Skip to main content

Barlow Twins deep neural network for advanced 1D drug–target interaction prediction

Abstract

Accurate prediction of drug–target interactions is critical for advancing drug discovery. By reducing time and cost, machine learning and deep learning can accelerate this laborious discovery process. In a novel approach, BarlowDTI, we utilise the powerful Barlow Twins architecture for feature-extraction while considering the structure of the target protein. Our method achieves state-of-the-art predictive performance against multiple established benchmarks using only one-dimensional input. The use of our hybrid approach of deep learning and gradient boosting machine as the underlying predictor ensures fast and efficient predictions without the need for substantial computational resources. We also propose the use of an influence method to investigate how the model reaches its decision based on individual training samples. By comparing co-crystal structures, we find that BarlowDTI effectively exploits catalytically active and stabilising residues, highlighting the model’s ability to generalise from one-dimensional input data. In addition, we further benchmark new baselines against existing methods. Together, these innovations improve the efficiency and effectiveness of drug–target interactions predictions, providing robust tools for accelerating drug development and deepening the understanding of molecular interactions. Therefore, we provide an easy-to-use web interface that can be freely accessed at https://www.bio.nat.tum.de/oc2/barlowdti.

Scientific contribution

Our computationally efficient and effective hybrid approach, combining the deep learning model Barlow Twins and gradient boosting machines, outperforms state-of-the-art methods across multiple splits and benchmarks using only one-dimensional input. Furthermore, we advance the field by proposing an influence method that elucidates model decision-making, thereby providing deeper insights into molecular interactions and improving the interpretability of drug-target interactions predictions.

Graphical Abstract

Introduction

Studying drug-target interactions (DTI) is crucial for understanding the biochemical mechanisms that govern how molecules interact with proteins [1]. Key challenges in drug discovery are the identification of proteins that can be used as targets for the treatment of diseases [2]. To achieve the desired therapeutic effects, the discovery of molecules that interact with and activate or inhibit target proteins is essential [3,4,5].

Recent advances in computational methods have transformed the drug discovery landscape, providing robust tools for cost-effective exploration of the chemical space. These in silico approaches facilitate the prediction and analysis of drug-target interactions, aiding in the identification of potential drug candidates and their corresponding protein targets [6,7,8,9,10,11]. The use of computational techniques allows researchers to gain a comprehensive understanding of the molecular mechanisms underlying drug-target interactions, thereby accelerating the drug discovery process and minimising reliance on traditional, resource-intensive experimental methods [12, 13]. Different methods have been used to understand how drugs interact with target proteins. These methods are grouped into three main categories: structure-agnostic, structure-based and complex-based.

Structure-agnostic approaches use one-dimensional (1D) representations like molecule simplified molecular-input line-entry system (SMILES) strings and protein amino acid sequences, graphs, or two-dimensional (2D) representations like predicted contact maps [14,15,16,17]. These methods are cost-effective and sufficiently accurate compared to experimental or in silico structure prediction [18], as they are independent of the protein’s structure when predicting effects.

Structure-based approaches require three-dimensional (3D) protein structures and 1D or 2D molecular inputs. 3D structures are usually derived from experimental data, although computational predictions are increasingly employed [19,20,21,22,23]. These methods have great potential but can be unreliable. They depend on accurate 3D protein structures and may be limited in their ability to generalise beyond experimentally observed DTIs [24]. Due to the complexity of the experimental setup, 3D protein structures can be difficult to obtain. In addition, models often overlook the fact that proteins are not rigid structures, but are generally in motion, e.g., ligand binding induces a conformational change [20, 22, 23].

Finally, complex-based approaches require protein–ligand co-crystal structures, which additionally require 3D information, as well as protein interaction information about the ligand [25]. For this reason, complex-based approaches can provide a more detailed insight into the interactions, but they are by far the most difficult to obtain data for.

Considering these different approaches, we designed BarlowDTI as a fully data-driven, sequence-based approach that relies on SMILES and amino acid sequences as the most accessible data, avoiding costly and time-consuming experimental data such as crystal structures. Additionally, we use a specialised bilingual protein language model (PLM) to embed the 1D amino acid sequence, which uses a 3D-alignment method that results in a “structure-sequence” representation [26, 27]. This approach makes BarlowDTI input data structure-agnostic, yet benefits from “structure-sequence” PLM embeddings. Unlike most other methods, we have developed a system that uses a hybrid “best of both worlds” machine learning (ML) and deep learning (DL) approach to improve drug-target interactions prediction performance in low data regimes where training data is limited [28, 29]. We have found that DL architectures such as Barlow Twins [30, 31] are excellent at learning representations [29] that can then be used for gradient boosting machine (GBM) training to achieve state-of-the-art performance, as the size of datasets is usually too small to reliably train a DL model that will perform competitively.

Fig. 1
figure 1

BarlowDTI architecture. Drug and target serve as 1D input, where they are processed and converted into vectors. Molecules are provided as SMILES and converted to ECFP. On the other hand, the primary amino acid sequence is vectorised using a bilingual 3D structure-aware PLM. The Barlow Twins architecture learns to understand drug-target interactions. The objective function forces both representations of the drug-target interactions to be as close as possible to the unity matrix. Finally, this DL model is used as a feature-extractor and a GBM is trained on the embeddings and the interaction label. The GBM is then used as the predictor

To overcome the limitation of data scarcity, we built BarlowDTIXXL, which is trained on millions of curated drug-target interactions pairs [32], to apply the model to real-world examples, as we have done in case studies. Here, BarlowDTIXXL captures the correlation between experimentally determined affinities and the predicted likelihood of interaction, proving our approach useful in drug discovery settings. By comparing co-crystal biochemical structures and their active sites, we also investigate and explain how BarlowDTIXXL arrives at its decision. We conduct our investigation by employing an influence method and adapting it in a novel way to identify the most important training DTIs [33]. We also assess whether BarlowDTIXXL can identify ligand binding sites. Additionally, we evaluate its performance in virtual screening experiments, focusing on early detection of hit molecules. This work culminates in a freely available web interface that takes 1D input of molecule and protein information and predicts the likelihood of interaction.

Results and discussion

BarlowDTI design

We propose a novel method for predicting DTIs using SMILES notations, primary amino acid sequences, both 1D, and annotated interaction properties. BarlowDTI relies on several key components, visualised in Fig. 1:

  1. 1.

    Firstly, the input needs to be vectorised. We investigate all combinations of several molecular and amino acid representations, and selected the best performing pair of modality representations (Additional file 1: Table S3). This is achieved by converting SMILES into extended-connectivity fingerprint (ECFP). Furthermore, we process amino acid sequences with a PLM that uses both modalities, combining 1D protein sequences and 3D protein structure [26].

  2. 2.

    Secondly, we teach the self-supervised learning (SSL) based Barlow Twins model interaction of molecule and protein [30, 31]. The objective function implements invariance of both representations of one interaction while ensuring non-redundancy of the features [30, 31].

  3. 3.

    Finally, BarlowDTI takes a combination of embeddings generated by the encoders from the Barlow Twins DL model and uses them as features to train a GBM based on the interaction annotations [28]. This approach exploits two key strengths: it uses DL to refine representations, and it leverages the power of machine learning in scenarios with limited data. This is particularly relevant for current drug–target interactions benchmarks/datasets, where only around 50000 annotated pairs are publicly available [34,35,36,37]. Consequently, we propose BarlowDTIXXL which is trained on more than 3600000 curated drug-target interactions pairs, additionally sourced from PubChem and ChEMBL [38, 39], to obtain generalisability in real-world scenarios [32].

Benchmark selection

We selected a comprehensive set of literature-based benchmarks to evaluate the performance of BarlowDTI against several leading methods. The benchmarks considered in this study are derived from several key sources. These sources include biomedical networks [34], the US patent database [35], and data detailing the interactions of 72 kinase inhibitors with 442 kinases, representing over 80 % of the human catalytic protein kinome [36], as well as a collection of binding affinities for the protein–ligand complexes in the Protein Data Bank [40]. These datasets provide DTIs as pairs of molecules and amino acid sequences, each coupled to an interaction annotation.

To ensure a fair comparison, BarlowDTI was retrained across all benchmarks. Finally, we assessed the model’s performance in a binary classification as well as regression setting, where the task is to distinguish between interacting and non-interacting drug–target pairs:

  • We compared BarlowDTI with a total of seven established drug-target interactions classification models: the model by Kang et al. (1D structure-agnostic) [41], MolTrans (1D substructure-based) [42], DLM-DTI (1D structure-agnostic) [17], ConPLex (1D structure-agnostic) [43], DrugBAN (2D structure-agnostic) [44], PSICHIC (2D structure-agnostic),[16] and STAMP-DPI (2D structure-based) [45]. For instance, Kang et al. fine-tuned a large language model (LLM) based on amino acid sequences [41]. MolTrans uses an efficient transformer architecture to increase the scalability of the model [42]. DLM-DTI introduced a dual language model approach combined with hint-based learning to improve prediction accuracy [17]. ConPLex leveraged contrastive learning to better understand DTIs [43], while DrugBAN focused on interpretable attention mechanisms that provide insights into the interaction process [44]. PSICHIC utilised physicochemical properties to predict interactions more accurately [16], and STAMP-DPI incorporated structure-aware, multi-modal learning to enhance its predictive capabilities [45]. Overall, we evaluated our architecture against the various model implementations. These models have demonstrated state-of-the-art performance in benchmarks.

  • This comparison is performed on a total of four classification datasets with twelve predefined literature-proposed splits: 4 \(\times\) BioSNAP [16, 34, 41], 4 \(\times\) BindingDB [16, 35, 41], 1 \(\times\) DAVIS [36, 41] and 3 \(\times\) Human [16, 42]. In addition, regression performance is evaluated on the benchmarks PDBBind v2016 and v2020.[25, 46,47,48] Our aim is to investigate the behaviour of different methods in diverse splitting scenarios, where a whole dataset is split into model training, validation, and evaluation subsets. These predefined splits help us to assess how well models generalise under challenging evaluation conditions, for example where either the drug or the target has not been seen before, thus providing insight into their real-world applicability. A detailed analysis of all benchmarks can be found in the Supporting Information “Dataset analysis”.

  • In addition, we investigated the addition of a more rigorous model baseline. The GBM XGBoost is known to be one of the best models, e.g. in quantitative structure-activity relationship (QSAR) tasks, often outperforming DL-based approaches [49,50,51].

BarlowDTI shows state-of-the-art performance in predicting DTIs

We assessed the performance of BarlowDTI in binary classification across four distinct datasets, each employing different data splitting procedures. For each dataset, we predicted whether drug–target pairs in the predefined test subset interact or not. We then statistically evaluated these predictions by comparing them to the actual outcomes provided in the benchmark test set, using the metrics receiver operating characteristic area under curve (ROC AUC) and precision recall area under curve (PR AUC). Overall, BarlowDTI significantly outperforms all other models in Fig. 2a and Tables 1 and Additional file 1: Table S5. Looking at BioSNAP, we improve 6 % over the leading method DLM-DTI in terms of PR AUC. Furthermore, as shown in Table 2 BarlowDTI again outperforms the PSICHIC method with a 7 % PR AUC improvement independent of the split.

Table 1 Benchmarking BarlowDTI against other models using Kang et al. splits [41]

When switching to BindingDB, BarlowDTI significantly outperforms DLM-DTI in terms of PR AUC with a >14 % improvement (Table 1). Investigating the BindingDB splits shows that BarlowDTI outperforms all existing methods when looking at unseen ligands, matches the ROC AUC performance of DrugBAN in the random setting and becomes second best in the unseen protein split (Table 2). Overall, BarlowDTI performs best in two out of four splits in this benchmark.

Table 2 Benchmarking BarlowDTI against other models using Koh et al. splits [16]

BarlowDTI once again outperforms all of the established approaches when looking at the DAVIS benchmark, with a 21 % improvement over the leading ConPLex model in terms of PR AUC (Table 1).

Furthermore, we evaluated the performance on the Human benchmark. BarlowDTI shows the best performance when looking at the unseen protein split as well as the random split (Table 2). PSICHIC comes first in the unseen ligand setting, when looking at ROC AUC, while DrugBAN is best in PR AUC. In summary, BarlowDTI outperforms all other models in two out of three splits.

Lastly, BarlowDTI and XGBoost regression performance was determined on two regression benchmarks: PDBBind v2016 and v2020 (Additional file 1: Tables S4 and S8) [25, 46,47,48]. Overall, BarlowDTI demonstrates competitive regression performance, ranking third on PDBBind v2016 and second on PDBBind v2020 among twelve literature-known models (Supporting Information “Regression performance”). XGBoost places third and fourth, still putting our proposed baseline ahead of two-thirds of all methods.

We looked at the architecture and its components, removing one at a time and measuring the effect on performance to investigate why BarlowDTI outperforms other methods in various benchmarks.

Fig. 2
figure 2

A comparison of the performance of methods established in the literature. a The state-of-the-art performance of BarlowDTI in terms of PR AUC was visualised in comparison to other models (for metrics and their statistics refer to Table 1). b The change in performance was examined as key elements of the BarlowDTI architecture were incrementally removed. Ablations are denoted as Abl. 1, 2 and 3. c The newly introduced model baseline, XGBoost, was compared with other established methods. A per dataset and split difference in PR AUC was calculated based on BarlowDTI in (b) performance or the baseline model in (c). The overall change was investigated for statistical significance (****\(p < 0.0001\), two-sided Welch’s t-test [52, 53], with Benjamini-Hochberg [54] multiple testing correction)

Unravelling the performance contributions of the BarlowDTI architecture

To investigate the impact of each element of the BarlowDTI architecture, we removed them one at a time. We have done this across all baselines and splits with the following ablations:

  1. 1.

    We removed the hyperparameter optimisation step of the BarlowDTI classifier (Fig. 2b Abl. 1).

  2. 2.

    From the first removal, we replaced the Barlow Twins architecture entirely and instead concatenate ECFP and PLM embeddings for training (Fig. 2b Abl. 2). We kept the hyperparameter optimisation procedure as in BarlowDTI.

  3. 3.

    Finally, we removed the hyperparameter optimisation procedure from the previous ablation, analogous to the first modification (Fig. 2b Abl. 3).

We observe a significant decline in performance, as illustrated in Fig. 2b and Additional file 1: Table S6 for the initial ablation, emphasising the crucial role of hyperparameter optimisation for achieving optimal model performance.

The second ablation also indicates a significant reduction in performance. However, for the DAVIS benchmark, the optimised GBM demonstrated surprisingly strong performance. We hypothesise that this may result from the hyperparameter search potentially leading to overfitting on the test set. This would be consistent with the improvement from the third ablation experiment to the first experiment comparing the two non-optimised model variants. On the whole, model performance is likely attributed to the DL architecture based on the SSL Barlow Twins model, which effectively learns embeddings to describe DTIs. The Barlow Twins objective promotes orthogonality between drug and target modalities while ensuring the non-redundancy of both, thus preventing informational collapse. As a result, this leads to an overall state-of-the-art predictive performance.

The final ablation shows a further decline in performance, consistent with the results of the initial ablation experiment.

In summary, the sustained reduction in performance of our ablation experiments demonstrates that each component of our BarlowDTI pipeline is needed to maximise performance. This architecture integrates the “best of both worlds”: DL and GBM to enhance predictive performance. Compared to other pure machine learning- or DL-based approaches, we can demonstrate a performance boost. In particular, the use of a state-of-the-art PLM [26] could offer an advantage over other methods. Other PLM variants are ProtTrans [55] in ConPLex [43] and ProtBERT proposed by Kang et al. also used in DLM-DTI [41]. The structural awareness of BarlowDTI added by the inclusion of 3D-alignment in ProstT5 [26] hints towards better generalisation capabilities, yielding increased performance.

Choosing baseline models

Selecting an appropriate baseline model is critical to effectively comparing different machine learning and DL techniques. Robust baselines are the basis for meaningful comparisons and highlight improvements from new methods. Without appropriate baselines, it becomes difficult to determine whether new approaches are truly advancing the field.

Current leading drug-target interactions models predominantly use DL methods and are often evaluated against simple baseline models such as logistic regression, ridge or deep neural network (DNN) classifiers [42, 43]. To improve the benchmarking process, we propose to add GBMs as a baseline for drug-target interactions benchmarking purposes, as shown in the final ablation configuration. GBMs such as XGBoost have demonstrated broad adaptability, e.g. in QSAR modelling, offering strong predictive performance and fast training times, particularly in scenarios with limited data availability, such as drug-target interactions prediction.

We compared the overall model performance across all datasets in Fig. 2c and Tables 1, 2, and Additional file 1: Tables S4, S7, and S8. Here, the performance of XGBoost trained on ECFP and PLM embeddings is highlighted as it shows competitive performance across all methods and datasets.

Demonstration of the capabilities of BarlowDTIXXL

To use BarlowDTI in real-world applications, more training data is needed to predict meaningful interactions. For this purpose, we have built BarlowDTIXXL, which is trained on more than 3600000 curated drug-target interactions pairs [32]. We have kept the same model design to ensure the comparability and performance of our hybrid approach. We looked at several co-crystal structures as case studies to provide insight into the possibilities using BarlowDTIXXL. In order to demonstrate the ability to generalise beyond the learnt DTIs, we evaluated our approach on structures which are not part of the training set. Our aim is to demonstrate the applicability of the model to multiple structures and affinities, as in the study performed by Dienemann et al. The importance of this work is further emphasised by its relevance to the malaria-causing parasite Plasmodium falciparum [56].

We first analysed the co-crystal structures Plasmodium falciparum lipoate protein ligase 1 LipL1 (5T8U) and Listeria monocytogenes lplA1 (8CRI), which share a low sequence identity (28.7 %) despite their structural similarity. Our objective is to evaluate the model’s ability to generalise, particularly when only 1D input is provided. This evaluation focuses on the model’s performance in capturing both biological function and structural attributes under these conditions. Secondly, we examined the predictive shifts induced by ligand methylation and explored the interaction dynamics of a novel enzyme inhibitor C3 (8CRL). This case study is further enriched with isothermal titration calorimetry (ITC) data [56], offering insights into the ligand’s affinity towards the target proteins.

Our results indicate, that BarlowDTIXXL is able to accurately predict the correlation between the experimentally determined affinity measured via ITC and the likelihood of the DTI (Fig. 3b). These capabilities provide useful insight in the drug discovery process, as researchers are able to prioritise chemical scaffolds. BarlowDTIXXL is able to catch small changes in the ligands structure and accurately predict the shift in interaction likelihood. This is illustrated by the methylation of LA, where our method predicts a significant decrease in interaction likelihood, consistent with the decrease in affinity measured by ITC.

To further validate the performance of BarlowDTIXXL, we conducted a virtual screening experiment (Additional file 1: Fig. S2) focused on identifying kinase inhibitors. Kinase inhibitors are pivotal in drug discovery, targeting dysregulated protein kinases linked to cancer, autoimmune disorders, and inflammation [57,58,59]. Our results show that BarlowDTIXXL effectively prioritised kinase inhibitors while remaining computationally efficient, reducing costs and time, and thereby accelerating the drug discovery process (Supporting Information “Virtual screening”).

We looked at Shapley additive explanation (SHAP) values to examine the influence of each input modality on the model (Additional file 1: Fig. S6). Regardless of the ligand molecule chosen, each modality proved equally important for prediction. This finding highlights the functionality and predictive power of BarlowDTI’s architecture.

Explaining BarlowDTI by investigating sample importance

We analysed the importance of individual samples within the training set to understand how BarlowDTI classifies DTIs. Our adjusted influence method was therefore applied. In Fig. 3d,e, we identified the most influential training pairs by examining those with the highest Jaccard similarity, calculated from the leaf indices of the GBM in BarlowDTIXXL. The most influential training sample is the Homo sapiens lipoyl amidotransferase LIPT1 for both lplA1 and LipL1, with LA as the common ligand (Fig. 3a,e). LIPT1 and lplA1 (\(J = 0.909\)) share a sequence identity of 31.8 %, while LIPT1 and LipL1 (\(J = 0.913\)) only share 29.7 % (Fig. S7).

Fig. 3
figure 3

Structure-based explanation of BarlowDTIXXL predictions. a Co-crystal structures of lplA1 and LipL1 with LA as ligand are shown in superposition, together with the most influential training sample (structure predicted using RoseTTAFold-AllAtom (RF-AA) [21]). b The squared Pearson R [60] correlation of BarlowDTIXXL and ITC measurements is presented [56]. c The protein residue–ligand interactions at the active site are compared. d We identified the most influential training samples for LA predictions. The distribution of Jaccard similarity for all training samples is shown. We applied kernel density estimation to the histogram to improve visibility, due to the large training set size. e The most influential training samples are highlighted (\(\downarrow\))

To investigate the biochemical implications of the training sample to the model’s prediction, we performed a structural study. We leveraged protein crystal structures to perform in-depth 3D analyses on lplA1 (8CRI) and LipL1 (5T8U). The superposition of lplA1 with LIPT1 revealed a root mean square deviation of atomic positions (RMSD) of 2.07 Å, while LipL1 exhibited a RMSD of 1.72 Å. These RMSD values reflect a significant structural congruence among these enzymes, notwithstanding their low sequence identity. Despite this structural similarity, it is noteworthy that human LIPT1 does not catalyse the same reaction as lplA1 and LipL1 [61].

Furthermore, we looked at the active site of LipL1, where all residues are conserved relative to LIPT1 (Fig. 3c). In lplA1, one notable substitution can be observed. L181 in LIPT1 is replaced by M151, possibly explaining the higher Jaccard similarity of LipL1 over lplA1. This conservation pattern underscores a highly conserved binding pocket across species, as confirmed by sequence alignment data (Additional file 1: Fig. S7). Furthermore, we investigated whether BarlowDTIXXL demonstrates sequence-based awareness of ligand interaction sites. The strongest shifts in predicted drug–target interaction likelihood is observed when active site residues are substituted (Additional file 1: Figs. S3 and S4, Supporting Information “Ligand interaction”). These results highlight the awareness of BarlowDTIXXL to ligand-binding residues and help to understand how the prediction of the model is achieved.

In summary, BarlowDTIXXL effectively learns DTIs by leveraging catalytically active and stabilising residues, demonstrating the model’s ability to generalise from 1D input data. This capability makes BarlowDTIXXL well-suited for applications in drug discovery.

Conclusions

Our proposed method, BarlowDTI, integrates sequence information with the Barlow Twins SSL architecture and GBM models, representing a powerful fusion of machine learning and DL techniques.

Our approach demonstrates state-of-the-art drug-target interactions prediction capabilities, validated across multiple benchmarks and data splits. Notably, our method outperforms existing literature benchmarks in ten out of fourteen datasets evaluated.

To elucidate the efficacy of BarlowDTI, we conducted an ablation study to investigate the contribution of its core components and their impact on performance. In addition, we re-evaluated the choice of baselines in numerous publications and advocate the inclusion of GBM baselines. Furthermore, we explored the classification mechanism of BarlowDTI for DTIs by performing a structure-based analysis of the most influential training samples. This was done by adapting a previously developed influence method to gain deeper insight into training sample importance.

Given the model’s exceptional performance, we are confident that BarlowDTI can significantly accelerate the drug discovery process and offer significant time and cost savings through the use of virtual screening campaigns. To make BarlowDTI accessible to the scientific community, we provide an easy-to-use and free web interface at https://www.bio.nat.tum.de/oc2/barlowdti.

Methods

Datasets

To evaluate the performance of BarlowDTI, three established benchmarks are used. They all provide fixed splits for training, evaluation and testing. In some publications the training and evaluation is merged to improve predictive performance. To ensure comparability, this was not done in this work. All the metrics presented are taken from other publications in which only the training set is used.

In addition, Kang et al. first proposed splits for large drug-target interactions datasets, BioSNAP [34], BindingDB [35] and DAVIS [36, 41].

The addition of a variety of splits with an additional benchmark Human [42] are proposed by Koh et al. We evaluate these separately [16]. Regression performance is evaluated on two regression benchmarks: PDBBind v2016 and v2020 [25, 46,47,48]. For all datasets, to reduce bias and improve model performance, the SMILES are cleaned using the Python ChEMBL curation pipeline [62]. All duplicate and erroneous molecule and protein information that could not be parsed is removed. Training is performed on the predefined training splits.

Representations

Molecular information

The SMILES are converted into Atom Pair [63], ECFP [64], Electrotopological State (EState) [65], MACCS [66], MinHashed Atom Pair (MAP) [67], PubChem and RDKit fingerprints [68] using scikit-fingerprints and RDKit [68, 69]. We used 1024bit and a radius of 2 where possible, otherwise the default parameters were used.

Amino acid sequence information

The amino acid sequences are converted into vectors, by using the PLM ProtTrans [55], ProtT5 [55] and ProstT5 [26]

Additionally, the protein sequences were encoded using one-hot encoding, In this method, each amino acid is represented by a unique binary vector where one position is set to 1, and all others are set to 0. For this encoding, we used the standard set of 20 amino acids \(\mathcal {A}\): Each amino acid \(a \in \mathcal {A}\) is mapped to a unique index: \(\text {index}(a) = i, \quad \text {where} \, i \in \{0, 1, \dots , 19\}.\) For a given amino acid sequence \(S = (s_1, s_2, \dots , s_n)\), where each \(s_j \in \mathcal {A}\), we encode each amino acid \(s_j\) as a one-hot vector \(\textbf{v}_j \in \mathbb {R}^{20}\), defined as:

$$\textbf{v}_j[i] = {\left\{ \begin{array}{ll} 1 & \text {if } i = \text {index}(s_j),\\ 0 & \text {otherwise}. \end{array}\right. }$$

For sequences shorter than a predefined maximum length \(l_{\max }\), padding is applied using a placeholder amino acid \(\text {X}\), which is mapped to a zero vector: \(\textbf{v}_{\text {X}} = (0, 0, \dots , 0) \in \mathbb {R}^{20}.\) Given a sequence of length \(n\), we ensure the final encoded vector has length \(20 \times l_{\max }\) by either truncating or padding the sequence. Thus, for each sequence \(S\) of length \(n\), the one-hot encoded representation \(\textbf{V}(S)\) is given by:

$$\textbf{V}(S) = [ \textbf{v}_1, \textbf{v}_2, \dots , \textbf{v}_{l_{\max }} ] \in \mathbb {R}^{20 \times l_{\max }}.$$

Barlow Twins model configuration

The proposed method is based on the Barlow Twins [30] network architecture, which employs one encoder for each modality and a unified projector. The encoders and projector are multilayer perceptron (MLP) based.

Both encoders as well as the projector have the following structure

$$\begin{aligned} \varvec{l_{i+1}} = \textrm{Linear} \left( {\textrm{ReLU} \left( \textrm{BatchNorm} \left( \textrm{Linear} \left( \varvec{W l_i + b} \right) \right) \right) }^{n} \right) , \end{aligned}$$

where \(\varvec{l_{i}}\) is the input layer and \(\varvec{l_{i+1}}\) is its output, with a flexible number of layers n and adjustable dimensionality of input and output. Furthermore, variables \(\varvec{W}\), \(\varvec{b}\) represent learnable weights and biases. A linear layer is followed by batch normalisation [70], ReLU activation function [71], and the last linear layer. The network was constructed using PyTorch [72].

The loss function \(\mathcal {L_{BT}}\) is adapted from the original Barlow Twins publication and enforces cross-correlation (matrix \(\mathcal {C}\)) between the projections of the modalities [30].

$$\begin{aligned} \mathcal {L_{BT}} \triangleq \underbrace{\sum _i (1-\mathcal {C}_{ii})^2}_\text {invariance term} + ~~\lambda \underbrace{\sum _{i}\sum _{j \ne i} {\mathcal {C}_{ij}}^2}_\text {redundancy reduction term} \end{aligned}$$

where \(\lambda\) is a constant that trades off the invariance term and redundancy reduction term.

$$\begin{aligned} \mathcal {C}_{ij} \triangleq \frac{ \sum _b z^A_{b,i} z^B_{b,j}}{\sqrt{\sum _b {(z^A_{b,i})}^2} \sqrt{\sum _b {(z^B_{b,j})}^2}} \end{aligned}$$

Pre-training Barlow Twins

Here we pre-train the Barlow Twins architecture on our joint drug-target interactions dataset, based on BioSNAP, BindingDB, DAVIS (Kang et al. splits) and DrugBank [37], removing duplicates and without labels to teach DTIs. Early stopping is implemented to avoid overfitting, which is carried out using a 15 % validation split.

Hyperparameter optimisation

Manual hyperparameter optimisation is performed, shown in Table 3.

Table 3 Barlow Twins hyperparameters

Feature-extractor

When performing feature-extraction, we use the pre-trained BarlowDTI model. For training and prediction, we extract the embeddings after the encoders for each modality and concatenate them. Finally, a GBM, XGBoost [28] Python implementation, is trained on the embeddings in combination with the labels for each training sets respectively.

Hyperparameter optimisation

If a benchmark provides a dedicated validation set, this was used for Optuna [73] hyperparameter optimisation. Therefore, in classification \(\mathcal {L}_{c} = \text{ROC AUC} + \text{PR AUC}\) was used as validation loss and Optuna was configured to maximise the summed loss. For regression \(\mathcal {L}_{r} = - \rho + \text{MAE}\) was applied as validation loss and Optuna was configured to minimise the summed loss.

The optimisation was carried out for 100 trials with the parameters shown in Table 4. The obtained benchmark specific hyperparameters were then used to fit the GBM on the training set. All detailed hyperparameters are provided in the Additional File gbm_hyperparameters.csv.

Table 4 GBM hyperparameters
BarlowDTI XXL

We introduce BarlowDTIXXL, a model trained for use in real-world applications. To build BarlowDTIXXL, we curated and standardised the large drug-target interactions dataset proposed by Golts et al. (procedure adapted from the “Datasets” section) [32]. Furthermore, we used random undersampling with a 3:1 ratio of non-interactors to interactors to improve model generalisation. Then we added the training splits from BioSNAP, BindingDB and DAVIS (Kang et al. splits), resulting in a model trained with 3653631 drug-target interactions pairs (2 789 498 non-interactors, 864 133 interactors).

BarlowDTIXXL uses the same architecture as BarlowDTI, using the powerful Barlow Twins network as feature-extraction method in combination with the GBM XGBoost [28, 30].

Baseline model configuration

As a baseline, we have selected a GBM. Similar to our feature-extraction implementation, for all features we concatenate both ECFP and PLM embeddings. Finally, a GBM, XGBoost Python implementation, is trained on the ECFP and PLM embedding concatenation in combination with the labels for each training set, respectively.

Case study

Amino acid sequence information as well as ligand information is taken from The Protein Data Bank to perform predictions using BarlowDTI [74]. Complex structures were generated using RoseTTAFold-AllAtom (RF-AA) [21].

Sequence identity was determined. Therefore, sequences were aligned using the BLASTP [75, 76] algorithm at https://blast.ncbi.nlm.nih.gov [77]. PyMOL 2 is used for structure visualisation and RMSD value calculation [78].

Explainability based on Shapley additive explanation values

We applied the TreeExplainer [79, 80] algorithm to the GBM of BarlowDTIXXL extracted and visualised the SHAP values.

Explainability based on sample importance

To assess how the model decides to classify drug–target pairs as interacting or non-interacting, we looked at the influence of training samples, as similarly proposed by Brophy et al. for uncertainty estimation [33]. We used a similar concept but changed the approach to identify the most influential training data. This is done by obtaining the leaf indices of the GBM of all training samples. Then we compare the leaf indices at inference time with the leaf indices of the training samples. Finally, we find the most influential samples by computing the pairwise Jaccard similarity of the leaf index vectors [81],

$$\begin{aligned} J(A, B)=\frac{|A \cap B|}{|A \cup B|}. \end{aligned}$$

The most influential training sample is represented by the maximum Jaccard similarity.

Availability of data and materials

The easy-to-use web interface can be found at https://www.bio.nat.tum.de/oc2/barlowdti. The code is available on GitHub https://github.com/maxischuh/BarlowDTI. Also available on GitHub are the curated and extensive BarlowDTIXXL training, as well as the benchmark data. The system used for computational work is equipped with an AMD Ryzen Threadripper PRO 5995WX CPU with 64/128 cores/threads and 1024GB RAM. The server is also powered by an NVIDIA RTX 4090 GPU with 24GB VRAM.

Abbreviations

1D:

One-dimensional

2D:

Two-dimensional

3D:

Three-dimensional

BEDROC:

Boltzmann-enhanced discrimination of receiver operating characteristic

CI:

Concordance index

DL:

Deep learning

DNN:

Deep neural network

DTI:

Drug-target interaction

ECFP:

Extended-connectivity fingerprint

GBM:

Gradient boosting machine

ITC:

Isothermal titration calorimetry

LA:

Lipoic acid

LLM:

Large language model

MAE:

Mean absolute error

ML:

Machine learning

MLP:

Multilayer perceptron

PLM:

Protein language model

PR AUC:

Precision recall area under curve

QSAR:

Quantitative structure-activity relationship

RF-AA:

RoseTTAFold-AllAtom

RMSD:

Root mean square deviation of atomic positions

ROC AUC:

Receiver operating characteristic area under curve

SHAP:

Shapley additive explanation

SMILES:

Simplified molecular-input line-entry system

SSL:

Self-supervised learning

References

  1. Rang HP, Dale MM, Ritter JM, Flower RJ, Henderson G (2011) Rang & Dale’s pharmacology. Elsevier Health Sciences, Amsterdam

    Google Scholar 

  2. Strittmatter SM (2014) Overcoming drug development bottlenecks with repurposing: old drugs learn new tricks. Nat Med 20:590–591

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hughes J, Rees S, Kalindjian S, Philpott K (2011) Principles of early drug discovery. Br J Pharmacol 162:1239–1249

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Blundell TL et al (2006) Structural biology and bioinformatics in drug design: opportunities and challenges for target identification and lead discovery. Philos Trans R Soc B Biol Sci 361:413–423

    Article  CAS  Google Scholar 

  5. Tautermann CS (2020) Current and future challenges in modern drug discovery. In: Heifetz A (ed) Quantum mechanics in drug discovery. Springer US, New York, pp 1–17

    Google Scholar 

  6. Agu PC et al (2023) Molecular docking as a tool for the discovery of molecular targets of nutraceuticals in diseases management. Sci Rep 13:13398

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Bender BJ et al (2021) A practical guide to large-scale docking. Nat Protoc 16:4799–4832

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hollingsworth SA, Dror RO (2018) Molecular dynamics simulation for all. Neuron 99:1129–1143

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Karplus M, Petsko GA (1990) Molecular dynamics simulations in biology. Nature 347:631–639

    Article  CAS  PubMed  Google Scholar 

  10. Dhakal A, McKay C, Tanner JJ, Cheng J (2022) Artificial intelligence in the prediction of protein-ligand interactions: recent advances and future directions. Brief Bioinform 23:bbab476

    Article  PubMed  Google Scholar 

  11. You Y et al (2022) Artificial intelligence in cancer target identification and drug discovery. Signal Transduct Target Ther 7:1–24

    Article  CAS  Google Scholar 

  12. Kitchen DB, Decornez H, Furr JR, Bajorath J (2004) Docking and scoring in virtual screening for drug discovery: methods and applications. Nat Rev Drug Discov 3:935–949

    Article  CAS  PubMed  Google Scholar 

  13. Hopkins AL (2009) Predicting promiscuity. Nature 462:167–168

    Article  CAS  PubMed  Google Scholar 

  14. Chen L et al (2020) TransformerCPI: Improving compound-protein interaction prediction by sequence-based deep learning with self-attention mechanism and label reversal experiments. Bioinformatics 36:4406–4414

    Article  CAS  PubMed  Google Scholar 

  15. Jiang M et al (2020) Drug-target affinity prediction using graph neural network and contact maps. RSC Adv 10:20701–20712

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Koh HY, Nguyen ATN, Pan S, May LT, Webb GI (2024) Physicochemical graph neural network for learning protein-ligand interaction fingerprints from sequence data. Nat Mach Intell 6:673–687

    Article  Google Scholar 

  17. Lee J, Jun DW, Song I, Kim Y (2024) DLM-DTI: a dual language model for the prediction of drug-target interaction with hint-based learning. J Cheminform 16:1–12

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Jiang M et al (2022) Sequence-based drug-target affinity prediction using weighted graph neural networks. BMC Genom 23:449

    Article  CAS  Google Scholar 

  19. Ahdritz G et al (2024) OpenFold: retraining AlphaFold2 yields new insights into its learning mechanisms and capacity for generalization. Nat Methods. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41592-024-02272-z

    Article  PubMed  PubMed Central  Google Scholar 

  20. Abramson J et al (2024) Accurate structure prediction of biomolecular interactions with AlphaFold 3. Nature 630:493–500

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Krishna R et al (2024) Generalized biomolecular modeling and design with RoseTTAFold All-Atom. Science 384:eadl2528

    Article  CAS  PubMed  Google Scholar 

  22. Trott O, Olson AJ (2010) AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem 31:455–461

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Corso G, Stärk H, Jing B, Barzilay R, Jaakkola T. DiffDock: diffusion steps, twists, and turns for molecular docking. 2210.01776. 2023.

  24. He X-H et al (2023) AlphaFold2 versus experimental structures: evaluation on G protein-coupled receptors. Acta Pharmacol Sin 44:1–7

    Article  CAS  PubMed  Google Scholar 

  25. Li S, et al. Structure-aware interactive graph neural networks for the prediction of protein-ligand binding affinity. 2021.

  26. Heinzinger M, et al. Bilingual language model for protein sequence and structure. 2024.

  27. van Kempen M et al (2024) Fast and accurate protein structure search with Foldseek. Nat Biotechnol 42:243–246

    Article  PubMed  Google Scholar 

  28. Chen T, Guestrin C. XGBoost: a scalable tree boosting system. 1603.02754. 2016.

  29. Schuh MG, Boldini D, Sieber SA (2024) Synergizing chemical structures and bioassay descriptions for enhanced molecular property prediction in drug discovery. J Chem Inform Model 64:4640–4650

    Article  CAS  Google Scholar 

  30. Zbontar J, Jing L, Misra I, LeCun Y, Deny S. Barlow twins: self-supervised learning via redundancy reduction. 2103:03230. 2021.

  31. Barlow HB et al (1961) Possible principles underlying the transformation of sensory messages. Sens Commun 1:217–233

    Google Scholar 

  32. Golts A, et al. A large dataset curation and benchmark for drug target interaction. 2401:17174. 2024

  33. Brophy J, Lowd D. Instance-based uncertainty estimation for gradient-boosted regression trees. 2205:11412. 2022.

  34. Zitnik M, Sosič R, Maheshwari S & Leskovec J. BioSNAP datasets: stanford biomedical network dataset collection. 2018. http://snap.stanford.edu/biodata.

  35. Liu T, Lin Y, Wen X, Jorissen RN, Gilson MK (2007) BindingDB: a web-accessible database of experimentally determined protein-ligand binding affinities. Nucleic Acids Res 35:D198–D201

    Article  CAS  PubMed  Google Scholar 

  36. Davis MI et al (2011) Comprehensive analysis of kinase inhibitor selectivity. Nat Biotechnol 29:1046–1051

    Article  CAS  PubMed  Google Scholar 

  37. Knox C et al (2024) DrugBank 6.0: the DrugBank knowledgebase for 2024. Nucleic Acids Res 52:D1265–D1275

    Article  CAS  PubMed  Google Scholar 

  38. Kim S et al (2023) PubChem 2023 update. Nucleic Acids Res 51:D1373–D1380

    Article  PubMed  Google Scholar 

  39. Mendez D et al (2019) ChEMBL: towards direct deposition of bioassay data. Nucleic Acids Res 47:D930–D940

    Article  CAS  PubMed  Google Scholar 

  40. Wang R, Fang X, Lu Y, Yang C-Y, Wang S (2005) The PDBbind database: methodologies and updates. J Med Chem 48:4111–4119

    Article  CAS  PubMed  Google Scholar 

  41. Kang H et al (2022) Fine-tuning of BERT model to accurately predict drug-target interactions. Pharmaceutics 14:1710

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Huang K, Xiao C, Glass LM, Sun J (2021) MolTrans: molecular interaction transformer for drug-target interaction prediction. Bioinformatics 37:830–836

    Article  CAS  PubMed  Google Scholar 

  43. Singh R, Sledzieski S, Bryson B, Cowen L, Berger B (2023) Contrastive learning in protein language space predicts interactions between drugs and protein targets. Proc Natl Acad Sci 120:e2220778120

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Bai P, Miljković F, John B, Lu H (2023) Interpretable bilinear attention network with domain adaptation improves drug-target prediction. Nat Mach Intell 5:126–136

    Article  Google Scholar 

  45. Wang P et al (2022) Structure-aware multimodal deep learning for drug-protein interaction prediction. J Chem Inform Model 62:1308–1317

    Article  CAS  Google Scholar 

  46. Su M et al (2019) Comparative assessment of scoring functions: the CASF-2016 update. J Chem Inform Model 59:895–913

    Article  CAS  Google Scholar 

  47. Lu W, et al. TANKBind: trigonometry-aware neural networks for drug-protein binding structure prediction. 2022.

  48. Stärk H Ganea O Pattanaik L Barzilay DR, Jaakkola T. Geometric deep learning for drug binding structure prediction, EquiBind. 2022.

  49. Wu Z et al (2021) Do we need different machine learning algorithms for QSAR modeling? A comprehensive assessment of 16 machine learning algorithms on 14 QSAR data sets. Brief Bioinform 22:3, bbaa321

    Article  PubMed  Google Scholar 

  50. Sheridan RP, Wang WM, Liaw A, Ma J, Gifford EM (2016) Extreme gradient boosting as a method for quantitative structure-activity relationships. J Chem Inform Model 56:2353–2360

    Article  CAS  Google Scholar 

  51. Asselman A, Khaldi M, Aammou S (2023) Enhancing the prediction of student performance based on the machine learning XGBoost algorithm. Interact Learn Environ 31:3360–3379

    Article  Google Scholar 

  52. Welch BL (1947) The generalization of ‘student’s’ problem when several different population varlances are involved. Biometrika 34:28–35

    CAS  PubMed  Google Scholar 

  53. Virtanen P et al (2020) SciPy 1.0: fundamental algorithms for scientific computing in python. Nat Methods 17:261–272

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Benjamini Y, Hochberg Y (1995) Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B Methodol 57:289–300

    Article  Google Scholar 

  55. Elnaggar A et al (2022) ProtTrans: toward understanding the language of life through self-supervised learning. IEEE Trans Pattern Anal Mach Intell 44:7112–7127

    Article  PubMed  Google Scholar 

  56. Dienemann J-N et al (2023) A chemical proteomic strategy reveals inhibitors of lipoate salvage in bacteria and parasites. Angew Chem Int Ed 62:e202304533

    Article  Google Scholar 

  57. Cohen P, Cross D, Jänne PA (2021) Kinase drug discovery 20 years after imatinib: progress and future directions. Nat Rev Drug Discov 20:551–569

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Attwood MM, Fabbro D, Sokolov AV, Knapp S, Schiöth HB (2021) Trends in kinase drug discovery: targets, indications and inhibitor design. Nat Rev Drug Discov 20:839–861

    Article  CAS  PubMed  Google Scholar 

  59. Zhavoronkov A et al (2019) Deep learning enables rapid identification of potent DDR1 kinase inhibitors. Nat Biotechnol 37:1038–1040

    Article  CAS  PubMed  Google Scholar 

  60. Pearson K (1895) Note on regression and inheritance in the case of two parents. Proc R Soc Lond Ser I(58):240–242

    Google Scholar 

  61. Cao X, Zhu L, Song X, Hu Z, Cronan JE (2018) Protein moonlighting elucidates the essential human pathway catalyzing lipoic acid assembly on its cognate enzymes. Proc Natl Acad Sci 115:E7063–E7072

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Bento AP et al (2020) An open source chemical structure curation pipeline using RDKit. J Cheminform 12:51

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Carhart RE, Smith DH, Venkataraghavan R (1985) Atom pairs as molecular features in structure-activity studies: definition and applications. J Chem Inform Computer Sci 25:64–73

    Article  CAS  Google Scholar 

  64. Rogers D, Hahn M (2010) Extended-connectivity fingerprints. J Chem Inform Model 50:742–754

    Article  CAS  Google Scholar 

  65. Hall LH, Kier LB (1995) Electrotopological state indices for atom types: a novel combination of electronic, topological, and valence state information. J Chem Inform Computer Sci 35:1039–1045

    Article  CAS  Google Scholar 

  66. Durant JL, Leland BA, Henry DR, Nourse JG (2002) Reoptimization of mdl keys for use in drug discovery. J Chem Inform Computer Sci 42:1273–1280

    Article  CAS  Google Scholar 

  67. Capecchi A, Probst D, Reymond J-L (2020) One molecular fingerprint to rule them all: drugs, biomolecules, and the metabolome. J Cheminform 12:43

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Landrum G, et al. Rdkit/rdkit: 2020_03_1 (Q1 2020) release. Zenodo. 2020.

  69. Adamczyk J, Ludynia P. Scikit-fingerprints: easy and efficient computation of molecular fingerprints in Python. 2407.13291. 2024.

  70. Ioffe S, Szegedy C. Batch normalization: accelerating deep network training by reducing internal covariate shift. 1502:03167. 2015.

  71. Agarap AF. Deep Learning using rectified linear units (ReLU). 1803:08375. 2019.

  72. Paszke A, et al. PyTorch: an imperative style, high-performance deep learning library. 1912.01703. 2019.

  73. Akiba T, Sano S, Yanase T, Ohta T, Koyama M. A next-generation hyperparameter optimization framework, Optuna. 2019.

  74. Berman HM et al (2000) The protein data bank. Nucleic Acids Res 28:235–242

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ (1990) Basic local alignment search tool. J Mol Biol 215:403–410

    Article  CAS  PubMed  Google Scholar 

  76. Altschul SF et al (1997) Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res 25:3389–3402

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Sayers EW et al (2022) Database resources of the national center for biotechnology information. Nucleic Acids Res 50:D20–D26

    Article  CAS  PubMed  Google Scholar 

  78. Schrödinger LLC. The PyMOL molecular graphics system, version 1.8. 2015.

  79. Lundberg SM, Lee S-I. A unified approach to interpreting model predictions. 2017.

  80. Lundberg SM et al (2020) From local explanations to global understanding with explainable AI for trees. Nat Mach Intell 2:56–67

    Article  PubMed  PubMed Central  Google Scholar 

  81. Jaccard P (1901) Étude comparative de la distribution florale dans une portion des Alpes et du Jura. Bull Soc Vaud Sci Nat 37:547

    Google Scholar 

Download references

Acknowledgements

All authors thank Prof. Michael Groll for his insight into the crystal structure data. M.G.S. thanks Joshua Hesse and Aleksandra Daniluk for their valuable input and helpful feedback and Leonard Gareis for assistance with the website.

Funding

Open Access funding enabled and organized by Projekt DEAL. Open Access funding enabled and organized by Projekt DEAL. The authors thank Merck KGaA Darmstadt for their generous support with the Merck Future Insight Prize 2020. This project is also cofunded by the European Union (ERC, breakingBAC, 101096911).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualisation: M.G.S. and D.B.; Benchmarking: M.G.S.; Software: M.G.S.; Methodology: M.G.S., D.B. and A.I.B.; Analysis of the results: all authors.; Writing original draft: M.G.S. and A.I.B.; Writing review and editing: all authors.; All authors have given approval to the final version of the manuscript.

Corresponding authors

Correspondence to Davide Boldini or Stephan A. Sieber.

Ethics declarations

Competing interests

The authors declare no Conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schuh, M.G., Boldini, D., Bohne, A.I. et al. Barlow Twins deep neural network for advanced 1D drug–target interaction prediction. J Cheminform 17, 18 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13321-025-00952-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13321-025-00952-2

Keywords