13,14

Although Noam Chomsky formed the basis of modern language modelling in the 1950s, 15 the field has advanced considerably over the decades. A pivotal point was the development of the transformer in 2017, 16 which sparked a discernible shift towards the use of so-called large language models (LLMs) to solve a plethora of language modelling tasks in bioinformatics, including variant effect prediction.17,18 These LLMs are transformer-based models with billions of parameters, trained on large corpora of sequence data, and have been favoured due to their ability to accurately model long-range dependencies within sequences.19,20

Large language models have been used extensively in bioinformatics, and many excellent reviews have been published on several aspects. However, existing review papers either focus broadly on LLMs for general bioinformatics applications21-23 or on an overview of machine learning techniques for variant effect prediction. 18 Our review, however, focuses specifically on the applications of language models for variant effect prediction. In addition, we provide an in-depth analysis of LLMs, including post-transformer techniques, which are as yet underrepresented in the existing reviews. Hence, this review addresses this gap by first presenting an introduction to variant effect prediction and biological language modelling, before entering an in-depth exploration of language models applied to the prediction of effects of genetic variations within DNA, RNA, and protein sequences. Following a brief presentation of the history of language modelling, in line with the rapid advancement in the field, the core of the review covers models produced since the inception of the transformer in 2017. This review focuses on variants within the human genome and their impacts on disease causality; however, models trained on multi-species data are also considered.

Methods

This review details language modelling approaches to predicting the effects of variants (mutations) in DNA, RNA, and protein sequences. The papers in this review were selected from exhaustive searches across Google Scholar and Science Direct, using the following keywords and phrases: ‘variant effect prediction’, ‘mutation effect prediction’, ‘language modelling’, ‘natural language processing’, and ‘large language model’. Selected papers were required to fulfil the following criteria: (1) to capture recent innovations and emerging trends in this rapidly evolving field, the review was limited to publications from the past decade (2014-2024), along with a select few from early 2025 published prior to the submission of this article, (2) input data must include a DNA, RNA, or protein sequence, (3) the task must be variant effect prediction, and (4) the technique must involve language modelling, for instance, a traditional natural language processing (NLP) approach, a convolutional neural network, or an LLM.

Background

As this article details the applications of language modelling to variant effect prediction tasks, this section provides a brief introduction to both aspects – variant effect prediction and NLP – to set out the main problems in the field, and the technologies that can be used to address them.

Variant effect prediction

Uncovering the associations between genetic variants and human diseases necessitates an understanding of the many different possible types of variants. The variants most commonly explored in the field are single base-pair substitutions, referred to as single-nucleotide polymorphisms (SNPs). Still, a small number of models have been developed to analyse the combined effect of several SNPs.5,24 While several single base-pair substitutions can co-occur independently, they can also occur as a single event; in such cases, they are referred to as multiple base-pair substitutions. 25 However, there is no evidence they have been addressed in the literature. In addition to substitutions, 2 other significant forms of variation are insertions and deletions, collectively known as indels. Insertion refers to the case where additional nucleotides are inserted into a genetic sequence, while deletion refers to the case where nucleotides are deleted from such a sequence. Similarly to substitutions, these events can occur across single or multiple nucleotides. While some papers have investigated indel effect prediction,26-29 this has been explored to a substantially lesser extent than substitutions.

Existing work focuses largely on variants within genes, which code for proteins. However, these protein-coding regions comprise less than 2% of the human genome. 30 As illustrated in Figure 1, variants can also occur in the non-coding regions of the genome, including in regulatory elements such as promoters and enhancers. In fact, 90% of disease-associated variants identified by genome-wide association studies have mapped to non-coding regions, and the majority of these remain unannotated. 31 Hence, the discovery of non-coding variant effects remains a largely untapped source of potential knowledge that could aid in illuminating human disease mechanisms.

Figure 1.

SNP in coding DNA vs. non-coding DNA for transcription

Illustration of coding vs non-coding DNA, and an SNP in a promoter region, for a eukaryotic cell. Non-coding DNA consists of transcription factors, such as promoters, and transcription factor binding sites. Promoters drive the initiation of transcription. 32 Other cis-regulatory elements (CREs) include enhancers and silencers, which positively and negatively regulate gene expression, respectively. Insulators are an additional type of CRE, which interact with nearby CREs and can block distal enhancers or regulate chromatin interactions. 33

Source: Created in BioRender. Hegde (2024) https://BioRender.com/e16b233.

Natural language processing

Natural language processing techniques have long been used to model the structure of DNA, from statistical models 34 to LLMs. 21 The most frequently observed pipeline among the models reviewed here is shown in Figure 2; the sequences are tokenised before being input to the model, which is first pre-trained on a large corpus of data, and then fine-tuned for specific downstream tasks, such as the examples listed in the figure.17,35,36 Although unlabelled data sets of genetic sequences are abundant, labelled data sets are in shorter supply, causing a roadblock in the supervised fine-tuning of LLMs. For variant effect predictors, this can become a concern due to the lack of labelled data related to novel or emerging diseases. 37 The field is starting to innovate to tackle this problem. For instance, a small number of models developed in recent years have circumvented the fine-tuning stage by implementing zero-shot prediction, 38 where models progress straight from pre-training to inference, without needing additional data for fine-tuning.39-41 An alternative solution is data augmentation, where artificially generated examples are used to increase the size of the training data set. In genomics, this is often done by taking the reverse complement (RC) of a sequence,42,43 or by introducing small random translations to the sequence.44,45 However, a recent study presented an evolution-based method of DNA sequence augmentations, hence increasing the genetic diversity of the training data set while preserving the biological functionality of the sequences. 46

Figure 2.

The diagram depicts a language model pipeline for variant effect prediction, classified into genomics (Dna, RNA, and Protein sequences), proteomics, and an optional MSA. The process includes tokenization of sequences, input into a language model, and further classification of variant effect tasks like pathogenicity or disease association, as well as regression of variant effects and functional effects.

Generic language modelling pipeline, including the main categories of tasks covered in this review. The DNA, RNA, or protein sequences are tokenised before being input to the model. The model is initially pre-trained on a large corpus of data, and then fine-tuned on a data set specific to the planned downstream tasks, eg, variant pathogenicity classification.

Source: Icons from BioRender https://app.biorender.com/.

The first step of the pipeline is tokenisation, where the input sequence is segmented into discrete units, referred to as tokens, using defined separators. This process converts the unstructured input data into a standardised format, hence enabling the model to create a numerical representation of the data so it can be processed.47-49 The chosen tokenisation method may have a significant impact on model performance. For instance, k-mer tokenisation produces a set of tokens with the same length k. The use of these constant-length tokens can lead to heterogeneous token frequencies due to the relative rarity of certain sequence patterns, such as CG dinucleotides; 50 this can negatively impact the model training process by causing the model to focus on token frequency patterns rather than the contextual relationships within a sequence. 51 Recent papers have addressed this limitation with the use of byte-pair encoding, 52 which creates a frequency-balanced vocabulary by creating combined tokens for more frequent sequence patterns.53,54

After tokenisation, the data can be used as an input to the model. There, the selected architecture plays a key role in the quality of predictions produced; the ensuing review will analyse and compare the state-of-the-art architectures in the field. The concepts of pre-training and fine-tuning date back to the introduction of transfer learning in 1976. 55 The pre-training stage allows the model to capture knowledge and context that can be used across a wide range of downstream tasks, while the fine-tuning stage builds task-specific understanding.56,57 Pre-training is most frequently done using unsupervised learning tasks such as masked language modelling (MLM), on large, unlabelled corpora of genetic sequences; this enables the model to learn without relying on the availability of large labelled data sets, which are scarce in the biomedical field.21,58 The smaller, labelled data sets are then used for task-specific fine-tuning. Frequently used data sets for both stages are detailed in the main review. After fine-tuning, the model can be used for downstream tasks. Figure 2 details some of the most common variant effect prediction tasks. It is important to note that there are several types of variant effect that can be measured, including fitness effect, pathogenicity, and functional change. 18 These result in different data types, and hence, model functionality will be informed by the specific task at hand. For example, some models may aim to classify a variant as pathogenic or non-pathogenic, whereas others may look to predict a numerical value representing its functional effect.18,59

Baseline clinical tools and guidelines for variant effect prediction

To understand the impact and effectiveness of the tools reviewed in this study, it is important to consider baseline clinical tools and guidelines. The simplest variant effect predictor can be built by creating a substitution matrix from a sequence alignment. For instance, the BLOSUM substitution matrix is created by aligning multiple proteins and providing a score to confer the likelihood of each amino acid substitution being conservative, ie, resulting in a protein with similar properties. 60 In fact, these scores have proven to be somewhat effective in predicting variant pathogenicity,61,62 and as such provide a useful baseline to assess whether a computational approach is adding value. The first widely adopted algorithmic approach to variant effect prediction is the Sorting Intolerant From Tolerant (SIFT) algorithm, 63 which is often used as a baseline to this day. The SIFT uses multiple alignment data to predict normalised probabilities of substitutions in each position of an input sequence. Then, a chosen cut-off is used to determine whether or not each substitution is deleterious. The original study showed that SIFT correctly predicted the deleteriousness of substitutions at a rate 14% higher than a BLOSUM62 substitution matrix; hence, it became a mainstay in the field. 63 Another frequently used baseline tool is PROVEAN (Protein Variation Effect Analyser), 64 an alignment-based algorithm for predicting the functional effects of variants in protein sequences, including single and multiple amino acid substitutions, insertions, and deletions. The PROVEAN achieves high results without machine learning, achieving balanced accuracy scores of over 80% for insertions and deletions and over 75% for amino acid substitutions. 64 PolyPhen 65 and PolyPhen-2 66 both combine features from open-access databases to predict the effects of variants in the coding region of the human genome. The former uses empirically determined rules for classification and achieves a true positive rate of 82% on the SwissProt 67 database. The latter uses a Naive Bayes approach and achieves true positive rates of 73% on the HumVar 68 database and 92% on a data set compiled of Mendelian disease-causing variants and their benign counterparts from UniProt. 69 Other widely adopted variant effect tools include the Ensembl Variant Effect Predictor 70 and the evolutionary model of variant effect (EVE). 71 Many of the papers reviewed below use these as baselines against which to compare their results. While there are a plethora of variant effect prediction tools and techniques, these must be held to a consistently high standard in order to be used clinically. Globally, regulators have set guidelines for reporting variants as pathogenic or benign. In the United States of America, the American College of Medical Genetics and Genomics and the Association for Molecular Pathology jointly produced a set of standards and guidelines for the interpretations of sequence variants. 72 In the United Kingdom, the Association for Clinical Genomic Science produced a similar set of best practice guidelines, with UK-specific adaptations. 73 These guidelines provide detailed criteria for when a variant can be reported as benign or pathogenic, based on clinical or experimental data. Although they can assist in interpreting the data when building computational variant effect predictors, these guidelines have not yet set standards in the field.

Language Models for Variant Effect Prediction

Pre-transformer models

Although researchers used forms of language modelling to solve machine translation as early as the 1940s,74,75 The Chomsky work on grammars and syntactical structures in the mid-1950s formed the basis of what we consider NLP today, where machines are able to ‘understand’ structure and context within languages. 15 A detailed historical review of the field can be found in Sparck Jones. 76

Since its inception, the field has undergone many changes and innovations. Figure 3 shows the evolution of models up to the development of the transformer in 2017.

Figure 3.

The diagram is a timeline of developments in language models from 1980 until the development of the transformer in 2017. It starts with the use of grammars, markov models, and n-gram models in the 1980s. The timeline moves to the year 2001, where the first neural language model, a feedforward neural network (FFNN), was developed. This was followed by the development of convolutional neural networks (CNN), long short-term memory (LSTM), and recurrent neural network (RNN) language models. The timeline culminates in 2017 with the development of the transformer, which combines attention mechanisms and convolutional neural networks (CNN). Classical ML refers to classical machine learning techniques such as support vector machine and Naive Bayes. Markov models are often used to construct grammars.

Timeline of models from 1980 until the development of the transformer. Classical ML refers to classical machine learning techniques such as support vector machine and Naive Bayes. FFNN, feed-forward neural network; CNN, convolutional neural network; LSTM, long short-term memory. Markov models are often used to construct grammars.77,78

There were significant advancements in the 1980s and 1990s, with the use of statistical models such as n-gram 79 and Hidden Markov Models. 80 The development of neural networks led to a further turning point in the field, leading to the use of neural language models, which were better able to learn semantic relationships between words, and generalise to unseen test sets, compared to their predecessors. 81 The original feed-forward neural network (FFNN) was created in the 1980s 82 and adopted in language modelling in the 2000s. 83 A widely used neural network architecture is the convolutional neural network (CNN), which was developed in the late 1990s, 84 and introduced in NLP in the mid-2000s. 85 Instead of relying on manually selected features, CNNs learn features directly from the input data, making them superior to implement end-to-end compared to traditional machine learning methods. Hence, they have become prevalent in DNA sequence modelling and classification.86-89 While CNNs are excellent at learning short-range dependencies, they struggle to model relationships between words (or nucleotides) far from each other. 90 This limitation underscores the need for more advanced architectures to address such dependencies.

Recurrent neural networks (RNNs) 82 were introduced in NLP as a possible alternative to CNNs, as the use of recurrent connections enabled these models to incorporate many previous inputs into future steps. 91 However, traditional RNNs suffer from a problem referred to as ‘vanishing gradients’, which makes them prone to ‘forgetting’ inputs that are further back in the sequence. Two main alternatives have been brought forth in an attempt to circumvent this problem: (1) the long short-term memory (LSTM) network, 92 which is able to handle long-term dependencies using a more complex architecture formed of different gates and (2) the gated recurrent unit (GRU), 93 which uses a simplified version of the LSTM architecture to streamline sequence handling. Several variants of these models have been utilised for language modelling over the past decades, both individually and as part of ensembles with other neural networks such as CNNs.94,95

Despite the fact that the introduction of transformers in 2017 marked a significant milestone in deep learning, the development of models using other architectures has continued. As shown in Table 1, many recent models using pre-transformer technologies, including those using CNNs, have demonstrated notable performance. In particular, the Genomic Pre-trained Network (GPN) model, a CNN-based approach for genome-wide effects of variants in DNA, has demonstrated state-of-the-art performance. 96 The architecture of the convolutional model was selected after it was observed that it converged faster than its transformer-based counterpart during pre-training, and the results showed that it outperformed other genome-wide variant effect predictors for Arabidopsis. Another noteworthy finding of this study was the performance gain observed from training on multi-species data instead of single-species data. This suggests that incorporating cross-species data can provide richer context for understanding genetic variation and can potentially improve the generalisability of the model.

Table 1.

Summary of neural language models for variant effect prediction (see Table 5 for code/data availability).

Paper Task Year Architecture Data type Variant type
Kim et al 88 Identifying cancer driver mutations 2018 CNN DNA Coding
Pejaver et al 97 Inferring the molecular and phenotypic impact of SAVs 2020 CNN Protein Coding
Shin et al 26 Protein variant effect prediction 2021 CNN Protein, RNA
Dunham et al 98 Protein variant effect prediction 2023 CNN Protein Coding
Benegas et al 96 Prediction of genome-wide DNA variant effects 2023 CNN DNA Coding, Non-coding
Tan and Shen 99 Non-coding variant effect prediction using genome sequence and chromatin structure 2023 CNN, GCN DNA Non-coding
Cheng et al 100 Self-supervised learning for DNA sequences with circular dilated convolutional networks 2024 CNN DNA Non-coding

In addition to CNNs, the graph convolutional network (GCN) has also proved to be a performant non-transformer language modelling approach for variant effect prediction. Notably, its enhanced ability to capture graph-like structural information compared to other neural network architectures has proven useful in DNA variant effect prediction approaches incorporating structural data alongside sequence data. 99

These findings underscore the ongoing relevance of pre-transformer neural network architectures in genomics and highlight the potential benefits of leveraging diverse data sets for training.

Transformer-based models

History and overview

The origination of the transformer architecture was a pivotal point in the NLP field, resulting in models that pushed the boundaries of human ability to process natural and biological languages. Figure 4 summarises the timeline of the most impactful models that have been produced, starting with the original transformer in 2017. After 7 years, it is still an active field of investigation; 2023, in particular, was a year of many developments for both transformer-based and non-transformer language models. A significant limitation common to statistical and neural language models is the need to specify a fixed context length prior to training; this restricts the capacity of these models to utilise extended contexts for predictions. 101 The attention mechanism was created to address this limitation, by computing weights for each token in the input sequence to capture its relation to the others, and applying scaling to focus (or ‘give attention’) on the tokens relevant to the task. 102 Several models achieved good results on machine translation tasks by combining this attention mechanism with recurrent networks.87,103 The attention mechanism was eventually developed into the self-attention mechanism, which forms the basis of the modern transformer. 16 Self-attention (Figure 5) is applied within a single sequence to compute a representation of that sequence and provides a method of learning long-range dependencies within input sequences. The original transformer architecture, summarised in Figure 6A and shown in detail in Figure 6B, 16 combines self-attention with fully connected layers. Multiple self-attention mechanisms are used in parallel; this is referred to as multi-head self-attention (Figure 5) and reduces the complexity per layer, hence increasing the capability for parallelisation. The attention layers are stacked with fully connected layers to form an encoder-decoder model. The input sequence is encoded by the encoder into a representation, which is then stored as a latent state. The decoder then decodes the representation into an output sequence, which is subsequently passed to the linear and softmax layers to produce the output predictions. The transformer architecture enables the modelling of complex patterns and long-range dependencies within sequence data, making it well-suited to tasks on DNA, RNA, and protein sequences.22,104 These features make the transformer more performant on complex tasks compared to recurrent or convolutional networks and enhance its efficiency. In particular, the multi-head attention mechanism enables efficient feature extraction and context-aware modelling of these biological sequences. 105 As illustrated in Figure 3, the transformer has been used both independently and in conjunction with other models such as LSTM and CNN.

Figure 4.

Describe the information in the image.

Timeline of developments in NLP since 2017.

Figure 5.

Comparison of self-attention, Multi-head attention, Hyena operator, and Mamba operator mechanisms in neural network architectures.

Comparison of the self-attention mechanism and alternatives. (A) Scaled dot-product attention, as shown in Avsec et al. 106 The attention mechanism is applied simultaneously to a set of queries Q, with keys K and values V . Hence, the output matrix is computed as: Attention(Q, K,V) = softmax(QK T / p[dk])V . MatMul = matrix multiplication. The Mask between the Scale and Softmax is used only in the decoder to preserve the auto-regressive property, by preventing the flow of data from right to left. 16 (B) Multi-head attention, as shown in Avsec et al. 106 The presence of h heads indicates that h attention layers run in parallel. (C) Hyena operator of order N, as shown in Nguyen et al. 107 Combinations of dense layers and convolutions are applied to the input; the resulting projections are then fed to the element-wise gate layers. An MLP is used to implicitly parameterise the long convolutions, hence producing the convolutional filters. 107 x indicates the input. (D) Mamba operator, adapted from Gu and Dao. 108 The Mamba operator combines a state space model (SSM) with an MLP. x indicates the input. For the activation function σ, either a sigmoid linear unit 109 or Swish 110 is used.

Figure 6.

Caption A: Transformer architecture with encoder-decoder, as per Vaswani et al.16; Caption B: Detailed transformer, from Vaswani et al.16, includes multi-head and masked attention; Caption C: Encoder-only transformer, adapted from DNABERT.35’s model; Caption C: Decoder-only transformer, adapted from GPT-1.111’s design.

Transformer architectures. (A) High-level representation of the encoder-decoder architecture comprising the vanilla transformer architecture. The encoder encodes the input sequence into a representation, which is stored as a latent state. The decoder decodes this representation into an output sequence. This is passed into the linear and softmax layers to produce the output predictions. (B) Detailed transformer architecture, adapted from Vaswani et al. 16 The multi-head attention modules consist of multiple self-attention modules used in parallel. These are stacked with fully connected layers to create an encoder-decoder model as shown in (A). (C) Encoder-only transformer architecture, adapted from DNABERT. 35 (D) Decoder-only transformer architecture, adapted from GPT-1. 111

While the original transformer uses an encoder-decoder architecture, it is possible to have models consisting of only one or the other. For instance, the generative pre-trained transformer (GPT) series of models111-113 are decoder-only generative models, which, when given an input sequence, output the probabilities of possible subsequent tokens. By feeding the extended sequence back into the model and repeating the process many times, it is possible to generate a body of text. Figure 6D shows a decoder-only model based on GPT-1. 111 These models have undergone significant developments since the release of GPT-1 111 and now form the basis of the notorious ChatGPT chatbot. A significant limitation of models using the standard transformer architecture is their unidirectionality; each token can only incorporate context from the previous tokens, hence limiting the model’s ability to perform sentence-level tasks.114,115 This was addressed by the development of BERT (Bidirectional Encoder Representations from Transformers), 114 an encoder-only model that transforms text embeddings into a representation that can be used for a variety of tasks. The BERT achieves bidirectionality by using a masked language modelling (MLM) pre-training objective, in which the model attempts to predict the identity of randomly masked tokens in the input sequences, hence learning a representation that combines the context from the left and right. Although originally designed to process text, BERT has also been extensively applied in the field of molecular biology, resulting in models such as DNABERT 35 (Figure 6C) and ProteinBERT. 116 As the bidirectional architecture incorporates contextual information from the entire input sequence, it performs well at uncovering relationships between different elements in DNA sequences. 35

Although LLMs have led to a paradigm shift in computational solutions for biological problems, they still experience several limitations. Data scarcity is a significant challenge; limited high-quality labelled data are available for several biological problems of interest, including non-coding variant effect prediction.117,118 This limits the use of LLMs for these problems due to their requirement for large quantities of training data. In addition, training on insufficiently diverse data can lead to poor generalisation across tasks. 21 Efforts to address these limitations have led to the emergence of foundation models, LLMs which are pre-trained on very large-scale data for parameter initialisation and are then able to be fine-tuned for an extensive range of downstream applications.119,120 The data-intensive pre-training stage enables fine-tuning with comparatively limited data, hence improving the models’ generalisability and allowing the models to be applied to biological problems with insufficient data to train an LLM from scratch. 121 Notable foundation models in bioinformatics, highlighted in red text in Figure 4, are DNABERT, 35 DNABERT-2, 54 Nucleotide Transformer, 122 and the ESM series of models.39,123,124

Despite the many successes of transformers, they also have a major drawback: the time and memory used by the self-attention mechanism scale quadratically with sequence length, leading to high computational costs and creating a performance bottleneck.125-127 These models are hence impractical to train and use without access to extensive computational equipment and power. Crucially, this is also an environmental concern, with LLMs having huge carbon and water footprints.128,129 Hence, research is required to produce models that can achieve excellent results without being highly resource-intensive. These concerns have sparked a trend in the field of creating computationally efficient models as an alternative to the transformer; these are explored in detail in the next section. Notwithstanding the benefits of these post-transformer technologies, development of transformer-based models has continued, with the release of highly performant models such as DNABERT-2 54 and VespaG 130 as recently as 2024.

Review of existing models

Transformer-based LLMs are by far the most common language models used in the variant effect prediction field. This section reviews the existing models in the field, identifying key trends.

While all models surveyed take a sequence input – DNA, protein, or RNA – the precise input type varies. Some models take both the mutated and wild-type sequences as input,131-134 while others take a wild-type sequence alongside tabular data describing a variant.97,135 Whereas the majority of models report taking an input sequence of length up to 10 000 bases (Figure 7), the Enformer 106 is notable as it can process significantly longer sequences, ie, up to 96 608 bases.

Figure 7.

A graph in Figure B shows training time in GPU hours for different state-of-the-art LLMs, indicating an upward trend with a few exceptions.

Input sequence length, number of parameters, and training time for models which have reported these statistics in the original papers. (A) Maximum input sequence length (x-axis) and number of parameters (y-axis) as reported in original papers for each model. The model names are indicated on the chart. There is no clear trend shown over time. Compared to the majority of transformer-based models, Caduceus, a Mamba-based model, has far fewer parameters and can handle longer input sequences. (B) Training time in GPU hours for state-of-the-art LLMs. GPU hours = number of hours × number of GPUs. In general, the training time required for LLMs has increased over the years. However, DNABERT and ESM-1b are outliers, having very high training times; this is likely due to the fact that both are foundation models, which were trained on very large data sets. The GPN-MSA is another outlier and has a particularly low training time, likely due to the use of retrieval augmented processing 136 to increase computational efficiency. 43

In addition to sequence input, several methods integrate multiple sequence alignments (MSAs) as an additional input. Indeed, the conserved residues predicted by MSA can be predictive of variant effect.137,138 Thus, it has been observed across many models that incorporating MSA as an auxiliary form of data improves the quality of predictions.43,139 However, this is largely dependent on the quality of the MSA, which is variable, and often poor due to a lack of appropriate data.140,141 Despite the positive results observed in variant effect predictors using MSA, they are not appropriate for all use cases, as many variants lie outside MSA coverage. 142 In addition, several predictors not using MSA have matched or outperformed MSA-based predictors while eliminating the additional computational cost associated with having a larger training data set. 132 For example, a benchmarking study 143 showed that ESM-1v, 39 which does not use MSA, outperformed several MSA-based state-of-the-art models. Hence, many recent approaches to variant effect prediction have eschewed MSA in favour of sequence-only input.

Human data are most predominantly used to train and test the models surveyed here. However, a few studies have demonstrated that incorporating data from multiple species during training can improve results compared to models trained on human data only. Indeed, it has been suggested that learning the variability across various genomes can assist a model in learning about the degree of conservation across genetic sites, hence improving its ability to predict variant pathogenicity.43,122,144

The majority of models surveyed adhere to the pipeline described in Figure 2, which includes pre-training and fine-tuning stages. Traditionally, language models used the pre-training task of next-token prediction. While this is still used in some contemporary models, 126 the field has generally moved to favour MLM 114 due to its ability to incorporate bidirectional context. However, MLM is not always the optimal choice, as it has been suggested that it may be insufficiently challenging for the model in cases where the training data include a multi-species MSA containing sequences very similar to the human genome; this has previously been addressed by excluding these very similar genomes during training. 43

To maximise efficiency and minimise computational cost, recent work has explored zero-shot prediction, where prediction is performed straight after pre-training, without fine-tuning. A benchmarking study 17 compared the ability of several state-of-the-art models to perform a non-coding variant effect prediction task 145 without additional fine-tuning. There, 2 transformer models, ie, Nucleotide Transformer 122 and Enformer, 106 were compared with CNN models GPN 96 and ResidualBind. 146 Eventually, Enformer performed best, achieving a Pearson correlation of 0.68 between the experimental and predicted values. Then, the CNN methods achieved correlations between 0.35 and 0.55, whereas Nucleotide Transformer performed worst, with a correlation lower than 0.1. Based on these results, it was suggested that specialised supervised models may be a better choice for zero-shot prediction compared to current LLMs, which are pre-trained on broad data sets. 17

While the original transformer architecture consists of an encoder-decoder framework (Figure 6A and B), the decoder portion is often not required for biological language models, as sequence generation tasks are uncommon in this field. Hence, the majority of models summarised in Table 2 employ an encoder-only framework, often based on BERT to implement bidirectionality (Figure 6C). Indeed, state-of-the-art papers have demonstrated that such architectures are able to successfully model genetic sequences without the need for a decoder.35,39,54,123 Still, a few encoder-decoder models, based on the original transformer, 16 are also present.106,147-149 There is a lack of decoder-only models; however, this is to be expected, as such models are generally better suited to generating sequences, an ability that is not required for most variant effect prediction tasks. Furthermore, novelty does not always reside in the architecture; many models are based on pre-trained LLMs, which are then fine-tuned, hence eliminating the additional time and computational expense associated with pre-training a new model for a similar set of tasks. A prominent example is ESM-1b, 123 which has been exploited by many studies attempting protein variant effect prediction, as shown in Table 2. Another use of pre-trained models in the field has been to provide input into models that can be considered meta-predictors. 150 Such models input data into a pre-trained LLM, extract the output embeddings, and add a simple neural network-based classifier or regressor on top to make predictions based on these embeddings. This approach is highly data- and time-efficient in comparison to other LLM workflows, as it eliminates any training or fine-tuning of the LLM and requires only training of a simple neural network. Models using this methodology have achieved state-of-the-art results, showcasing this as an accurate and efficient framework for variant effect prediction.106,151 Recent work has also discovered benefits from integrating embeddings from multiple pre-trained LLMs, hence combining important context from diverse sources. 134

Table 2.

Summary of transformer-based language models for variant effect prediction (see Table 6 for code/data availability).

Paper Task Year Architecture Data type Variant type
Li et al 152 Prediction of pathogenicity of protein sequences 2020 Encoder-only (BERT) Protein Coding sequences
Rives et al 123 Prediction of protein variant effects 2020 ESM-1b 123 – encoder-only Protein
Meier et al 39 Prediction of functional effects of protein mutations 2021 ESM-1v – encoder-only Protein
Amadeus et al 24 Polygenic risk model for colorectal cancer 2021 Encoder-only DNA Coding, non-coding
Avsec et al 106 Prediction of non-coding DNA variant effects on gene expression 2021 Encoder-decoder DNA Non-coding
Ji et al 35 Identification of functional variants in non-coding DNA 2021 Encoder-only DNA Non-coding
Yamaguchi and Saito 153 Prediction of variant effects on multi-domain proteins 2021 Encoder-only Protein
Liu et al 40 a Zero-shot protein mutation pathogenicity prediction 2022 ESM-1b 123 Protein, MSA
Marquet et al 154 Prediction of protein variant effects 2022 ProtBert, 155 ESM-1b, 123 ProtT5-XL-U50 155 Protein
Yang et al 156 Prediction of deleteriousness of SNPs in non-coding DNA 2022 Encoder-only DNA Non-coding
Olenyi et al 157 Predicting SAV effects 2022 Marquet et al 154 Protein
Zhou et al 158 a Prediction of protein variant pathogenicity 2022 Elnaggar et al 155 Protein
Manfredi et al 135 Prediction of SAV pathogenicity 2022 ESM-1v39,155 Protein Coding
Dampier et al 159 Prediction of protease inhibitor resistance in HIV-1 mutations 2022 Encoder-only (BERT) RNA
Jiang et al 160 Prediction of SAV pathogenicity 2023 Encoder-only (BERT) Protein
Sun et al 161 a Prediction of SAV pathogenicity from sequence and structure 2023 Encoder-only (BERT) Protein, MSA
Brandes et al 142 Prediction of protein variant pathogenicity 2023 ESM-1b 123 Protein
Fan et al 27 Prediction of pathogenicity of insertion and deletion variants from protein sequences 2023 ESM-1b123,144 Protein, MSA
Benegas et al 43 a Genome-wide variant effect prediction in human DNA 2023 Encoder-only DNA, MSA Coding, non-coding
Derbel et al 131 Prediction of functional effect of SAVs 2023 Protein Coding
Hidayat et al 147 Prediction of BRCA1 variant pathogenicity 2023 ESM2 124 DNA Coding
James et al 162 Prediction of protein-coding SAV pathogenicity in the low-density lipoprotein receptor (LDLR) protein 2023 Frazer et al, 71 ESM-1v39,163 Protein, MSA Coding
Zhou et al 54 a SARS-CoV-2 variant classification 2023 Encoder-only RNA Non-coding
Cheng et al 164 Proteome-wide missense variant effect prediction 2023 Encoder-only, based on Jumper et al 163 Protein
Danzi et al 28 Variant prioritisation in Mendelian diseases 2023 Elnaggar et al 155 Protein, MSA Coding
Truong et al 148 Prediction of protein variant fitness 2023 Encoder-decoder Protein, MSA
Qu et al 165 Prediction of protein mutation effects using ensemble learning 2023 Ensemble:16,166 Protein, MSA
Blaabjerg et al 139 Protein variant effect prediction from sequence and structure 2023 Based on Rao et al 144 Protein, MSA
Dalla-Torre et al 122 Prediction of DNA variant effects 2024 Encoder-only DNA Coding
Lin et al 132 Prediction of protein missense variant pathogenicity 2024 ESM-1b 123 used in twin network configuration Protein Coding
Wild et al 151 a Prediction of DNA variant pathogenicity 2024 Benegas et al 43 and Dalla-Torre et al 122 DNA Coding, non-coding
Luo et al 29 Prediction of off-target effects of mismatches and indels 2024 Encoder-only (BERT) DNA, RNA
Gao et al 149 a (1) Prediction of DNA variant effects
(2) SARS-CoV-2 variant prioritisation
2024 Encoder-decoder DNA, RNA Non-coding
Zhan and Zhang 133 a Prediction of coding and non-coding variant effects 2024 ESM-1b 123 DNA, protein Coding, non-coding
Lafita et al 167 a Prediction of SAV pathogenicity 2024 ESM-1b, 123 ESM-1v, 39 ESM2 124 Protein Coding
Marquet et al 130 Prediction of SAV effect score 2024 Shallow neural network on top of Lin et al 124 Protein
Yan et al 134 Prediction of SAV pathogenicity 2024 Ensemble: ESM-1b, 123 ESM-1v, 39 ESM2124,155 Protein Coding
Shulgina et al 168 a Identification of RNA mutations beneficial to thermostability 2024 Decoder-only (GPT) RNA
Yang et al 169 Pathogenicity scoring for structural variants 2024 Tabtransformer 170 DNA Coding
Li et al 171 Prediction of missense coding variant pathogenicity 2024 Gated transformer Protein Coding
Zhong et al 172 a Prediction of functional effects of protein missense variants 2024 Graph attention transformer Protein Coding
Linder et al 173 Predicting the impact of genetic variation on gene expression 2025 Encoder-decoder (based on Avsec et al 106 ) DNA Coding, non-coding
Joshi et al 174 Prediction of coding VUS pathogenicity 2025 ESM-1b 123 DNA Coding
Glaser et al 175 a Prediction of functional effect of protein mutations 2025 ESM2 124 Protein Coding
a

Preprint.

By far one of the most significant transformer models has been AlphaFold, 163 which became famous for placing first in the 13th and 14th annual Critical Assessment of Structure Prediction (CASP) competitions,176,177 and significantly outperforming competitors. Due to its success, this architecture was adapted for human protein variant effect prediction, resulting in AlphaMissense. 164 This method fine-tunes AlphaFold on human and primate variant population frequency databases and classifies missense variants as likely benign, likely pathogenic, or uncertain. As in AlphaFold, an ‘Evoformer’ block 163 is used to process encodings of residue-residue interactions and MSA. The model eventually predicts the structure of the reference sequence, and the pathogenicity score for the variant, which is then converted into a classification. The structure prediction performance of AlphaMissense is comparable to that of AlphaFold, and it performs well on variant effect prediction across a variety of data sets. AlphaMissense achieves an area under the receiver operating characteristic curve (AUROC) of 0.94 on classifying missense variants in ClinVar, 178 outperforming EVE (AUROC = 0.911). 71 Among a set of proteins encoded by clinically actionable genes prioritised by the American College of Medical Genetics, 179 77% displayed improvements in accurate pathogenicity prediction when using AlphaMissense over EVE. AlphaMissense also outperformed the state-of-the-art methods on the 2 other evaluation data sets, achieving an AUROC of >0.8 on both.

While significant developments in model architecture have occurred, work on model interpretability is still limited. The majority of models mentioned in Table 2 function as black boxes, taking an input and returning an output. Although some of them have provided promising results, it is difficult for humans to understand and interpret the underlying logic.

Currently, it is uncommon for this issue to be addressed in papers in the field; however, a recent study on predicting CRISPR/Cas9 off-target activities included interpretability as a key contribution. 29 In the CRISPR/Cas9 gene editing system, base mismatches can occur during pairing of DNA and single-guide RNA sequences, leading to poor gene editing outcomes, and increasing the risk of ‘off-target’ mutations. Deep SHAP, 180 a statistical method to calculate the contribution of each hidden unit to the predictions of a model, was used to evaluate the importance of specific nucleotide positions in the model’s classification of off-target or on-target for each single-guide RNA and DNA pair. This method is easily interpretable by humans and can be used to plot a heatmap to visually identify key positions that contribute significantly to the decision-making process of the model. The resultant heatmap from the paper is shown in Figure 8. 29 The colour of each square indicates the strength of the contribution of the nucleotide position to the predicted class label; the legend is shown on the right-hand side.

Figure 8.

Heatmap from Luo et al28 showing DNA editing importance with Deep SHAP, for five cell lines. Y-axis: data sets, X-axis: nucleotide position. Colors show nucleotide importance, 1/−1 for positive/negative influence. ’N’ is PAM’s flexible nucleotide, ’G’ and ’G’ the PAM sequence itself.

Heatmap adapted from that produced in Luo et al 29 using the Deep SHAP method. 180 Evaluation was done on 5 independent data sets, each for a different cell line. The y-axis denotes the data set, while the x-axis denotes the nucleotide position. The colours indicate the importance of the nucleotide position towards the predicted class label; the legend is shown on the right-hand side. 1 and −1 respectively indicate a significant positive or negative contribution.

A key element of the CRISPR-Cas9 DNA editing system is the single-guide RNA sequence consisting of a 20-nucleotide protospacer and a 3-nucleotide protospacer adjacent motif (PAM) sequence. 181 The ‘N’, ‘G’, and ‘G’ positions represent the PAM sequence, which consists of any 1 nucleotide (N) followed by 2 guanines (GG).

The developments described above have resulted in the models described in Table 2. Comparing the performance of models across papers is challenging, as different studies tend to evaluate models on different data sets often using different metrics. One therefore cannot definitively conclude that a certain model is state-of-the-art in all aspects. It is, however, possible to assess trends across the models for specific tasks. For instance, transformer-based models have demonstrated good performance in classifying single amino acid variant (SAV) pathogenicity from protein sequences, with a number of models achieving an AUROC >0.8,135,160,161,167 and a few studies achieving an AUROC >0.9.116,123,162 The unique study published on predicting the effects of protein indels also showed promising performance; AUROC >0.8 was achieved when predicting the pathogenicity of both insertions and deletions across 2 separate data sets. However, this outperformed the previous state-of-the-art (non-transformer) methods by less than 0.1. Outcomes in predicting the functional scores of protein variants show a greater degree of variability, with the correlation between true and predicted values varying from below 0.5 139 to above 0.9. 123 However, this significant disparity in results may be due to the fact that these models were evaluated on different data sets. Performance on DNA variant effect prediction is similarly varied, although the best-performing models have achieved AUROC >0.9 for SNP classification.43,133,149,156 Although both coding and non-coding regions are addressed by these models, performance on some non-coding variant effect prediction tasks is still low; for instance, state-of-the-art models have achieved a correlation of less than 0.6 between true and predicted values on the Variant Effect Causal eQTL data set (Table 3). 106 Existing work on RNA tasks is promising, although limited. The evaluation of 3 models on a SARS-CoV-2 variant classification task yielded a best F1-score of 73.04, indicating potential for further enhancement. 54 Overall, the models demonstrating state-of-the-art performance across multiple tasks have been the Nucleotide Transformer, 122 DNABERT-2, 54 and ESM1b. 123 These are all foundation models, the former 2 for DNA and the latter for proteins. These results suggest that foundation models represent a promising direction for future research.

Table 3.

Most common data sets used in papers on language modelling for variant effect prediction.

Data set Data type Description Size Pub. year No. of citations Papers Open access
ClinVar 182 DNA ‘. . . germline and somatic variants of any size, type or genomic location.’ 182 500 000 variants 178 2016 2875 Fan et al, 27 Danzi et al, 28 Ji et al, 35 Liu et al, 40 , Benegas et al, 43 Dalla-Torre et al, 122 Lin et al, 132 Zhan and Zhang, 133 Yan et al, 134 Manfredi et al, 135 Brandes et al, 142 Hidayat et al, 147 Gao et al, 149 Wild et al, 151 Yang et al, 156 James et al, 162 Cheng et al, 164 Lafita et al, 167 and Joshi et al 174 Yes
gnomAD 183 DNA Genome and exome sequences 76 215 genomes, 730 947 exomes 2020 8243 Fan et al, 27 Danzi et al, 28 Benegas et al, 43 Lin et al, 132 Brandes et al, 142 and Jiang et al 160 Yes
Human Gene Mutation Database (HGMD) 184 DNA ‘. . . all known gene lesions underlying human inherited disease . . .’ 184 291 329
entries (free version) and 510 804 entries (paid version)
2020 1008 Pejaver et al, 97 Dalla-Torre et al, 122 Brandeset al, 142 Yang et al, 156 and Jiang et al 160 Yes – free version excluding past the 3 years’ data
UniProt 185 Protein Protein sequences + annotations, including functional information 253 206 171 entries 2004 2900 Shin et al, 26 Manfredi et al, 135 Hidayat et al, 147 and Olenyi et al 157 Yes
CAGI5 Regulation Saturation 145 DNA Non-coding SNPs + effect scores 175 000 variants across 9 promoters and 5 enhancers 2019 56 Tang and Koo 17 and Avsec et al 106 Yes
Variant Effect Causal eQTL 106 DNA Non-coding SNPs + effect scores 97 563 variants 186 2021 Unknown a Avsec et al 106 and Schiff et al 187 Yes

Source: Data sourced from ClinVar has been employed for both training and evaluation.

ClinVar: a large open-access database of human genomic variants, is the most widely used; Pub. year: publication year; No. of citations: overall number of citations as per Google Scholar; Papers: papers in this review using the data set.

a

While the paper reporting the creation of the data set 106 has 835 citations, it was not possible to determine the number of citations for the data set itself.

The transformer has led to a plethora of interesting and valuable studies on variant effect prediction. However, the lack of standard evaluation data sets and protocols has made performance comparison particularly difficult. Overall, performance on protein variant pathogenicity classification has been high; however, non-coding DNA and RNA variant effect prediction tasks have proved challenging and, thus, require further investigation to improve results. Recent approaches have aimed to reduce the computational cost associated with training and testing transformer-based models alongside enhancing the prediction quality. The increasing number of papers published on such models since 2020 (Figure 9), and the fact that such papers have been published as recently as January 2025 (Table 2), suggest that the transformer remains competitive for variant effect prediction.

Figure 9.

The graphs depict the number of published papers and citations for high-impact papers between 2020 and 2024. The total number of papers published per year has generally increased, with a slight decrease from 2023 to 2024. The number of LLM papers has far exceeded the number of neural LM papers each year. The number of citations per year for these papers has steadily increased since their publication.

Analysis of the number of published papers and the number of annual citations for the highest-impact papers. (A) Number of papers published per year on language models for variant effect prediction, as described in Tables 1, 2, and 4. Neural LM is the neural language model (Table 1). The LLM refers to both Transformer-based and post-transformer models (Tables 2 and 4). During the period 2018 to 2024, the overall number of papers per year has generally increased, with a slight decrease from 2023 to 2024. The number of LLM papers has far exceeded the number of neural LM papers each year. (B) Number of citations per year for the most impactful papers. The number of citations per year for these papers has steadily increased since their publication.

Beyond the transformer

In recent years, extensions and alternatives to the self-attention mechanism have been developed in order to tackle the high computational cost currently associated with training transformer-based LLMs. The timeline of these emerging technologies is displayed in Figure 4. Figure 5 provides a visual representation of the self-attention mechanism (A), multi-head self-attention (B), and the 2 major alternatives (C and D). The first such approach to gain traction was the Hyena operator (Figure 5C), which was developed in 2023 as a direct replacement for the self-attention mechanism. Using a recurrence of multiplicative gating interactions and long convolutions, 126 this approach scales linearly with sequence length, unlike the attention mechanism, which scales quadratically. Thus, the Hyena operator is 100 times faster than attention at a sequence length of 100 000 bases while delivering similar results. 107 This operator forms the basis of HyenaDNA, 107 a foundation model for DNA, which has achieved excellent results on tasks such as chromatin profile prediction and species classification. The subquadratic scaling of the Hyena operator makes it much more efficient in modelling long DNA sequences, a feature that is necessary for deciphering long-range interactions such as those involved in gene regulation.188-190 While current models often sacrifice single-nucleotide context to reach longer context,35,54,106 HyenaDNA maintains single-nucleotide resolution by using single-nucleotide tokens while also retaining the ability to process long contexts. The combination of these properties makes it well-suited to capturing the genome-wide effects of single-nucleotide alterations in DNA sequences.107,191

An alternative replacement for the attention mechanism is the state-space model-based Mamba operator 108 (Figure 5D). Unlike conventional state space models, which experience performance bottlenecks due to repeated matrix multiplications, Mamba uses a structured state space sequence (S4) model, which overcomes this by employing matrix diagonalisation. During pre-training on the Hg38 human reference genome data set, Mamba scaled significantly better than the HyenaDNA and Transformer++ baselines, achieving a lower perplexity (better ability to predict the next token) with the same or fewer parameters. In addition, the Mamba perplexity continued to decrease at sequences lengths over 105 base pairs (bp), whereas that of HyenaDNA increased above this length. The Mamba-based model outperformed HyenaDNA on a species classification task while using the same number of parameters, suggesting that Mamba models biological sequences more accurately and efficiently. Despite several developments in post-transformer methods, few of these models have been applied to variant effect prediction (Table 4).

Table 4.

Summary of post-transformer large language models for variant effect prediction (see Table 7 for code/data availability).

Paper Task Year Architecture Data type Variant type
Schiff et al 187 Non-coding variant effect prediction 2024 Caduceus; based on Mamba 108 DNA Non-coding
Nguyen et al 41 Predicting mutational effects on bacterial protein fitness
Predicting mutational effects on non-coding RNA fitness
2024 Evo, based on StripedHyena 192 DNA, RNA, protein Coding,
Non-coding

One of the first post-transformer models applied to variant effect prediction is Caduceus, 187 which is based on the Mamba operator. 108 The implementation leverages the RC nature of the 2 strands in a double-helix DNA structure, recognising that both strands contain semantically equivalent information. The Mamba operator is applied twice, once to the original DNA sequence, and again to a reversed copy of the sequence; the parameters are shared between these 2 applications to increase efficiency. This double application of the operator is termed BiMamba and is used as the basis of the MambaDNA block, which additionally defines an RC mathematical operation to re-combine the forward and reverse sequences. Parameter sharing enables bidirectional models that are much deeper while using fewer parameters than transformer-based equivalents and hence are more efficient in sequence modelling. The performance of Caduceus was evaluated on a non-coding variant effect prediction data set 106 and was compared with the state-of-the-art foundation models HyenaDNA 107 and Nucleotide Transformer. 122 Caduceus outperformed both state-of-the-art models, achieving an AUROC of 0.68 on variants that were 0 to 30 kbp (kilo-base pairs) from the nearest transcription start site (TSS). However, performance degraded with increasing distance of the variant from the nearest TSS, with the AUROC decreasing to 0.61 for variants at a distance of 100+ kbp. Notably, Caduceus was able to surpass the performance of Nucleotide Transformer v2 using only a fraction of the parameters (7.7M compared to the Nucleotide Transformer’s 500M).

The other notable example of a post-transformer model applied to variant effect prediction is Evo. 41 This is a hybrid Transformer-Hyena model, where Hyena operators are combined with multi-head self-attention to improve performance on long sequences; this approach is termed StripedHyena. 192 The majority of the computation required for sequence processing is performed by the Hyena layers, while the attention layers supplement the ability of the model to store and incorporate contextual information. Furthermore, the composition of the Hyena layers using short convolutions makes them effective at filtering ‘noisy patterns’ that can occur in DNA sequences due to the stochasticity of transcription.41,193,194 Evo was pre-trained on a prokaryotic whole-genome data set of 300 billion nucleotides, resulting in a model with 7 billion parameters, which can handle a context length of up to 131 072 nucleotides. 41 Analysis during training showed that the model scaled far better with sequence length compared to state-of-the-art transformer models; while the transformer-based models scaled quadratically with sequence length, the scaling of Evo was almost linear. However, the training was highly resource-intensive, with the first stage taking 2 weeks across 64 GPUs and the second stage taking a further 2 weeks across 128 GPUs. Hence, the availability of the pre-trained model is a major contribution of this work, as it can be applied to different tasks without requiring re-training from scratch. The Evo performance on variant effect prediction was tested across 2 tasks. First, the prediction of variant effects on bacterial protein fitness. The Spearman correlation between the experimental and predicted fitness values was 0.45, underperforming compared to state-of-the-art models, including Nucleotide Transformer 122 and RNA-FM, 195 which achieved correlation values between 0.5 and 0.55. 41 The second task was the prediction of variant effects on non-coding RNA fitness, in which Evo achieved a Spearman correlation of 0.27 between its predictions and the true values. While this exceeds state-of-the-art models, which achieved a correlation of less than 0.2 on the same task, the performance indicates that further research is required to produce a model that can accurately predict variant fitness in non-coding RNA. Evo was also tested on predicting mutational effects on human protein fitness; however, these experiments were unsuccessful; it was hypothesised that this may be due to the model being trained only on prokaryotic sequences, without any human samples.

These models have achieved mixed results. While in some cases, they have matched or exceeded state-of-the-art performance while reducing the number of model parameters required, the state-of-the-art models demonstrate limited ability to predict variant effects. While improvements in computational efficiency have been achieved using models such as Caduceus, this remains an area requiring further attention. For instance, Evo has achieved results exceeding the current state-of-the-art, and the pre-trained model has been made available; however, it would be necessary to undertake the resource-intensive pre-training stage again in order to make it suitable for use on the human genome. The major contribution of these post-transformer models is their enhanced ability to efficiently model longer genomic sequences, a property which is desirable for understanding long-range gene regulation mechanisms. However, the results on variant effect prediction tasks indicate that significant further work is required to use these models as reliable variant effect predictors.

Model Evaluation

This section details the approaches to model evaluation for language models in variant effect prediction. First, the main data sets used in the field are reviewed. Then, benchmarking studies are evaluated. Finally, relevant metrics and evaluation protocols are surveyed.

Data sets and benchmarking

A considerable challenge in the field is the difficulty of accurately comparing different models. The papers reviewed employ a variety of data sets and metrics, which seldom align. Even in the case of data sets or tasks that are used to assess multiple models, different papers select different subsets of the data set, or apply different metrics to measure model performance. This makes it challenging to compare the performance of various methods and hence can obscure the effect of different architectures on prediction quality. Hence, there is a pressing need for benchmarks that can enable comparison of models.

Table 3 summarises the main data sets used in the papers reviewed above; data sets used across multiple papers were identified, and their characteristics were summarised. The most impactful DNA database in the field is ClinVar; 182 its coverage of many different variants across the whole human genome makes it suitable for training and evaluating a wide range of models. Two other similar databases are also very popular – gnomAD 183 and the Human Gene Mutation Database. 184 The former is unique due to its inclusion of several different ancestry groups from around the globe. In addition, an equivalent for proteins exists in the form of UniProt, which contains over 200 000 000 protein sequences and annotations and is exploited by several protein language models. Although these databases are used across many scientific articles, it is rare for different models to be evaluated on the same subset of a database. As shown in Tables 5 to 7, the data sets used in the field are numerous, vary significantly across papers, and frequently are not open access. Even the data sets that have been the most popular, such as the CAGI5 Regulation Saturation 145 and Variant Effect Causal eQTL 106 data sets, have only been employed across a small number of papers (Table 3). These constraints make it challenging to compare the performance of different models, as their data sets may vary significantly in the type of data or category of task.

Table 5.

Code and data availability for neural language models in Table 1.

Paper Data source Data availability Code availability Model
Kim et al 88 Hudson et al 196 and Gonzalez-Perez et al 197 N/A N/A N
Pejaver et al 97 Stenson et al, 184 Sherry et al 198 and Mottaz et al 199 http://mutpred.mutdb.org/wo_exclusive_hgmd_mp2_training_data.txt https://github.com/vpejaver/mutpred2 N
Shin et al 26 Apweiler et al 185 and Riesselman et al 200 https://zenodo. org/records/
4606785
https://github.com/debbiemarkslab/SeqDesign N
Dunham et al 98 Kryshtafovych et al 176 and AlQuraishi 201 https://zenodo. org/records/
7621269
N/A https://www.ebi.ac.uk/biostudies/studies/S-BSST732
Benegas et al 96 Sayers et al 202 and Togninalli et al 203 N/A https://github.com/songlab-cal/gpn https://huggingface.co/collections/songlab/gpn-653191edcb0270ed05ad2c3e
Tan and Shen 99 Zhou and Troyanskaya, 204 Chen et al, 205 and Biggs et al 206 https://zenodo.
org/record/
7975777
https://github.com/Shen-Lab/ncVarPred-1D3D N
Cheng et al 100 Avsec et al 106 and Zhou and Troyanskaya 204 N/A https://github.com/wiedersehne/cdilDNA N

Table 7.

Code and data availability for post-transformer models in Table 4.

Paper Data source Data availability Code availability Model
Schiff et al 187 eQTL: 106 https://tinyurl.com/29nafrsw https://github.com/kuleshov-group/caduceus Y
Nguyen et al 41 Notin et al 221 and Koboriet al 222 N/A https://github.com/evo-design/evo Y

Table 6.

Code and data availability for transformer-based language models in Table 2.

Paper Data source Data availability Code availability Model
Li et al 152 ClinVar 182 https://www.ncbi.nlm.nih.gov/clinvar/ https://github.com/xzenglab/BertVS Y
Rives et al 123 Riesselman et al 200 and Grey et al 207 N/A https://github.com/facebookresearch/esm Y
Meier et al 39 UniRef90 208 https://www.uniprot.org/uniref https://github.com/facebookresearch/esm Y
Amadeus et al 24 Yusuf et al 209 N/A N/A N
Avsec et al 106 MPRA: 145 , eQTL: Original MPRA: http://www.genomeinterpretation.org/cagi5-regulation-saturation.html, eQTL: https://tinyurl.com/29nafrsw https://github.com/google-deepmind/deepmind-research/tree/master/enformer Y
Ji et al 35 Sherry et al 198 https://github.com/jerryji1993/DNABERT Y
Yamaguchi and Saito 153 N/A N/A https://github.com/dlnp2/evotuning_protocols_for_transformers N
Liu et al 40 a ClinVar182,210 https://www.ncbi.nlm.nih.gov/clinvar/ N/A N
Marquet et al 154 Riesselman et al 200 and Reeb et al 211 https://zenodo.org/records/5238537 https://github.com/Rostlab/VESPA Y
Yang et al 156 Rentzsch et al 212 https://figshare.com/articles/dataset/LOGO_dbSNP_score_chr/19149827/2 https://github.com/melobio/LOGO Y
Olenyi et al 157 UniProt 185 accession: Q9NZC2 https://www.uniprot.org/ https://github.com/Rostlab/LambdaPP/tree/main N
Zhou et al 158 a ClinVar 182 https://www.ncbi.nlm.nih.gov/clinvar/ N/A N
Manfredi et al 135 ClinVar 182 , UniProt 185 https://esnpsandgo.biocomp.unibo.it/datasets/ N/A N
Dampier et al 159 Original https://huggingface.co/damlab https://github.com/DamLabResources/hiv-transformers Y
Jiang et al 160 HGMD 184 professional version https://www.hgmd.cf.ac.uk/ac/index.php https://github.com/WGLab/MutFormer Y
Sun and Shen 161 a 200 , MSA: UniRef100 208 https://www.uniprot.org/uniref https://github.com/Stephen2526/Structure-informed_PLM Y
Brandeset al 142 ClinVar 182 ,
HGMD184,183
https://github.com/ntranoslab/esm-variants https://github.com/ntranoslab/esm-variants Y
Fan et al 27 Chang et al 213 and Kaplanis et al 214 https://github.com/xf-omics/SHINE https://github.com/xf-omics/SHINE Y
Benegas et al 43 a ClinVar182,183 https://huggingface.co/collections/songlab/gpn-msa-65319280c93c85e11c803887 https://github.com/songlab-cal/gpn Y
Derbel et al 131 N/A N/A https://github.com/qgenlab/Rep2Mut N
Hidayat et al 147 ClinVar 182 ,
UniProt 185
https://www.ncbi.nlm.nih.gov/clinvar/, https://www.uniprot.org/ N/A N
James et al 162 ClinVar 182 https://www.ncbi.nlm.nih.gov/clinvar/ https://github.com/facebookresearch/esm, https://github.com/OATML/EVE Y
Zhou et al 54 a Khare et al 215 https://github.com/MAGICS-LAB/DNABERT_2 https://github.com/MAGICS-LAB/DNABERT_2 Y
Cheng et al 164 Notin et al, 166 Landrum et al, 182 Sundaram et al, 216 Original curated
Benchmark
https://github.com/OATML-Markslab/Tranception, https://github.com/google-deepmind/alphafold/tree/main/afdb https://github.com/google-deepmind/alphamissense N
Danzi et al 28 ClinVar182,183 https://www.ncbi.nlm.nih.gov/clinvar/ https://github.com/ZuchnerLab/Maverick Y
Truong and Bepler 148 Notin et al 166 https://github.com/OATML-Markslab/Tranception https://github.com/OpenProteinAI/PoET Y
Qu et al 165 Notin et al 166 https://github.com/OATML-Markslab/Tranception N/A N
Dalla-Torre et al 122 ClinVar: 182 ,
HGMD:184,217
See ‘Data Availability’ section in original paper. https://github.com/instadeepai/nucleotide-transformer Y
Blaabjerg et al 139 Notin et al 166 https://zenodo.org/records/12798019 https://github.com/KULL-Centre/_2023_Blaabjerg_SSEmb Y
Lin et al 132 ClinVar:182,183 https://www.ncbi.nlm.nih.gov/clinvar/ https://github.com/wlin16/VariPred Y
Wild et al 151 a ClinVar:182,218 https://www.ncbi.nlm.nih.gov/clinvar/ N/A N
Luo et al 29 Lin et al 95 and Chuai et al 219 N/A https://github.com/BrokenStringx/CRISPR-BERT Y
Gao et al 149 a ClinVar:182,220 See ‘Data Availability’ section in original paper. https://github.com/ZjGaothu/EpiGePT Y
Zhan and Zhang 133 a N/A N/A https://github.com/zhanglab-aim/DYNA Y
Lafita et al 167 a ClinVar 182 https://www.ncbi.nlm.nih.gov/clinvar/ N/A N
Marquet et al 130 Notin et al 166 https://github.com/OATML-Markslab/Tranception https://github.com/JSchlensok/VespaG Y
Yan et al 134 Manfredi et al 135 https://github.com/yzh9607/TransEFVP/tree/master https://github.com/yzh9607/TransEFVP/tree/master N
Shulgina et al 168 a Original https://tinyurl.com/5abszup9 https://github.com/Doudna-lab/GARNET_DL Y
Li et al 171 Original https://github.com/genemine/MVFormer https://github.com/genemine/MVFormer N
Zhong et al 172 a Original https://huggingface.co/gzhong/PreMode https://github.com/ShenLab/PreMode Y
Joshi et al 174 UniProt 185 https://www.uniprot.org/ Available upon request N
Glaser and Braegelmann 175 a N/A N/A https://github.com/moritzgls/ESM-Effect N
a

= preprint.

Addressing this limitation either requires the community to agree on a set of data sets on which to evaluate new models or the compilation of a framework or data set that covers several different tasks. A source of inspiration should be the CASP, 223 a recurring set of experiments to determine the state-of-the-art in protein structure prediction methods. Every 2 years since 1994, research groups worldwide have been encouraged to submit results to ensure that a thorough and complete review of existing methods is conducted. The experiments provide a method for researchers across the community to evaluate their models on a common data set and provide several categories of tasks on which models can be assessed. This format could be highly applicable for the variant effect prediction community. A regular competition or community experiment comprising multiple categories of variant effect prediction tasks on varied context lengths would be invaluable in determining the state-of-the-art and deciding the course of future research. Furthermore, input from the clinical community on desired standards and ideal tasks could be used to assess the real-world applicability of such models.

Currently, benchmarking studies in the field are limited. The small number of existing benchmarks in the field are summarised in Table 8, with access links in Table 9. However, significant progress has been made by Livesey and Marsh at the University of Edinburgh in benchmarking protein variant effect predictors, with 2 successive studies published in 2020 140 and 2023. 143 They provide a comprehensive review of protein variant effect predictors at the time of publication, comparing their performance on deep mutational scanning data sets of human proteins, and ranking the models based on their results. The difference between the 2 articles highlights the progress in protein language modelling over the early 2020s. While the 2020 study identified DeepSequence 200 – a non-language modelling, probabilistic model – as the best variant effect predictor for proteins, the 2023 one revealed that LLM methods such as ESM-1v 39 produced even better results. Another notable finding was the increase in data availability; in the 2023 study, there were over twice as many data sets available on which to evaluate the models. A particular strength of this study is that models were compared across multiple metrics – AUROC, AUPRC, and correlation; the benefits of this are discussed further in the metrics section. Overall, these 2 studies provide a thorough review of the existing models for protein variant effect prediction. However, language modelling of specific aspects is not explored, as deep learning models of various methodologies are assessed.

Table 8.

Summary of existing benchmarks for large language models in variant effect prediction field (see Table 9 for access links).

Benchmark Task Year Data type No. of samples Organisms No. of predictors evaluated
Benchmarking of variant effect predictors using deep mutational scanning 140 Prediction of variant effect scores for missense SAVs 2020 Protein 7239 Human, yeast, bacteria, virus 46
BEND 224 Binary classification of non-coding SNPs as effect/no effect. (1) Gene expression and (2) disease 2023 DNA 105 263
295 495
Human 13
Updated benchmarking of variant effect predictors using deep mutational scanning 143 Prediction of variant effect scores for missense SAVs 2023 Protein 9310 Human 55
Genome understanding evaluation 54 Classification of SARS-CoV-2 variant pathogenicity 2024 RNA 91 669 SARS-CoV-2
Genomics Long-Range Benchmark 186 Prediction of SNP effect on gene expression 2024 DNAs Avsec et al 106 : 97 563.
Benegas et al 43 : 39 652.
Benegas et al 43 : 2 321 473
Human 3
Benchmarking computational variant effect predictors by their ability to infer human traits 225 Prediction of functional scores for rare-disease-associated variants in the human genome 2024 DNA 100 000 Human 24

Table 9.

Links to the benchmarks summarised in Table 8.

Benchmark Link
Benchmarking of variant effect predictors using deep mutational scanning 140 https://doi.org/10.6084/m9.figshare.12369359.v1, https://doi.org/10.6084/m9.figshare.12369452.v1
BEND 224 https://github.com/frederikkemarin/BEND
Updated benchmarking of variant effect predictors using deep mutational scanning 143 https://figshare.com/articles/dataset/Compiled_DMS_and_VEP_predictions/21581823/1
Genome understanding evaluation 54 https://github.com/Zhihan1996/DNABERT_2
Genomic long-range benchmark 186 https://huggingface.co/datasets/InstaDeepAI/genomics-long-range-benchmark

Although variant-specific benchmarks are scarce, variant effect prediction tasks are included in some benchmarking studies that evaluate the performance of LLMs on genomic modelling in general. For instance, the Genome Understanding Evaluation benchmark 54 consists of genomic modelling tasks across multiple species, including the classification of SARS-CoV-2 variants based on sequences of 1000 bp in length. Comparison of DNABERT-2 with several different versions of DNABERT and Nucleotide Transformer showed that a version of the Nucleotide Transformer pre-trained on multi-species data performed best, while DNABERT-2 was close behind (accuracies of 73.04% and 71.21%, respectively). A complementary study is the Genomics Long-Range Benchmark, 186 which evaluates model performance specifically on genomics tasks requiring modelling of long-range dependencies and includes the prediction of SNP effect on gene expression, using data derived from Avsec et al. 106 It was discovered that increasing context length improved models’ ability for variant effect prediction. In addition, models with longer context lengths were able to more accurately predict the effects of variants further from the TSS. Indeed, Enformer outperformed more recent models such as Nucleotide Transformer and HyenaDNA due to its ability to handle longer context.

While past benchmarks focused on the quality of predictions, there is also a need to understand and compare the computational cost of variant effect prediction models. Recent research has highlighted the immense impact of deep learning technologies on the natural environment, from carbon emissions to water consumption.128,129 Transformer-based LLMs are a significant culprit due to the quadratic scaling of the attention mechanism with context length. The computational cost of training on large data sets can be extensive; as shown in Figure 7, training can span across days or weeks, using multiple GPUs. Although large foundation models such as DNABERT and ESM-1b are particularly computationally expensive to train, the training time in general has increased since 2020. However, training is not the only computational expense associated with LLMs; while training only occurs once, inference occurs repeatedly, with the frequency depending on the application of the LLM. For instance, ChatGPT was visited over 3 billion times in December 2024. 226 Hence, since the total inference cost over time can match or exceed the training cost, it is crucial to understand and reduce its impact in the pursuit of environmentally conscious models. Table 10 lists the inference time as per the original paper for each model. Notably, not all LLM methods have high inference time, and many improve on traditional methods. In addition, recent methods have aimed to perform inference on consumer-grade machines rather than high-specification GPUs, hence making the models more accessible to run in clinical settings. For instance, VespaG 130 took only 5.7 seconds on a 12-core CPU to make predictions for 73 unique proteins from ProteinGym, 221 while a non-LLM method, GEMME, 227 took 1.27 hours to perform the same task on the same hardware. However, inference time is still far less frequently reported than training time – the only models for which this is reported are listed in Table 10. Hence, it is also challenging to compare existing methods based on this criterion.

Table 10.

Comparison of reported inference time for LLM methods.

Model Publication year Transformer-based CPU/GPU Inference time
E-SNP&GO 135 2022 Yes 1 × 12-core CPU 12.464 seconds per variant
VariPred 132 based on ESM-2 124 2024 Yes 1 × GPU – 12 GB Nvidia GTX 1010Ti 0.360 seconds per variant
VespaG 130 2024 Yes 1 × 12-core CPU 0.078 seconds per protein

Hence, to accurately compare models, simply testing on the same data set is insufficient. There must be a framework within the field for the evaluation metrics to use and the aspects of the model to report (for instance, training time, inference time, and computational resource usage).

Metrics

Pre-training metrics

A significant and specific metric in NLP is perplexity, 228 which can be calculated continuously throughout the pre-training stage to identify the optimal number of parameters. 41 Language models represent sequences by calculating the probability of each token based on the context from previous tokens. The perplexity is calculated by taking the inverse probability assigned to each token in a given set of data and normalising it by the number of words, as shown in equation (1) for a data set W = w1w2. . .wN. 229

perplexity(W)=P(w1w2...wN)1/N (1)

For a given model, a lower perplexity indicates an enhanced ability to predict the next token of a sequence. However, while an improvement in perplexity often correlates with an improvement in performance on downstream tasks, this relationship is not guaranteed, and hence, further evaluation metrics are required to directly evaluate the performance of the model on the task of interest.229,230 For instance, although Evo achieved a lower pre-training perplexity compared to transformer-based models, the latter still achieved better Spearman correlation between true and predicted values when predicting bacterial protein fitness. 41

Fine-tuning metrics

The fine-tuning metrics for NLP generally align with those for standard machine learning models. The accuracy and loss are measured throughout the fine-tuning process, which stops when the model converges (ie, when the chosen metric has remained the same for a set number of iterations).

Evaluation metrics

Three main categories of metrics are used to evaluate computational variant effect predictors. The first such category contains metrics that align with those used for standard machine learning models and use true and false positive rates to evaluate the predictions. These include AUROC,43,161 accuracy,147,160 precision, 159 recall, 159 and F1-score.54,156

The second category of metrics assesses the relationship between the true values and those predicted by the model. In cases where a numerical value such as a variant effect score is predicted, this is done by calculating the correlation between the 2. The Spearman rank correlation coefficient is most frequently used;39,123,131,147 however, some papers also use the Pearson98,106 correlation coefficient. All such metrics used in the reviewed papers are summarised in Table 11. While all of these metrics measure the agreement between the true and predicted values, they each measure this in a different way. For instance, the Pearson correlation coefficient assesses whether there is a linear relationship between the 2, while the Spearman correlation coefficient determines whether a monotonic relationship exists. A unique case is the Matthews correlation coefficient (MCC), 231 which is used to evaluate the agreement between the true and predicted classes in a classification problem.54,132 Unlike accuracy or AUROC, it takes into account all 4 aspects of a confusion matrix (true and false positive rates, and true and false negative rates), hence better representing the overall quality of predictions produced by the model. 232

Table 11.

Metrics used for assessing the relationship between the values predicted by the model and the true values.

Metric Measures Range Note
Pearson correlation coefficient 233 Linear relationship (Strong negative) −1 to 1 (strong positive) 0 = no relationship
Spearman correlation coefficient 234 Monotonic relationship (Strong negative) −1 to 1 (strong positive) 0 = no relationship
Matthews correlation coefficient 231 Agreement of classes (All inverse) −1 to 1 (all correct) 0 = no agreement
Jaccard similarity index 235 Similarity (No similarity) 0 to 1 (complete similarity)

Pearson, Spearman, and Jaccard metrics are used for prediction of numerical values. Matthews is used for classification.

To compare the agreement of these 2 categories of metrics, a simple meta-predictor was created by using the pre-trained Enformer model 106 to generate embeddings from SNPs in the ncVarDB 206 and using a simple machine learning classifier on top to perform a binary pathogenicity classification. The results of the different models tested are displayed in Table 12. It must be noted that, while Random Forest and Gradient Boosting achieved the same accuracy, their AUROC and MCC were different. In addition, the MCC achieved using support vector machine (SVM) with a linear kernel is very similar to that achieved using Random Forest, despite the latter having higher accuracy and AUROC values. These results demonstrate the importance of evaluating and comparing models across these different dimensions in order to fully understand the differences and determine the state-of-the-art.

Table 12.

Comparison of metrics for models performing variant pathogenicity classification on SNPs from ncVarDB, 206 using embeddings extracted from Enformer. 106 .

Model Accuracy AUROC MCC
SVM (RBF kernel) 69.4% 0.725 0.516
SVM (linear kernel) 73.6% 0.763 0.574
Random forest 77.5% 0.781 0.572
Gradient boosting 77.5% 0.778 0.558

MCC: Matthews correlation coefficient.

Beyond perplexity, no further NLP-specific metrics have been used to evaluate variant effect predictors based on language models. However, many such metrics have been developed to evaluate the ability to model natural languages, such as ROUGE 236 and its variants, and a variety of semantic embedding-based metrics.237,238 Moreover, recent papers have investigated the use of semantic similarity for assessing the ability of LLMs to appropriately model natural languages. Of particular interest is a 2024 paper in which the ability of an encoder to model substitution of a word with a synonym or antonym is tested; 239 this concept could be extended to genetic language modelling and hence evaluate the ability of an encoder to model substitution of a nucleotide. Despite the ability of non-NLP-specific metrics to evaluate the results of a model, they have no ability to assess the quality of language modelling or understand the underlying logic. Hence, to fully understand LLM performance, standard metrics must be combined with NLP-specific metrics.

While there are several metrics to assess the quality of model predictions, looking solely at the values of these metrics does not take into account other key aspects of a model, including computational cost. Although modifications such as including additional features in the training data, or increasing the size of the model, can enhance the predictive performance, they can also lead to a significantly higher computational cost. This calls into question the extent to which an increase in computational cost is justified for a corresponding increase in prediction quality. 240 For instance, usage of Pareto optimality has been adopted to attempt to select models with an appropriate trade-off between accuracy and inference latency. 241 In the future, it would be very valuable to define a metric to combine the information from each of the 3 categories above with data regarding computational cost.

Discussion

The advent of the transformer model in 2017 led to a paradigm shift in NLP and its applications to various fields, including the prediction of biological variant effects. Transformer-based language models have achieved mixed results in this area; while some models excel, others fail to make accurate predictions. Another significant limitation of transformers is the overwhelming computational cost associated with training and inference due to the quadratic scaling of the cost of the attention mechanism with sequence length. Research to address this has led to the development of several attention alternatives such as Mamba and Hyena. While these have garnered much attention in the LLM field, their capacity for variant effect prediction has not yet been fully explored, with only 2 models being used for this application so far. In addition, transformer-based models are still being proposed for variant effect prediction, as recently as early 2025, 174 demonstrating that this technology remains competitive.

The models produced to date have focused largely on single-nucleotide substitutions within proteins, or protein-coding regions of the human genome, often achieving promising results. However, there has been very little work on multiple base-pair variants, or non-substitution variant types, such as indels. Furthermore, while there has been extensive work on modelling DNA and protein sequences, there has been limited work on human RNA, despite the known associations between RNA variants and disease.242,243 Moreover, although extensive research has been conducted on the effects of variation within the human genome, very few recent studies have investigated the effects of variants in pathogenic organisms and viruses with a high disease burden. In particular, only 2 studies54,149 have looked at the mutational effects of SARS-CoV-2, which had a devastating impact on human health during the COVID-19 pandemic. Still, some work has been conducted on using deep learning to viral mutation data to predict individual risk 244 and the possibility of drug resistance. 245 Moreover, given that LLMs have already demonstrated effectiveness in modelling HIV, 159 they could potentially enhance results in this area.

Despite significant advancements in recent years, the field still faces several limitations. Many of the most prominent challenges are related to data rather than model architectures. A common issue observed among computational variant effect predictors is type 2 data circularity. Studies found that, in many cases, all variants within a particular gene are recorded with the same label (pathogenic or benign) across multiple different variant databases. This leads to models trained on these data performing well on known variants in known genes but poorly on de novo variants for newly identified risk genes. 246 Although a benchmarking study investigating this issue found that traditional machine learning models were the most prone to suffering from this issue, only 1 LLM (an ESM variant) was tested; hence, it is possible that others may still be at risk of suffering from this issue. 132 Therefore, it may be of interest to include such a test in future LLM benchmarking studies.

Another significant data-related challenge is that of demographic bias. Many large genomics data sets, such as UK Biobank, contain data largely from individuals of White European descent. 247 This poses a concern, as several mutations related to Mendelian diseases, including sickle cell anaemia and Tay-Sachs disease, have been shown to differ significantly in prominence across different groups.248,249 Hence, training on an ancestrally homogeneous data set risks the loss of valuable features when modelling the human genome and can lead to poor generalisation of models across different ancestral groups. The computational health care field has largely continued to uphold existing biases against underserved groups, with some widely used algorithms displaying clear racial bias. 250 As the field moves forward into an era where algorithms play an increasingly pivotal role in shaping personalised medicine, it is crucial to prioritise equity in future developments to ensure fair and unbiased outcomes for all.

In addition to addressing data set composition, the privacy of patient data is another key consideration when using LLMs for health care-related applications. As LLMs have already demonstrated their ability to identify sensitive information in documents such as electronic health records,251,252 this raises concerns around accidental patient identification via training data. Genomic data must be treated as particularly sensitive, due to the possibility of identifying not only an individual but also their familial relationships and links to specific traits or diseases. 253 This is of particular concern in rare disease research, where access to data on diseases experienced by only a handful of individuals increases the risk of individuals being identified. Although privacy solutions for genomic data sharing are being rapidly explored and developed,254,255 it is crucial to consider these through the lens of LLMs and the handling of data by those who develop these models. Indeed, LLMs can be susceptible to Membership Inference Attacks (MIAs) 256 and User Inference Attacks (UIAs). 257 The MIA aims to determine whether a given data record is present in the training data of an LLM and is conducted by creating an adversarial model to recognise the differences in an LLM’s response to its training data and its response to other samples. Recent research has shown that such attacks are effective on clinical language models, with samples from individuals with rare diseases being at greater risk of privacy leakage. 258 On the contrary, UIA attempts to ascertain whether an individual’s data were used in fine-tuning an LLM. While MIA threatens the privacy of individual samples, UIA puts the privacy of users who have contributed multiple samples at risk. 257 Both sets of attacks can severely compromise patient data privacy and can lead to the revelation of sensitive information about participants. However, tests on MIA and UIA have not yet been applied to genomic language models, and the latter has not yet been tested for any clinical LLMs. Hence, a framework must be created for testing the resiliency of state-of-the-art models in the field against such attacks. Crucially, these tests must be performed before models are adopted into clinical settings, to avoid putting patients at risk. The lack of interpretability in existing models poses another significant concern. The LLMs, like many deep learning models, often function as ‘black boxes’, with little human-understandable logic connecting the input data to the decisions of the model. In order to trust the decision-making of such models in clinical settings, it is necessary to understand and validate the logic behind such decisions. Due to the limited interpretability of existing predictors, meta-predictors based on these models experience the same issues. For instance, some studies have proposed that the exclusive use of LLM embeddings leads to limited biological interpretability and suggested that including more protein structure data could make the model more interpretable without increasing training costs. 134 Although work on interpretability in the field is currently limited, 1 recent paper 29 explores a method based on SHAP 180 to calculate the relative importance of the model’s hidden features on the output score. The authors reported this method to be effective, and it translated well to a visual representation of the relationship between the input and output features (Figure 7). However, this approach has yet to be adopted across other variant effect prediction papers. Alternatively, in attention-based models such as transformers, the attention weights confer the relative importance given to certain tokens259,260 and hence can be used to infer the impact of certain tokens in the decision of the model. 261 While this approach has been used to interpret the results of transformer-based for a number of bioinformatics applications,262-264 it has not yet appeared in the literature for variant effect prediction. For high-dimensional models, t-distributed Stochastic Neighbour Embedding (t-SNE) 265 is able to visualise the relationships between features and predictions. Thus, it has been used in transformer-based protein language models. 266 While some excellent reviews have been published on the state of interpretability of LLMs in biology and medicine, 267 they focus only on attention-based models. Further research is required to incorporate interpretability into post-transformer models such as Mamba and Hyena.

Future trends

Due to the significant training and inference costs associated with transformer-based LLMs, many recent studies have focused on creating more computationally efficient models, either using transformers or substituting the attention mechanism with alternative operators such as Hyena or Mamba. Although the advent of small language models (SLMs) 244 has advanced this area of research in natural language-based LLMs, they have not yet been applied to genetic sequences. A notable SLM is TinyLlama, 268 which utilises the same architecture and tokeniser as Llama2, 269 while leveraging novel computational methods such as FlashAttention 270 to create a model with fewer parameters and increased computational efficiency compared to state-of-the-art LLMs. The SLMs have already demonstrated impressive performance in text classification 271 and text-based health monitoring, 272 matching or exceeding the results achieved using LLMs. These findings underscore the potential of SLMs in future research and suggest that they may be an interesting avenue of advancement for biological language modelling also.

Although the development of SLMs is on the horizon, LLMs continue to be widely used. Recent papers have shown a trend towards the use of foundation models, which are pre-trained on a large corpus of data and can be fine-tuned for a wide range of downstream tasks. For instance, 8 separate papers in Table 2 base their models on the ESM-1b 123 foundation model. As the field aims to reduce computational cost, it is likely that foundation models will be even more widely used as an alternative to ab initio pre-training of new LLMs.

As the number of models in the field rapidly increases, 5 often trained and evaluated on different data sets, it is becoming increasingly challenging to identify the true state-of-the-art. To address this rising need, the development of benchmarking data sets has accelerated since 2023, resulting in the creation of benchmarks such as the Genome Understanding Evaluation. 54 As interest in computational efficiency and model fairness grows, it is likely that future benchmarks will include methods to assess these features of models and that such measures will become more significant when comparing models. Moreover, although models may perform well during technical evaluations, it is crucial to define and adhere to specific standards in order to discern their efficacy in clinical settings. For instance, in 2018, the National Health Service (NHS) in England and the UK National Institute for Health and Care Excellence (NICE) developed an evidence standards framework 273 to provide guidance on the development and usage of digital health and care technologies. While this framework places a high emphasis on demonstrating valuable results and significant benefits to the target population, it is not specific to artificial intelligence (AI) or LLM-based technologies, and hence does not detail any expectations for numerical results or other aspects of models. It is therefore of the utmost importance that those in the computational field work closely with clinicians to decide appropriate standards for the performance of variant effect predictors and implement strategies to bridge the gap between research and practice. Existing frameworks for models predicting individual prognosis or diagnosis include TRIPOD, 274 which explores transparent reporting, and PROBAST, 275 which estimates the risk of bias – these could be used to inform the creation of similar frameworks for language model-based variant effect predictors.

Alongside appropriate performance, the adoption of computational models in the clinical field requires the exploration of clinically relevant problems. While the bulk of work in the field has focused on the coding regions of the genome, research continues to uncover associations between non-coding variants and rare but highly impactful diseases in humans.276-278 Thus, although there has recently been increasing interest in predicting the impact of human genetic variation in the non-coding regions, further computational exploration of the non-coding genome is required. Furthermore, although current research focuses mainly on SNPs, diseases such as haemophilia have been linked to multiple base-pair variants or combinations of co-occurring SNPs.279,280 Very few papers exist on computational prediction of the effects of such variants;281,282 hence, this is an area of great interest for future work.

Conclusion

Although language models have proven effective in modelling DNA, RNA, and protein sequences, their results on variant effect prediction tasks remain mixed. The best performance on these tasks has been achieved by large transformer-based foundation models, pre-trained on large corpora of sequence data. However, such models incur a high-computational cost in terms of training and inference. While this has begun to be addressed via the creation of alternatives and extensions to the attention mechanism, these have had limited use in bioinformatics thus far. Initial studies show that models based on these technologies, such as Caduceus and Evo, achieve results comparable to transformer-based models while consuming less time and fewer resources for training and inference. Nevertheless, the state-of-the-art results for some tasks of importance, including non-coding variant effect prediction, require improvement. Despite the substantial progress in the field in recent years, there are still a number of limitations that persist, including demographic bias in training data sets, and the limited work on variants spanning multiple base pairs or situated in the non-coding regions of the genome.

Acknowledgments

The authors thank the anonymous reviewers for their valuable suggestions.

Footnotes

Author Contributions: Megha Hegde: Conceptualisation; Investigation; Writing – original draft; Formal analysis; Methodology; Visualisation; Writing – review & editing; Data curation.

Jean-Christophe Nebel: Conceptualisation; Writing – review & editing; Supervision; Project administration.

Farzana Rahman: Conceptualisation; Writing – review & editing; Supervision; Project administration.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work is supported by funds from the Kingston University Graduate Research School PhD Studentship.

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

References

  • 1. Karki R, Pandya D, Elston RC, Ferlini C. Defining ‘mutation’ and ‘polymorphism’ in the era of personal genomics. BMC Med Genomics. 2015;8:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Goetz LH, Schork NJ. Personalized medicine: motivation, challenges, and progress. Fertil Steril. 2018;109:952-963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Jamuar SS, Tan E-C. Clinical application of next-generation sequencing for Mendelian diseases. Hum Genomics. 2015;9:1-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Rahit KMTH, Tarailo-Graovac M. Genetic modifiers and rare Mendelian disease. Genes. 2020;11:239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Castro-Giner F, Ratcliffe P, Tomlinson I. The mini-driver model of polygenic cancer evolution. Nat Rev Cancer. 2015;15:680-685. [DOI] [PubMed] [Google Scholar]
  • 6. Jia G, Lu Y, Wen W, et al. Evaluating the utility of polygenic risk scores in identifying high-risk individuals for eight common cancers. JNCI Cancer Spectr. 2020;4:pkaa021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Lvovs D, Favorova OO, Favorov AV. A polygenic approach to the study of polygenic diseases. Acta Naturae. 2012;4:59-71. [PMC free article] [PubMed] [Google Scholar]
  • 8. Visscher PM, Yengo L, Cox NJ, Wray NR. Discovery and implications of polygenicity of common diseases. Science. 2021;373:1468-1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Zhang F, Lupski JR. Non-coding genetic variants in human disease. Hum Mol Genet. 2015;24:R102-R110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Moyon L, Berthelot C, Louis A, Nguyen NTT, Roest Crollius H. Classification of non-coding variants with high pathogenic impact. PLoS Genet. 2022;18:e1010191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Brendel V, Busse HG. Genome structure described by formal languages. Nucleic Acids Res. 1984;12:2561-2568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Brendel V, Beckmann JS, Trifonov EN. Linguistics of nucleotide sequences: morphology and comparison of vocabularies. J Biomol Struct Dyn. 1986;4:11-21. [DOI] [PubMed] [Google Scholar]
  • 13. Searls DB. The linguistics of DNA. Am Sci. 1992;80:579-591. [Google Scholar]
  • 14. Solan Z, Horn D, Ruppin E, Edelman S. Unsupervised learning of natural languages. Proc Natl Acad Sci U S A. 2005;102:11629-11634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Chomsky N. Three models for the description of language. IRE Trans Inf Theory. 1956;2:113-124. [Google Scholar]
  • 16. Vaswani A, Shazeer N, Parmar N, et al. Attention is all you need. In: Guyon I, Von Luxburg U, Bengio S, Wallach H, Fergus R, Vishwanathan S, Garnett R, eds. Advances in Neural Information Processing Systems. vol 30. Curran Associates Inc; 2017. [Google Scholar]
  • 17. Tang Z, Koo PK. Building foundation models for regulatory genomics requires rethinking large language models. Paper presented at: Proceedings of the ICML Workshop on Computational Biology; July 29, 2023; Honolulu, HI. [Google Scholar]
  • 18. Bromberg Y, Prabakaran R, Kabir A, Shehu A. Variant effect prediction in the age of machine learning. Cold Spring Harb Perspect Biol. 2024;16:a041467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Zhao WX, Zhou K, Li J, et al. A survey of large language models. arXiv [preprint]. doi: 10.48550/arXiv.2303.18223 [DOI] [Google Scholar]
  • 20. Shanahan M. Talking about large language models. Commun ACM. 2024;67:68-79. [Google Scholar]
  • 21. Zhang S, Fan R, Liu Y, Chen S, Liu Q, Zeng W. Applications of transformer-based language models in bioinformatics: a survey. Bioinform Adv. 2023;3:vbad001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Sarumi OA, Heider D. Large language models and their applications in bioinformatics. Comput Struct Biotechnol J. 2024:23:3498-3505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Tian S, Jin Q, Yeganova L, et al. Opportunities and challenges for ChatGPT and large language models in biomedicine and health. Brief Bioinform. 2024;25:bbad493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Amadeus S, Cenggoro TW, Budiarto A, Pardamean B. A design of polygenic risk model with deep learning for colorectal cancer in multiethnic Indonesians. Procedia Comput Sci. 2021;179:632. [Google Scholar]
  • 25. Hampsey DM, Ernst JF, Stewart JW, Sherman F. Multiple base-pair mutations in yeast. J Mol Biol. 1988;201:471-486. [DOI] [PubMed] [Google Scholar]
  • 26. Shin JE, Riesselman AJ, Kollasch AW, et al. Protein design and variant prediction using autoregressive generative models. Nat Commun. 2021;12:2403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Fan X, Pan H, Tian A, Chung WK, Shen Y. SHINE: protein language model-based pathogenicity prediction for short inframe insertion and deletion variants. Brief Bioinform. 2022;24:bbac584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Danzi MC, Dohrn MF, Fazal S, et al. Deep structured learning for variant prioritization in Mendelian diseases. Nat Commun. 2023;14:4167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Luo Y, Chen Y, Xie H, Zhu W, Zhang G. Interpretable CRISPR/Cas9 off-target activities with mismatches and indels prediction using BERT. Comput Biol Med. 2024;169:107932. [DOI] [PubMed] [Google Scholar]
  • 30. Lander ES. Initial impact of the sequencing of the human genome. Nature. 2011;470:187-197. [DOI] [PubMed] [Google Scholar]
  • 31. Gaulton KJ, Preissl S, Ren B. Interpreting non-coding disease-associated human variants using single-cell epigenomics. Nat Rev Genet. 2023;24:516-534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Andersson R, Sandelin A. Determinants of enhancer and promoter activities of regulatory elements. Nat Rev Genet. 2020;21:71-87. [DOI] [PubMed] [Google Scholar]
  • 33. West AG, Gaszner M, Felsenfeld G. Insulators: many functions, many mechanisms. Genes Dev. 2002;16:271-288. [DOI] [PubMed] [Google Scholar]
  • 34. Mantegna RN, Buldyrev SV, Goldberger AL, et al. Systematic analysis of coding and noncoding DNA sequences using methods of statistical linguistics. Phys Rev E Stat Phys Plasmas Fluids Relat Interdiscip Topics. 1995;52:2939-2950. [DOI] [PubMed] [Google Scholar]
  • 35. Ji Y, Zhou Z, Liu H, Davuluri RV. DNABERT: pre-trained bidirectional encoder representations from transformers model for DNA-language in genome. Bioinformatics. 2021;37:2112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Diaz DJ, Kulikova AV, Ellington AD, Wilke CO. Using machine learning to predict the effects and consequences of mutations in proteins. Curr Opin Struct Biol. 2023;78:102518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Nilforoshan H, Moor M, Roohani Y, et al. Zero-shot causal learning. Adv Neural Inf Process Syst. 2023;36:6862-6901. [Google Scholar]
  • 38. Larochelle H, Erhan D, Bengio Y. Zero-data learning of new tasks. AAAI. 2008;1:3. [Google Scholar]
  • 39. Meier J, Rao R, Verkuil R, Liu J, Sercu T, Rives A. Language models enable zero-shot prediction of the effects of mutations on protein function. Adv Neural Inf Process Syst. 2021;34:29287-29303. [Google Scholar]
  • 40. Liu X, Yang X, Ouyang L, et al. Protein language model predicts mutation pathogenicity and clinical prognosis. Cold Spring Harb Lab. Published online October 9, 2022. doi: 10.1101/2022.09.30.510294 [DOI] [Google Scholar]
  • 41. Nguyen E, Poli M, Durrant MG, et al. Sequence modeling and design from molecular to genome scale with Evo. Science. 2024;386:eado9336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Zhou H, Shrikumar A, Kundaje A, eds. Towards a better understanding of reverse-complement equivariance for deep learning models in genomics. In: Machine Learning in Computational Biology. PMLR; 2022:1-33. [Google Scholar]
  • 43. Benegas G, Albors C, Aw AJ, Ye C, Song YS. GPN-MSA: an alignment-based DNA language model for genome-wide variant effect prediction. bioRxiv. 2023. doi: 10.1101/2023.10.10.561776 [DOI] [Google Scholar]
  • 44. Frazer KA, Murray SS, Schork NJ, Topol EJ. Human genetic variation and its contribution to complex traits. Nat Rev Genet. 2009;10:241-251. [DOI] [PubMed] [Google Scholar]
  • 45. Toneyan S, Tang Z, Koo PK. Evaluating deep learning for predicting epigenomic profiles. Nat Mach Intell. 2022;4:1088-1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Lee NK, Tang Z, Toneyan S, Koo PK. EvoAug: improving generalization and interpretability of genomic deep neural networks with evolution-inspired data augmentations. Genome Biol. 2023;24:105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Webster JJ, Kit C, eds. Tokenization as the initial phase in NLP. In: COLING 1992 Volume 4: The 14th International Conference on Computational Linguistics. ACM; 1992. [Google Scholar]
  • 48. Palmer DD, ed. Tokenisation and sentence segmentation. In: Handbook of Natural Language Processing. Taylor and Francis; 2000:11-35. [Google Scholar]
  • 49. Mielke SJ, Alyafeai Z, Salesky E, et al. Between words and characters: a brief history of open-vocabulary modeling and tokenization in NLP. arXiv [Preprint]. doi: 10.48550/arXiv.211210508 [DOI] [Google Scholar]
  • 50. Cooper DN, Youssoufian H. The CpG dinucleotide and human genetic disease. Hum Genet. 1988;78:151-155. [DOI] [PubMed] [Google Scholar]
  • 51. anabria M, Hirsch J, Joubert PM, Poetsch AR. DNA language model GROVER learns sequence context in the human genome. Nat Mach Intell. 2024:1-13. [Google Scholar]
  • 52. Gage P. A new algorithm for data compression. C User J. 1994;12:23-38. [Google Scholar]
  • 53. Lindsey LM, Pershing NL, Habib A, Stephens WZ, Blaschke AJ, Sundar H. A comparison of tokenization impact in attention based and state space genomic language models. bioRxiv. 2024. doi: 10.1101/2024.09.09.612081 [DOI] [Google Scholar]
  • 54. Zhou Z, Ji Y, Li W, Ramana PD, Davuluri V, Liu H. DNABERT-2: efficient foundation model and benchmark for multi-species genome. arXiv [preprint]. doi: 10.48550/arXiv.2306.15006 [DOI] [Google Scholar]
  • 55. Bozinovski S, Fulgosi A, eds. The influence of pattern similarity and transfer learning on the base perceptron training. In: Proceedings of Symposium Informatica. IEEE; 1976. [Google Scholar]
  • 56. Thrun S, Pratt L, eds. Learning to learn: introduction and overview. In: Learning to Learn. Springer; 1998:3-17. [Google Scholar]
  • 57. Pan SJ, Yang Q. A survey on transfer learning. IEEE Trans Knowl Data Eng. 2009;22:1345-1359. [Google Scholar]
  • 58. Kalyan KS, Rajasekharan A, Sangeetha S. AMMU: a survey of transformer-based biomedical pretrained language models. J Biomed Inform. 2022;126:103982. [DOI] [PubMed] [Google Scholar]
  • 59. Riccio C, Jansen ML, Guo L, Ziegler A. Variant effect predictors: a systematic review and practical guide. Hum Genet. 2024;143:625-634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Henikoff S, Henikoff JG. Amino acid substitution matrices from protein blocks. Proc Natl Acad Sci U S A. 1992;89:10915-10919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Chan PA, Duraisamy S, Miller PJ, et al. Interpreting missense variants: comparing computational methods in human disease genes CDKN2A, MLH1, MSH2, MECP2, and tyrosinase (TYR). Hum Mutat. 2007;28:683-693. [DOI] [PubMed] [Google Scholar]
  • 62. Shauli T, Brandes N, Linial M. Evolutionary and functional lessons from human-specific amino acid substitution matrices. NAR Genom Bioinform. 2021;3:lqab079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Ng PC, Henikoff S. Predicting deleterious amino acid substitutions. Genome Res. 2001;11:863-874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Choi Y, Sims GE, Murphy S, Miller JR, Chan AP. Predicting the functional effect of amino acid substitutions and indels. PLoS ONE. 2012;7:e46688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Ramensky V, Bork P, Sunyaev S. Human non-synonymous SNPs: server and survey. Nucleic Acids Res. 2002;30:3894-3900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Adzhubei IA, Schmidt S, Peshkin L, et al. A method and server for predicting damaging missense mutations. Nat Methods. 2010;7:248-249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Bairoch A, Boeckmann B. The Swiss-Prot protein sequence data bank. Nucleic Acids Res. 1992;20:2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Capriotti E, Calabrese R, Casadio R. Predicting the insurgence of human genetic diseases associated to single point protein mutations with support vector machines and evolutionary information. Bioinformatics. 2006;22:2729-2734. [DOI] [PubMed] [Google Scholar]
  • 69. UniProt Consortium. UniProt: the universal protein knowledgebase in 2021. Nucleic Acids Res. 2021;49:D480-D489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. McLaren W, Gil L, Hunt SE, et al. The Ensembl Variant Effect Predictor. Genome Biol. 2016;17:122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Frazer J, Notin P, Dias M, et al. Disease variant prediction with deep generative models of evolutionary data. Nature. 2021;599:91-95. [DOI] [PubMed] [Google Scholar]
  • 72. Richards S, Aziz N, Bale S, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American college of medical genetics and genomics and the association for molecular pathology. Genet Med. 2015;17:405-424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Durkie M, Cassidy E-J, Berry I, et al. ACGS best practice guidelines for variant classification in rare disease 2024. 2023. [DOI] [PubMed] [Google Scholar]
  • 74. Weaver W, ed. Translation. In: Machine Translation of Languages. Rockfeller Foundation; 1949. [Google Scholar]
  • 75. Shannon CE. The redundancy of English. In: Von Forester H, ed. Cybernetics; Transactions of the 7th Conference. Josiah Macy Jr. Foundation; 1951:248-272. [Google Scholar]
  • 76. Sparck Jones K. Natural language processing: a historical review. In: Zampolli A, Calzolari N, Palmer M, eds. Current Issues in Computational Linguistics: In Honour of Don Walker; 1994:3-16. [Google Scholar]
  • 77. Galley M, McKeown K, eds. Lexicalized Markov grammars for sentence compression. In: Human Language Technologies 2007: The Conference of the North American Chapter of the Association for Computational Linguistics; Proceedings of the Main Conference; 2007:180-187. [Google Scholar]
  • 78. Zhu L, Chen Y, Yuille A. Unsupervised learning of probabilistic grammar-Markov models for object categories. IEEE Trans Pattern Anal Mach Intell. 2008;31:114-128. [DOI] [PubMed] [Google Scholar]
  • 79. Brown PF, Cocke J, Della Pietra SA, et al. A statistical approach to machine translation. Comput Linguist. 1990;16:79-85. [Google Scholar]
  • 80. Ron D, Singer Y, Tishby N. The power of amnesia: learning probabilistic automata with variable memory length. Mach Learn. 1996;25:117-149. [Google Scholar]
  • 81. Lauriola I, Lavelli A, Aiolli F. An introduction to deep learning in natural language processing: models, techniques, and tools. Neurocomputing. 2022;470:443-456. [Google Scholar]
  • 82. Rumelhart DE, Hinton GE, Williams RJ. Learning internal representations by error propagation. In: De Rumelhart D, Mcclelland JL, eds. Parallel Distributed Processing, Explorations in the Microstructure of Cognition. vol 1. MIT Press; 1986:599-607. [Google Scholar]
  • 83. Bengio Y, Ducharme R, Vincent P. A neural probabilistic language model. In: Leen T, Dietterich T, Tresp V, eds. Advances in Neural Information Processing Systems. vol 13. MIT Press; 2000:1137-1155. [Google Scholar]
  • 84. LeCun Y, Bottou L, Bengio Y, Haffner P. Gradient-based learning applied to document recognition. Proc IEEE. 1998;86:2278-2324. [Google Scholar]
  • 85. Collobert R, Weston J, eds. A unified architecture for natural language processing: deep neural networks with multitask learning. In: Proceedings of the 25th International Conference on Machine Learning (ICML ’08). ACM; 2008:160-167. [Google Scholar]
  • 86. Zhou J, Lu Q, Xu R, Gui L, Wang H, eds. CNNsite: prediction of DNA-binding residues in proteins using convolutional neural network with sequence features. In: 2016 IEEE International Conference on Bioinformatics and Biomedicine (BIBM). IEEE; 2016:78-85. [Google Scholar]
  • 87. Kim Y, Denton C, Hoang L, Rush AM. Structured attention networks. arXiv [preprint]. doi: 10.48550/arXiv.1702.00887 [DOI] [Google Scholar]
  • 88. Kim S, Lee H, Kim K, Kang J. Mut2vec: distributed representation of cancerous mutations. BMC Med Genomics. 2018;11:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Le NQK, Yapp EKY, Nagasundaram N, Yeh HY. Classifying promoters by interpreting the hidden information of DNA sequences via deep learning and combination of continuous FastText n-grams. Front Bioeng Biotechnol. 2019;7:305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Wang W, Gang J, eds. Application of convolutional neural network in natural language processing. In: 2018 International Conference on Information Systems and Computer Aided Education (ICISCAE). IEEE; 2018:64-70. [Google Scholar]
  • 91. Mikolov T, Karafiát M, Burget L, Černocký J, Khudanpur S. Recurrent neural network based language model. Proc Interspeech. 2010;2:1045-1048. [Google Scholar]
  • 92. Hochreiter S. Long short-term memory. Neural Comput. 1997:9:1735-1780. [DOI] [PubMed] [Google Scholar]
  • 93. Cho K. Learning phrase representations using RNN encoder-decoder for statistical machine translation. arXiv [preprint]. 2014. doi: 10.48550/arXiv.1406.1078 [DOI] [Google Scholar]
  • 94. Rhanoui M, Mikram M, Yousfi S, Barzali S. A CNN-BiLSTM model for document-level sentiment analysis. Mach Learn Knowl Extr. 2019;1:832-847. [Google Scholar]
  • 95. Lin J, Zhang Z, Zhang S, Chen J, Wong K-C. CRISPR-Net: a recurrent convolutional network quantifies CRISPR off-target activities with mismatches and indels. Adv Sci. 2020;7:1903562. [Google Scholar]
  • 96. Benegas G, Batra S, Song YS. DNA language models are powerful predictors of genome-wide variant effects. Proc Natl Acad Sci U S A. 2023;120:e2311219120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Pejaver V, Urresti J, Lugo-Martinez J, et al. Inferring the molecular and phenotypic impact of amino acid variants with MutPred2. Nat Commun. 2020;11:5918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Dunham AS, Beltrao P, AlQuraishi M. High-throughput deep learning variant effect prediction with sequence UNET. Genome Biol. 2023;24:110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Tan W, Shen Y. Multimodal learning of noncoding variant effects using genome sequence and chromatin structure. Bioinformatics. 2023;39:btad541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Cheng L, Yu T, Khalitov R, Yang Z. Self-supervised learning for DNA sequences with circular dilated convolutional networks. Neural Netw. 2023;171:466. [DOI] [PubMed] [Google Scholar]
  • 101. Liu NF, Lin K, Hewitt J, et al. Lost in the middle: how language models use long contexts. Trans Assoc Comput Linguist. 2024;12:157-173. [Google Scholar]
  • 102. Bahdanau D. Neural machine translation by jointly learning to align and translate. arXiv [preprint]. 2014. doi: 10.48550/arXiv.1409.0473 [DOI] [Google Scholar]
  • 103. Wu Y. Google’s neural machine translation system: bridging the gap between human and machine translation. arXiv [preprint]. 2016. doi: 10.48550/arXiv.1609.08144 [DOI] [Google Scholar]
  • 104. Le NQK. Transforming peptide hormone prediction: the role of AI in modern proteomics. Proteomics. 2024;25:2400156. [DOI] [PubMed] [Google Scholar]
  • 105. Hu RS, Gu K, Ehsan M, Abbas Raza SH, Wang CR. Transformer-based deep learning enables improved B-cell epitope prediction in parasitic pathogens: a proof-of-concept study on Fasciola hepatica. PLoS Negl Trop Dis. 2025;19:e0012985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Avsec Ž, Agarwal V, Visentin D, et al. Effective gene expression prediction from sequence by integrating long-range interactions. Nat Methods. 2021;18:1196-1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Nguyen E, Poli M, Faizi M, et al. HyenaDNA: long-range genomic sequence modeling at single nucleotide resolution. In: Jordan MI, Solla SA, LeCun Y, eds. Advances in Neural Information Processing Systems. vol 36. MIT Press; 2023:43177-43201. [Google Scholar]
  • 108. Gu A, Dao T. Mamba: linear-time sequence modeling with selective state spaces. arXiv [preprint]. 2024. doi: 10.48550/arXiv.2312.00752 [DOI] [Google Scholar]
  • 109. Hendrycks D, Gimpel K. Gaussian error linear units (GELUs). arXiv [preprint]. 2016. doi: 10.48550/arXiv1606.08415 [DOI] [Google Scholar]
  • 110. Ramachandran P, Zoph B, Le QV. Searching for activation functions. arXiv [preprint]. 2017. doi: 10.48550/arXiv.1710.05941 [DOI] [Google Scholar]
  • 111. Radford A. Improving language understanding by generative pre-training. 2018. https://www.mikecaptain.com/resources/pdf/GPT-1.pdf
  • 112. Floridi L, Chiriatti M. GPT-3: its nature, scope, limits, and consequences. Minds Mach. 2020;30:681-694. [Google Scholar]
  • 113. Achiam J, Adler S, Agarwal S, et al. GPT-4 technical report. arXiv [preprint]. 2023. doi: 10.48550/arXiv.2303.08774 [DOI] [Google Scholar]
  • 114. Devlin J, Chang M-W, Lee K, Toutanova K, eds. BERT: pre-training of deep bidirectional transformers for language understanding. In: Proc NAACL-HLT. vol 1. ACL; 2019:2. [Google Scholar]
  • 115. Shreyashree S, Sunagar P, Rajarajeswari S, Kanavalli A. A literature review on bidirectional encoder representations from transformers. In: Smys S, Balas VE, Palanisamy R, eds. Inventive Computation and Information Technologies: Proceedings of ICICIT. Springer; 2022:305-320. [Google Scholar]
  • 116. Brandes N, Ofer D, Peleg Y, Rappoport N, Linial M. ProteinBERT: a universal deep-learning model of protein sequence and function. Bioinformatics. 2022;38:2102-2110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Wang X, Li F, Zhang Y, et al. Deep learning approaches for non-coding genetic variant effect prediction: current progress and future prospects. Brief Bioinform. 2024;25:bbae446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Yang J, Das Adhikari S, Wang H, et al. De novo prediction of functional effects of genetic variants from DNA sequences based on context-specific molecular information. Front Syst Biol. 2024;4:1402664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Bommasani R, Hudson DA, Adeli E, et al. On the opportunities and risks of foundation models. arXiv [preprint]. 2022. doi: 10.48550/arXiv.2108.07258 [DOI] [Google Scholar]
  • 120. Zhou C, Li Q, Li C, et al. A comprehensive survey on pretrained foundation models: a history from BERT to ChatGPT. Int J Mach Learn Cybern. 2024:1-65. [Google Scholar]
  • 121. Li Q, Hu Z, Wang Y, et al. Progress and opportunities of foundation models in bioinformatics. Brief Bioinform. 2024;25:bbae548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Dalla-Torre H, Gonzalez L, Mendoza-Revilla J, et al. Nucleotide transformer: building and evaluating robust foundation models for human genomics. Nat Methods. 2024;22:287-297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Rives A, Meier J, Sercu T, et al. Biological structure and function emerge from scaling unsupervised learning to 250 million protein sequences. Proc Natl Acad Sci U S A. 2021;118:e2016239118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Lin Z, Akin H, Rao R, et al. Evolutionary-scale prediction of atomic-level protein structure with a language model. Science. 2023;379:1123-1130. [DOI] [PubMed] [Google Scholar]
  • 125. Zhu C, Ping W, Xiao C, et al. Long-short transformer: efficient transformers for language and vision. In: Ranzato M, Beygelzimer A, Dauphin Y, Liang PS, Wortman Vaughan J, eds. Advances in Neural Information Processing Systems. vol 34. Curran Associates Inc; 2021:17723-17736. [Google Scholar]
  • 126. Poli M, Massaroli S, Nguyen E, et al. Hyena hierarchy: towards larger convolutional language models. In: Krause A, Brunskill E, Cho K, Engelhardt B, Sabato S, Scarlett J, eds. Proceedings of the 40th International Conference on Machine Learning. vol 202; 2023:28043-28078. [Google Scholar]
  • 127. Consens ME, Dufault C, Wainberg M, et al. To transformers and beyond: large language models for the genome. arXiv [preprint]. 2023. doi: 10.48550/arXiv.2311.07621 [DOI] [Google Scholar]
  • 128. Huang J, Tong S, Singh P, Shi M, Liu C. White paper on global artificial intelligence environmental impact. 2024. doi:10.13140/RG.2.2.17090.39368 [Google Scholar]
  • 129. Jiang P, Sonne C, Li W, You F, You S. Preventing the immense increase in the life-cycle energy and carbon footprints of LLM-powered intelligent chatbots. Engineering. 2024;40:202-210. [Google Scholar]
  • 130. Marquet C, Schlensok J, Abakarova M, Rost B, Laine E. Expert-guided protein language models enable accurate and blazingly fast fitness prediction. Bioinformatics. 2024;40:btae621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Derbel H, Zhao Z, Liu Q. Accurate prediction of functional effect of single amino acid variants with deep learning. Comput Struct Biotechnol J. 2023;21:5776-5784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Lin W, Wells J, Wang Z, Orengo C, Martin ACR. Enhancing missense variant pathogenicity prediction with protein language models using Varipred. Sci Rep. 2024;14:8136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Zhan H, Zhang Z. DYNA: disease-specific language model for variant pathogenicity. arXiv [preprint]. 2024. doi: 10.48550/arXiv.2406.00164 [DOI] [Google Scholar]
  • 134. Yan Z, Ge F, Liu Y, et al. TranSEFvP: a two-stage approach for the prediction of human pathogenic variants based on protein sequence embedding fusion. J Chem Inf Model. 2024;64:1407. [DOI] [PubMed] [Google Scholar]
  • 135. Manfredi M, Savojardo C, Martelli PL, Casadio R. E-SNPs\&GO: embedding of protein sequence and function improves the annotation of human pathogenic variants. Bioinformatics. 2022;38:5168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Borgeaud S, Mensch A, Hoffmann J, et al. Improving language models by retrieving from trillions of tokens. In: International Conference on Machine Learning. ICML; 2022:2206-2240. [Google Scholar]
  • 137. Liu M, Watson LT, Zhang L. Quantitative prediction of the effect of genetic variation using hidden Markov models. BMC Bioinformatics. 2014;15:1-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Capriotti E, Fariselli P. Evaluating the relevance of sequence conservation in the prediction of pathogenic missense variants. Hum Genet. 2022;141:1649-1658. [DOI] [PubMed] [Google Scholar]
  • 139. Blaabjerg LM, Jonsson N, Boomsma W, Stein A, Lindorff-Larsen K. SSEmb: a joint embedding of protein sequence and structure enables robust variant effect predictions. Nat Commun. 2024;15:9646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Livesey BJ, Marsh JA. Using deep mutational scanning to benchmark variant effect predictors and identify disease mutations. Mol Syst Biol. 2020;16:e9380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Ranwez V, Chantret NN. Strengths and limits of multiple sequence alignment and filtering methods. In: Scornavacca C, Delsuc F, Galtier N, eds. Phylogenetics in the Genomic Era. Open Access; 2020:2-36. [Google Scholar]
  • 142. Brandes N, Goldman G, Wang CH, Ye CJ, Ntranos V. Genome-wide prediction of disease variant effects with a deep protein language model. Nat Genet. 2023;55:1512-1522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Livesey BJ, Marsh JA. Updated benchmarking of variant effect predictors using deep mutational scanning. Mol Syst Biol. 2023;19:e11474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Rao RM, Liu J, Verkuil R, et al. , eds. MSA transformer. In: International Conference on Machine Learning. PMLR; 2021:8844-8856. [Google Scholar]
  • 145. Shigaki D, Adato O, Adhikari AN, et al. Integration of multiple epigenomic marks improves prediction of variant impact in saturation mutagenesis reporter assay. Hum Mutat. 2019;40:1280-1291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Koo PK, Majdandzic A, Ploenzke M, Anand P, Paul SB. Global importance analysis: an interpretability method to quantify importance of genomic features in deep neural networks. PLoS Comput Biol. 2021;17:e1008925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Hidayat AH, Trinugroho JP, Nirwantono R, Sudigyo D, Pardamean B. Utilizing semi-supervised method in predicting BRCA1 pathogenicity variants. Procedia Comput Sci. 2023;227:36. [Google Scholar]
  • 148. Truong TF, Bepler T, eds. POET: a generative model of protein families as sequences-of-sequences. In: Advances in Neural Information Processing Systems. 2023:77379-77415. [Google Scholar]
  • 149. Gao Z, Liu Q, Zeng W, Jiang R, Wong WH. EpiGePT: A Pretrained Transformer Model For Epigenomics. Technical Report. Cold Spring Harbor Laboratory; 2023. [Google Scholar]
  • 150. Liu Y, Yeung WSB, Chiu PCN, Cao D. Computational approaches for predicting variant impact: an overview from resources, principles to applications. Front Genet. 2022;13:981005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Wild B, Upmeier zu Belzen J, Herrmann L, Kittner P, Eils R, eds. DNA language models identify variants predictive across the human phenome. In: ICLR 2024 Workshop on Machine Learning for Genomics Explorations. ICLR; 2024:1-13. [Google Scholar]
  • 152. Li K, Zhong Y, Lin X, Quan Z. Predicting the disease risk of protein mutation sequences with pretraining model. Front Genet. 2020;11:605620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Yamaguchi H, Saito Y. Evotuning protocols for transformer-based variant effect prediction on multidomain proteins. Brief Bioinform. 2021;22:bbab234. [DOI] [PubMed] [Google Scholar]
  • 154. Marquet C, Heinzinger M, Olenyi T, et al. Embeddings from protein language models predict conservation and variant effects. Hum Genet. 2021;141:1629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Elnaggar A, Heinzinger M, Dallago C, et al. ProtTrans: toward understanding the language of life through self-supervised learning. IEEE Trans Pattern Anal Mach Intell. 2021;44:7112-7127. [DOI] [PubMed] [Google Scholar]
  • 156. Yang M, Huang L, Huang H, et al. Integrating convolution and self-attention improves language model of human genome for interpreting non-coding regions at base-resolution. Nucleic Acids Res. 2022;50:e81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Olenyi T, Marquet C, Heinzinger M, et al. LambdaPP: fast and accessible protein-specific phenotype predictions. Protein Sci. 2022;32:e4524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Zhou A, Landolfi NC, O’Neill DC. Unsupervised language models for disease variant prediction. arXiv [preprint]. 2022. doi: 10.48550/arXiv.2212.03979 [DOI] [Google Scholar]
  • 159. Dampier W, Link RW, Earl JP, et al. HIV-BERT: bidirectional encoder representations from transformers for accelerating HIV deep learning tasks. Front Virol. 2022;2:880618. [Google Scholar]
  • 160. Jiang TT, Fang L, Wang K. Deciphering ‘the language of nature’: a transformer-based language model for deleterious mutations in proteins. Innovation. 2023;4:100487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Sun Y, Shen Y. Structure-Informed Protein Language Models are Robust Predictors for Variant Effects. Technical Report. Research Square Platform LLC; 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. James JK, Norland K, Johar AS, Kullo IJ. Deep generative models of LDLR protein structure to predict variant pathogenicity. J Lipid Res. 2023;64:100455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Jumper J, Evans R, Pritzel A, et al. Highly accurate protein structure prediction with AlphaFold. Nature. 2021;596:583-589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Cheng J, Novati G, Pan J, et al. Accurate proteome-wide missense variant effect prediction with AlphaMissense. Science. 2023;381:eadg7492. [DOI] [PubMed] [Google Scholar]
  • 165. Qu Y, Niu Z, Ding Q, et al. Ensemble learning with supervised methods based on large-scale protein language models for protein mutation effects prediction. Int J Mol Sci. 2023;24:16496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Notin P, Dias M, Frazer J, et al. , eds. Tranception: protein fitness prediction with autoregressive transformers and inference-time retrieval. In: Proceedings of the International Conference on Machine Learning. ICML; 2022:16990-17017. [Google Scholar]
  • 167. Lafita A, Gonzalez F, Hossam M, et al. Fine-tuning protein language models with deep mutational scanning improves variant effect prediction. arXiv [preprint]. 2024. doi: 10.48550/arXiv.2405.06729 [DOI] [Google Scholar]
  • 168. Shulgina Y, Trinidad MI, Langeberg CJ, et al. RNA language models predict mutations that improve RNA function. bioRxiv. 2024. doi: 10.1101/2024.04.05.588317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Yang Y, Wang J, Wang X, et al. , eds. TransVPath: a TabTransformer-based model for predicting the pathogenicity of structural variants. In: 2024 IEEE Int Conf Bioinform Biomed. BIBM; 2024:1289-1295. [Google Scholar]
  • 170. Huang X, Khetan A, Cvitkovic M, Karnin Z. TabTransformer: tabular data modeling using contextual embeddings. arXiv [preprint]. 2020. doi: 10.48550/arXiv.2012.06678 [DOI] [Google Scholar]
  • 171. Li ZX, Huang WK, Li HD, eds. MVFormer: predicting the pathogenicity of missense variants using gated transformers. In: Proceedings 15th ACM International Conference Bioinform Computational Biology Health Informatics. ACM; 2024:1-6. [Google Scholar]
  • 172. Zhong G, Zhao Y, Zhuang D, Chung WK, Shen Y. PreMode predicts mode of action of missense variants by deep graph representation learning of protein sequence and structural context. bioRxiv. 2024. doi: 10.1101/2024.02.20.581321 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Linder J, Srivastava D, Yuan H, Agarwal V, Kelley DR. Predicting RNA-seq coverage from DNA sequence as a unifying model of gene regulation. Nat Genet. 2025;57:949-961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Joshi D, Pradhan S, Sajeed R, Srinivasan R, Rana S. An augmented transformer model trained on protein family specific variant data leads to improved prediction of variants of uncertain significance. Hum Genet. 2025;144:143-158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Glaser M, Braegelmann J. ESM-Effect: an effective and efficient fine-tuning framework towards accurate prediction of mutation’s functional effect. bioRxiv. 2025. doi: 10.1101/2025.02.03.635741 [DOI] [Google Scholar]
  • 176. Kryshtafovych A, Schwede T, Topf M, Fidelis K, Moult J. Critical assessment of methods of protein structure prediction (CASP) – round XIII. Proteins. 2019;87:1011-1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Kryshtafovych A, Schwede T, Topf M, Fidelis K, Moult J. Critical assessment of methods of protein structure prediction (CASP) – round XIV. Proteins. 2021;89:1607-1617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Landrum MJ, Lee JM, Benson M, et al. ClinVar: improving access to variant interpretations and supporting evidence. Nucleic Acids Res. 2018;46:D1062-D1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Miller DT, Lee K, Abul-Husn NS, et al. ACMG SF v3.1 list for reporting of secondary findings in clinical exome and genome sequencing: a policy statement of the American college of medical genetics and genomics (ACMG). Genet Med. 2022;24:1407-1414. [DOI] [PubMed] [Google Scholar]
  • 180. Scott M, Lee S-I, et al. A unified approach to interpreting model predictions. Adv Neural Inf Process Syst. 2017;30:4765-4774. [Google Scholar]
  • 181. Jiang F, Doudna JA. CRISPR–Cas9 structures and mechanisms. Annu Rev Biophys. 2017;46:505-529. [DOI] [PubMed] [Google Scholar]
  • 182. Landrum MJ, Lee JM, Benson M, et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 2016;44:D862-D868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Karczewski KJ, Francioli LC, Tiao G, et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature. 2020;581:434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Stenson PD, Mort M, Ball EV, et al. The human gene mutation database (HGMD®): optimizing its use in a clinical diagnostic or research setting. Hum Genet. 2020;139:1197-1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Apweiler R, Bairoch A, Wu CH, et al. UniProt: the universal protein knowledgebase. Nucleic Acids Res. 2004;32:D115-D119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Kao CH, Trop E, Polen M, et al. Advancing DNA language models: the genomics long-range benchmark. In: ICLR 2024 Workshop on Machine Learning for Genomics Explorations. Vol. 5. ICLR; 2024. [Google Scholar]
  • 187. Schiff Y, Kao CH, Gokaslan A, Dao T, Gu A, Kuleshov V. Caduceus: bidirectional equivariant long-range DNA sequence modeling. arXiv [preprint]. 2024. doi: 10.48550/arXiv.2403.03234 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Miele A, Dekker J. Long-range chromosomal interactions and gene regulation. Mol Biosyst. 2008;4:1046-1057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Noonan JP, McCallion AS. Genomics of long-range regulatory elements. Annu Rev Genomics Hum Genet. 2010;11:1-23. [DOI] [PubMed] [Google Scholar]
  • 190. Chen Y, Bates DL, Dey R, et al. DNA binding by GATA transcription factor suggests mechanisms of DNA looping and long-range gene regulation. Cell Rep. 2012;2:1197-1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Nasser J, Bergman DT, Fulco CP, et al. Genome-wide enhancer maps link risk variants to disease genes. Nature. 2021;593:238-243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Poli M, Hie B, Thomas AW, Bybee M. StripedHyena: moving beyond transformers with hybrid signal processing models. Github Repository. 2023. Accessed August 12, 2025. https://github.com/togethercomputer/stripedhyena [Google Scholar]
  • 193. Blake WJ, Kærn M, Cantor CR, Collins JJ. Noise in eukaryotic gene expression. Nature. 2003;422:633-637. [DOI] [PubMed] [Google Scholar]
  • 194. Munsky B, Neuert G, Van Oudenaarden A. Using gene expression noise to understand gene regulation. Science. 2012;336:183-187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Chen J, Hu Z, Sun S, et al. Interpretable RNA foundation model from unannotated data for highly accurate RNA structure and function predictions. arXiv [preprint]. 2022. doi: 10.48550/arXiv.2204.00300 [DOI] [Google Scholar]
  • 196. Hudson TJ, Anderson W, Artez A, et al. International network of cancer genome projects. Nature. 2010;464:993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Gonzalez-Perez A, Perez-Llamas C, Deu-Pons J, et al. IntOGen-mutations identifies cancer drivers across tumor types. Nat Methods. 2013;10:1081-1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Sherry ST, Ward MH, Kholodov M, et al. DbSNP: the NCBI database of genetic variation. Nucleic Acids Res. 2001;29:308-311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Mottaz A, David FPA, Veuthey A-L, Yip YL. Easy retrieval of single amino-acid polymorphisms and phenotype information using SwissVar. Bioinformatics. 2010;26:851-852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Riesselman AJ, Ingraham JB, Marks DS. Deep generative models of genetic variation capture the effects of mutations. Nat Methods. 2018;15:816-822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. AlQuraishi M. ProteinNet: a standardized data set for machine learning of protein structure. BMC Bioinformatics. 2019;20:1-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Sayers EW, et al. Database resources of the National Center for Biotechnology Information in 2025. Nucleic Acids Res. 2025;53(D1):D20-D29. doi:10.1093/nar/gkae979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Togninalli M, et al. The AraGWAS Catalog: a curated and standardized Arabidopsis thaliana GWAS catalog. Nucleic Acids Res. 2018;46(D1):D1150–D1156. doi.org/10.1093/nar/gkx954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Zhou J, Troyanskaya OG. Predicting effects of noncoding variants with deep learning–based sequence model. Nat Methods. 2015;12:931-934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Chen KM, Wong AK, Troyanskaya OG, Zhou J. A sequence-based global map of regulatory activity for deciphering human genetics. Nat Genet. 2022;54:940-949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Biggs H, Parthasarathy P, Gavryushkina A, Gardner PP. NCVarDB: a manually curated database for pathogenic non-coding variants and benign controls. Database. 2020;2020:baaa105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Gray VE, Hause RJ, Luebeck J, Shendure J, Fowler DM. Quantitative missense variant effect prediction using large-scale mutagenesis data. Cell Syst. 2018;6:116-124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Suzek BE, Huang H, McGarvey P, Mazumder R, Wu CH. UniRef: comprehensive and non-redundant UniProt reference clusters. Bioinformatics. 2007;23:1282-1288. [DOI] [PubMed] [Google Scholar]
  • 209. Yusuf I, Pardamean B, Baurley JW, et al. Genetic risk factors for colorectal cancer in multiethnic Indonesians. Sci Rep. 2021;11:9988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210. Hecht M, Bromberg Y, Rost B. Better prediction of functional effects for sequence variants. BMC Genomics. 2015;16 suppl 8:S1-S12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Reeb J, Wirth T, Rost B. Variant effect predictions capture some aspects of deep mutational scanning experiments. BMC Bioinformatics. 2020;21:1-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Rentzsch P, Witten D, Cooper GM, Shendure J, Kircher M. CADD: predicting the deleteriousness of variants throughout the human genome. Nucleic Acids Res. 2019;47:D886-D894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Chang MT, Bhattarai TS, Schram AM, et al. Accelerating discovery of functional mutant alleles in cancer. Cancer Discov. 2018;8:174-183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Kaplanis J, Samocha KE, Wiel L, et al. Evidence for 28 genetic disorders discovered by combining healthcare and research data. Nature. 2020;586:757-762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Khare S, Gurry C, Freitas L, et al. GISAID’s role in pandemic response. China CDC Wkly. 2021;3:1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216. Sundaram L, Gao H, Padigepati SR, et al. Predicting the clinical impact of human mutation with deep neural networks. Nat Genet. 2018;50:1161-1170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. National Heart Lung Blood Institute others. GRASP: genome-wide repository of associations between SNPs and phenotypes. 2021. https://grasp.nhlbi.nih.gov/
  • 218. Lambert SA, Gil L, Jupp S, et al. The polygenic score catalog as an open database for reproducibility and systematic evaluation. Nat Genet. 2021;53:420-425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Chuai G, Ma H, Yan J, et al. DeepCRISPR: optimized CRISPR guide RNA design by deep learning. Genome Biol. 2018;19:1-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. Wang QS, Kelley DR, Ulirsch J, et al. Leveraging supervised learning for functionally informed fine-mapping of cis-eQTLs identifies an additional 20,913 putative causal eQTLs. Nat Commun. 2021;12:3394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221. Notin P, Kollasch AW, Ritter D, et al. ProteinGym: large-scale benchmarks for protein design and fitness prediction. bioRxiv. 2023. doi: 10.1101/2023.12.07.570727 [DOI] [Google Scholar]
  • 222. Kobori S, Nomura Y, Miu A, Yokobayashi Y. High-throughput assay and engineering of self-cleaving ribozymes by sequencing. Nucleic Acids Res. 2015;43:e85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Shortle D. Protein fold recognition. Nat Struct Biol. 1995;2:91-93. [DOI] [PubMed] [Google Scholar]
  • 224. Marin FI, Teufel F, Horlacher M, et al. , eds. BEND: benchmarking DNA language models on biologically meaningful tasks. In: The Twelfth International Conference on Learning Representations. ICLR; 2024. [Google Scholar]
  • 225. Tabet DR, Kuang D, Lancaster MC, et al. Benchmarking computational variant effect predictors by their ability to infer human traits. Genome Biol. 2024;25:172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. SimilarWeb. chatgpt.com website analysis for December 2024. Accessed January 28, 2025. https://www.similarweb.com/website/chatgpt.com/
  • 227. Laine E, Karami Y, Carbone A. GEMME: a simple and fast global epistatic model predicting mutational effects. Mol Biol Evol. 2019;36:2604-2619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Jelinek F, Mercer RL, Bahl LR, Baker JK. Perplexity – a measure of the difficulty of speech recognition tasks. J Acoust Soc Am. 1977;62:S63. [Google Scholar]
  • 229. Jurafsky D, Martin JH. Speech and Language Processing. Prentice Hall; 2024. [Google Scholar]
  • 230. Meister C, Cotterell R. Language model evaluation beyond perplexity. arXiv. 2021. doi: 10.48550/arXiv.2106.00085 [DOI] [Google Scholar]
  • 231. Matthews BW. Comparison of the predicted and observed secondary structure of T4 phage lysozyme. Biochim Biophys Acta. 1975;405:442-451. [DOI] [PubMed] [Google Scholar]
  • 232. Chicco D, Jurman G. The Matthews correlation coefficient (MCC) should replace the ROC AUC as the standard metric for assessing binary classification. Biodata Min. 2023;16:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. Pearson KI. Mathematical contributions to the theory of evolution. – XI. On the influence of natural selection on the variability and correlation of organs. Philos Trans R Soc Lond A. 1903;200:1-66. [Google Scholar]
  • 234. Spearman C. The proof and measurement of association between two things. Am J Psychol. 1961;15:72-101. [PubMed] [Google Scholar]
  • 235. Jaccard P. Étude de la distribution florale dans une portion des Alpes et du Jura. Bull Soc Vaud Sci Nat. 1901;37:547-579. [Google Scholar]
  • 236. Lin CY, ed. ROUGE: a package for automatic evaluation of summaries. In: Text Summarization Branches Out. ACL; 2004:74-81. [Google Scholar]
  • 237. Rus V, Lintean M, eds. An optimal assessment of natural language student input using word-to-word similarity metrics. In: Intelligent Tutoring Systems: 11th International Conference. Springer; 2012:675-676. [Google Scholar]
  • 238. Forgues G, Pineau J, Larcheveque J-M, Tremblay R, eds. Bootstrapping dialog systems with word embeddings. In: NIPS Modern Machine Learning and Natural Language Processing Workshop. vol 2. NIPS; 2014:168. [Google Scholar]
  • 239. Xu S, Wu Z, Zhao H, et al. Reasoning before comparison: LLM-enhanced semantic similarity metrics for domain specialized text analysis. arXiv [preprint]. 2024. doi: 10.48550/arXiv.2402.11398 [DOI] [Google Scholar]
  • 240. Thompson NC, Greenewald K, Lee K, Manso GF. The computational limits of deep learning. arXiv [preprint]. 2020. doi: 10.48550/arXiv.2007.05558 [DOI] [Google Scholar]
  • 241. Justus D, Brennan J, Bonner S, McGough AS, eds. Predicting the computational cost of deep learning models. In: 2018 IEEE International Conference on Big Data (Big Data). IEEE; 2018:3873-3882. [Google Scholar]
  • 242. Pickrell JK, Marioni JC, Pai AA, et al. Understanding mechanisms underlying human gene expression variation with RNA sequencing. Nature. 2010;464:768-772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243. Manning KS, Cooper TA. The roles of RNA processing in translating genotype to phenotype. Nat Rev Mol Cell Biol. 2017;18:102-114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Tai KY, Dhaliwal J. Machine learning model for malaria risk prediction based on mutation location of large-scale genetic variation data. J Big Data. 2022;9:85. [Google Scholar]
  • 245. Das B, Kutsal M, Das R. Effective prediction of drug–target interaction on HIV using deep graph neural networks. Chemom Intell Lab Syst. 2022;230:104676. [Google Scholar]
  • 246. Grimm DG, Azencott CA, Aicheler F, et al. The evaluation of tools used to predict the impact of missense variants is hindered by two types of circularity. Hum Mutat. 2015;36:513-523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Biobank UK. Data-field 21000. Accessed February 2, 2025. https://biobank.ndph.ox.ac.uk/ukb/field.cgi?id=21000
  • 248. Lu YF, Goldstein DB, Angrist M, Cavalleri G. Personalized medicine and human genetic diversity. Cold Spring Harb Perspect Med. 2014;4:a008581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. Prohaska A, Racimo F, Schork AJ, et al. Human disease variation in the light of population genomics. Cell. 2019;177:115-131. [DOI] [PubMed] [Google Scholar]
  • 250. Obermeyer Z, Powers B, Vogeli C, Mullainathan S. Dissecting racial bias in an algorithm used to manage the health of populations. Science. 2019;366:447-453. [DOI] [PubMed] [Google Scholar]
  • 251. Chen A, Jonnagaddala J, Nekkantti C, Liaw S-T. Generation of surrogates for deidentification of electronic health records. In: Ohno-Machado L, Séroussi B, eds. MEDINFO 2019: Health and Wellbeing E-networks for All. IOS Press; 2019:70-73. [Google Scholar]
  • 252. Liu Z, Huang Y, Yu X, et al. DeID-GPT: zero-shot medical text de-identification by GPT-4. arXiv [preprint]. 2023. doi: 10.48550/arXiv.2303.11032 [DOI] [Google Scholar]
  • 253. Naveed M, Ayday E, Clayton EW, et al. Privacy in the genomic era. ACM Comput Surv. 2015;48:1-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Grishin D, Obbad K, Church GM. Data privacy in the age of personal genomics. Nat Biotechnol. 2019;37:1115-1117. [DOI] [PubMed] [Google Scholar]
  • 255. Bonomi L, Huang Y, Ohno-Machado L. Privacy challenges and research opportunities for genomic data sharing. Nat Genet. 2020;52:646-654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256. Shokri R, Stronati M, Song C, Shmatikov V, eds. Membership inference attacks against machine learning models. In: 2017 IEEE Symposium on Security and Privacy (SP). IEEE; 2017:3-18. [Google Scholar]
  • 257. Kandpal N, Pillutla K, Oprea A, Kairouz P, Choquette-Choo CA, Xu Z. User inference attacks on large language models. arXiv [preprint]. 2023. doi: 10.48550/arXiv.2310.09266 [DOI] [Google Scholar]
  • 258. Jagannatha A, Rawat BPS, Yu H. Membership inference attack susceptibility of clinical language models. arXiv [preprint]. 2021. doi.org/10.48550/arXiv.2104.08305 [Google Scholar]
  • 259. Jain S, Wallace BC. Attention is not explanation. arXiv [preprint]. 2019. doi: 10.48550/arXiv.1902.10186 [DOI] [Google Scholar]
  • 260. Wiegreffe S, Pinter Y. Attention is not not explanation. arXiv [preprint]. 2019. doi: 10.48550/arXiv.1908.04626 [DOI] [Google Scholar]
  • 261. Hao Y, Dong L, Wei F, Xu K, eds. Self-attention attribution: interpreting information interactions inside transformer. In: Proceedings of the AAAI Conference on Artificial Intelligence. vol 35. 2021:12963-12971. [Google Scholar]
  • 262. Yamada K, Hamada M. Prediction of RNA–protein interactions using a nucleotide language model. Bioinform Adv. 2022;2:vbac023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263. Mock F, Kretschmer F, Kriese A, Bocker S, Marz M. Taxonomic classification of DNA sequences beyond sequence similarity using deep neural networks. Proc Natl Acad Sci U S A. 2022;119:e2122636119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264. Qiu X, Shao S, Wang H, Tan X. BioK-transformer: a pre-trained transformer-based sequence-to-sequence model for adverse drug reactions prediction. Comput Methods Programs Biomed. 2025;260:108524. [DOI] [PubMed] [Google Scholar]
  • 265. Van der Maaten L, Hinton G. Visualizing data using t-SNE. J Mach Learn Res. 2008;9:2579-2605. [Google Scholar]
  • 266. Zhang L, Liu T. PDNAPred: interpretable prediction of protein-DNA binding sites based on pre-trained protein language models. Int J Biol Macromol. 2024;281:136147. [DOI] [PubMed] [Google Scholar]
  • 267. Lyu D, Wang X, Chen Y, Wang F. Language model and its interpretability in biomedicine: a scoping review. iScience. 2024;27:109334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268. Zhang P, Zeng G, Wang T, Lu W. TinyLlama: an open-source small language model. arXiv [preprint]. 2024. doi: 10.48550/arXiv.2401.02385 [DOI] [Google Scholar]
  • 269. Touvron H, Martin L, Stone K, et al. LLaMA 2: open foundation and fine-tuned chat models. arXiv [preprint]. 2023. doi: 10.48550/arXiv.2307.09288 [DOI] [Google Scholar]
  • 270. Dao T. FlashAttention-2: faster attention with better parallelism and work partitioning. arXiv [preprint]. 2023. doi: 10.48550/arXiv.2307.08691 [DOI] [Google Scholar]
  • 271. Dehan FN, Fahim M, Rahman AKM, Amin MA, Ali AA, eds. TinyLLM efficacy in low-resource language: an experiment on Bangla text classification task. In: International Conference on Pattern Recognition. Springer; 2025:472-487. [Google Scholar]
  • 272. Wang X, Dang T, Kostakos V, Jia H, eds. Efficient and personalized mobile health event prediction via small language models. In: Proceedings of the 30th Annual International Conference on Mobile Computing and Networking. MobiCom; 2024:2353-2358. [Google Scholar]
  • 273. National Institute for Health Care Excellence. Evidence standards framework for digital health technologies, 2018. Last updated 09/08/2022. Accessed February 16, 2025. https://www.nice.org.uk/corporate/ecd7
  • 274. Collins GS, Reitsma JB, Altman DG, Moons KGM. Transparent reporting of a multivariable prediction model for individual prognosis or diagnosis (TRIPOD) the TRIPOD statement. Circulation. 2015;131:211-219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275. Wolff RF, Moons KGM, Riley RD, et al. PROBAST: a tool to assess the risk of bias and applicability of prediction model studies. Ann Intern Med. 2019;170:51-58. [DOI] [PubMed] [Google Scholar]
  • 276. Christensen AH, Andersen CB, Wassilew K, et al. Rare non-coding desmoglein-2 variant contributes to arrhythmogenic right ventricular cardiomyopathy. J Mol Cell Cardiol. 2019;131:164-170. [DOI] [PubMed] [Google Scholar]
  • 277. Jiang X, Li T, Liu S, et al. Variants in a cis-regulatory element of TBX1 in conotruncal heart defect patients impair GATA6-mediated transactivation. Orphanet J Rare Dis. 2021;16:1-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278. Pagnamenta AT, Camps C, Giacopuzzi E, et al. Structural and non-coding variants increase the diagnostic yield of clinical whole genome sequencing for rare diseases. Genome Med. 2023;15:94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279. Bowen DJ. Haemophilia A and haemophilia B: molecular insights. Mol Pathol. 2002;55:127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280. Shetty S, Bhave M, Ghosh K. Challenges of multiple mutations in individual patients with haemophilia. Eur J Haematol. 2011;86:185-190. [DOI] [PubMed] [Google Scholar]
  • 281. Liu M, Watson LT, Zhang L. Predicting the combined effect of multiple genetic variants. Hum Genomics. 2015;9:1-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282. Holcomb D, Hamasaki-Katagiri N, Laurie K, et al. New approaches to predict the effect of co-occurring variants on protein characteristics. Am J Hum Genet. 2021;108:1502-1511. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Bioinformatics and Biology Insights are provided here courtesy of SAGE Publications

RESOURCES