Next Article in Journal
ChronoSort: Revealing Hidden Dynamics in AlphaFold3 Structure Predictions
Previous Article in Journal
Stoichiometric Multiprotein Assembly Scaffolded by a Heterotrimeric DNA Clamp for Enzyme Colocalization and DNA Functionalization
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Digital to Biological Translation: How the Algorithmic Data-Driven Design Reshapes Synthetic Biology

1
Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
2
Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar 25130, Pakistan
3
Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea
*
Author to whom correspondence should be addressed.
SynBio 2025, 3(4), 17; https://doi.org/10.3390/synbio3040017
Submission received: 16 September 2025 / Revised: 21 October 2025 / Accepted: 5 November 2025 / Published: 7 November 2025

Abstract

Synthetic biology, an emergent interdisciplinary field integrating principles from biology, engineering, and computer science, endeavors to rationally design and construct novel biological systems or reprogram extant ones to achieve predefined functionalities. The conventional approach relies on an iterative Design-Build-Test-Learn (DBTL) cycle, a process frequently hampered by the intrinsic complexity, non-linear interactions, and vast design space inherent to biological systems. The advent of Artificial Intelligence (AI), and particularly its subfields of Machine Learning (ML) and Deep Learning (DL), is fundamentally reshaping this paradigm by offering robust computational frameworks to navigate these formidable challenges. This review elucidates the strategic integration of AI/ML/DL across the synthetic biology workflow, detailing the specific algorithms and mechanisms that enable rational design, autonomous experimentation, and pathway optimization. Their advanced applications are specifically underscored across critical facets, including de novo rational design, enhanced predictive modeling, intelligent high-throughput data analysis, and AI-driven laboratory automation. Furthermore, pivotal challenges, such as data sparsity, model interpretability, the “black box” problem, computational resource demands, and ethical considerations, have been addressed, while concurrently forecasting future trajectories for this rapidly advancing and convergent domain. The synergistic convergence of these disciplines is demonstrably accelerating biological discovery, facilitating the creation of innovative and scalable biological solutions, and fostering a more predictable and efficient paradigm for biological engineering.

Graphical Abstract

1. Introduction

Synthetic biology represents a burgeoning interdisciplinary field that fundamentally redefines humanity’s interaction with biological systems. It integrates core principles from biology, engineering, and computer science to achieve ambitious goals: the design and construction of entirely new biological entities, such as enzymes, genetic circuits, and cells, or the systematic redesign of existing biological systems [1]. This discipline approaches biology with an engineering mindset, aiming to program biological processes with novel functions by starting from fundamental genetic components that can be assembled and programmed to perform specific functions [1,2]. Much like an engineer crafts a high-tech device, synthetic biologists seek to build with biology, where the “program” operates within a living system. A central tenet of synthetic biology is the application of engineering principles, including standardization and the creation of controlled circuits, to develop biological solutions across diverse sectors [3]. Importantly, this engineering-inspired perspective differentiates synthetic biology from traditional biotechnology, as it emphasizes modularity, scalability, and predictability in design.
The potential applications of synthetic biology are vast and span critical areas of human endeavor [3]. In healthcare, it facilitates the creation of new therapies, improves disease diagnosis and monitoring, and develops novel research tools. Specific examples include engineering cells to produce therapeutic molecules with precise targeting, advancing rational drug design research [4], and contributing to immunotherapy for cancer. The development of mRNA vaccines, for instance, stands as a significant innovation in combating infectious diseases and exploring new approaches for cancer treatment. These advancements highlight the translational potential of synthetic biology, bridging fundamental research with real-world clinical applications. Beyond medicine, synthetic biology offers transformative solutions in industry, agriculture, and environmental management. Industrial applications encompass the sustainable production of biofuels, the manufacture of enzymes, and the creation of bio-based specialty products and bulk chemicals. In agriculture, synthetic biologists are developing microbes that can produce their own fertilizer, enhancing crop yield and potentially addressing global hunger, alongside engineering disease-resistant crops and specialty foods. Environmental applications include the creation of microbial biosensors for detecting pollutants and the development of microbes or plants for bioremediation of contaminated sites or water pollution. Furthermore, synthetic biology enables the engineering of bacteria to convert carbon emissions into common chemicals like acetone and isopropanol, redirecting greenhouse gases from the atmosphere. Even consumer products, such as the Impossible Burger, which utilizes a lab-engineered non-meat-based heme molecule [5], demonstrate synthetic biology’s broad reach and potential to redesign traditional processes for greater environmental sustainability [3,6,7]. Together, these examples illustrate not only the breadth of synthetic biology but also its capacity to address global challenges in health, sustainability, and climate change.
The systematic development and optimization of biological systems in synthetic biology are typically guided by the Design-Build-Test-Learn (DBTL) cycle [8]. This iterative framework combines experimental techniques with computational modeling. The cycle comprises four distinct stages:
Design: This initial phase involves conjecturing a DNA pattern or a series of cellular alterations intended to achieve specific objectives. Researchers may design new genes, select genetic parts from existing libraries, or employ computer simulations to model the anticipated behavior of the biological system.
Build: Following the design phase, this stage focuses on the physical development of the DNA fragment and its effective incorporation into a host cell. Key tools enabling this include gene synthesis, which allows for precise design of genes with specific sequences, and genome editing technologies like CRISPR-Cas9, which facilitate targeted modifications to an organism’s genome.
Test: Once constructs are built, they are rigorously tested to determine how well the assessed phenotype aligns with the desired outcome and to evaluate any off-target or unintended effects. High-throughput DNA sequencing technologies are crucial here, enabling rapid analysis of large volumes of genetic information to provide the necessary data for assessment.
Learn: Based on the results from the testing phase, the constructs are modified or refined. This learning process informs subsequent design iterations, and the DBTL cycle is repeated until the desired function is robustly achieved.
Despite the systematic nature of the DBTL cycle, the inherent complexity of biological systems poses a significant challenge, often acting as a bottleneck to efficient and predictable engineering. Synthetic biology fundamentally involves designing intricate biological entities, and the biological systems themselves are characterized by their “intricacy and interconnectedness”. Traditional approaches to circuit engineering have historically relied on “first-principles biophysical models” to predict behavior [9]. However, these models struggle significantly with the “non-linear, high-dimensional interactions between genetic parts and host cell machinery”. Such complex interactions violate fundamental assumptions about part modularity, which is critical for predictable engineering, and consequently undermine the predictive power of these biophysical models. This often forces the circuit engineering process away from precise predictive design and into a “regime of ad hoc tinkering”. This highlights a persistent tension between the vision of rational design and the reality of biological complexity. The very nature of biological complexity, therefore, limits the efficiency and predictability of the traditional DBTL cycle, making it a laborious and technically challenging endeavor [10].
Synthetic biology is consistently framed as an engineering discipline, applying engineering principles to program biology with novel functions. The essence of engineering lies in its capacity for predictable design and controlled outcomes [1,2,7,8]. However, a fundamental tension exists between this engineering aspiration and the reality of biological systems: the impact of introducing foreign DNA into a cell can be inherently difficult to predict. This unpredictability manifests in the need to test multiple permutations to obtain a desired outcome, leading to considerable iteration within the DBTL cycle. For synthetic biology to truly mature as a robust engineering discipline, it must bridge this predictability gap. The ability to accurately forecast how engineered biological systems will behave before extensive physical experimentation is paramount for achieving the efficiency, reliability, and scalability characteristic of other engineering fields. This unmet need for enhanced predictive power creates a natural point of convergence with Artificial Intelligence (AI), Machine Learning (ML), and Deep Learning (DL). By leveraging their ability to capture complex, non-linear, and high-dimensional patterns, these computational tools promise to transform synthetic biology from an iterative, empirical practice into a truly predictive science.

2. Strategic Integration: ML/DL as Catalysts in Synthetic Biology

2.1. Addressing Biological Complexity and Non-Linearity with ML/DL

The inherent intricacy and interconnectedness of biosystems present significant challenges in designing biological entities with desired properties. Traditional first-principles biophysical models often fall short in predicting outcomes due to the non-linear, high-dimensional interactions between genetic parts and host cell machinery. AI algorithms offer powerful solutions by leveraging vast amounts of biological data and sophisticated computational models to navigate this complexity (Figure 1). ML models, in particular, are adept at capturing complex, non-linear patterns in data that are not known a priori, and can account for latent variables that are beyond those explicitly derived from biophysical models. This capability provides them with significantly higher predictive power, especially in systems where knowledge is sparse or incomplete. DL networks further enhance this by encoding intricate non-linear connections between input values, allowing them to discover subtle synergistic effects, such as how specific combinations of amino acids can dramatically increase protein activity beyond what individual contributions would suggest. This ability to discern hidden relationships within complex biological data is critical for moving synthetic biology beyond empirical trial-and-error [11,12]. Notably, such predictive models not only accelerate design but also reduce dependency on exhaustive experimentation, a shift that has profound implications for cost, scalability, and reproducibility.

2.2. Accelerating the Synthetic Biology DBTL Cycle Through Data-Driven Approaches

The traditional DBTL cycle in synthetic biology, while systematic, can be time-consuming and iterative. AI, ML, and DL act as powerful catalysts by integrating data-driven approaches and high-throughput experimental methods, thereby dramatically accelerating the pace and scope of circuit design (Table 1; Figure 1). AI can directly guide the subsequent set of designs, significantly reducing the number of DBTL iterations required to achieve desired results. By transforming the cycle from reactive testing into proactive prediction, AI-driven pipelines minimize uncertainty at each stage. This acceleration is profound; for instance, the intense application of AI and robotics/automation could potentially enable the creation of a new commercially viable molecule in approximately six months, a stark contrast to the traditional timeline of around ten years. AI-driven tools optimize the entire design process, leading to reduced time and cost, while simultaneously enhancing the precision and efficiency of synthetic biology projects. Such integration represents a paradigm shift from a labor-intensive, trial-based framework to a scalable, automated innovation pipeline, mirroring the acceleration previously seen in computational drug discovery. This transformation allows researchers to navigate complex design spaces more efficiently and converge on target behaviors within a limited number of cycles [9,13].

2.3. Synergistic Relationship: Data Generation from Synthetic Biology for ML/DL Training

A powerful synergy exists between synthetic biology and ML/DL, creating a mutually reinforcing ecosystem for innovation. Synthetic biology, through its experimental advancements, is uniquely positioned to “produce large data sets for training models,” for example, by leveraging high-throughput DNA synthesis. These vast datasets are precisely what ML/DL models require to learn complex patterns and make accurate predictions (Table 1). The growing availability of multi-omics datasets (genomics, transcriptomics, proteomics, and metabolomics) further strengthens this integration, enabling multi-layered models that can capture system-level dynamics.
Conversely, the trained ML/DL models are then “employed to inform design” within synthetic biology, guiding researchers by “generating new parts or advising unrivaled experiments to perform”. This creates a robust feedback loop: synthetic biology generates the necessary data for AI to learn, and AI, in turn, provides the predictive and generative capabilities to guide and optimize synthetic biology designs. This dynamic interaction accelerates the entire research and development process, transforming it from a largely empirical endeavor into a more rational and data-driven engineering discipline [9,13]. Looking ahead, this bidirectional flow is expected to mature into fully autonomous “closed-loop” platforms, where AI continuously designs, experiments, learns, and re-designs with minimal human intervention, ushering in the era of self-driving laboratories for biology.

2.4. AI as a “Predictive Powerhouse” for Non-Intuitive Design

Traditional synthetic biology faces significant hurdles in designing complex biological systems due to “non-linear and context-dependent interactions” that make predictive design challenging. Human intuition and explicit mechanistic understanding often fall short when confronted with the vast and intricate design spaces of biological components (Figure 1). This is where AI, particularly ML, emerges as a “predictive powerhouse.” ML models are uniquely capable of “capturing complex, non-linear patterns in the data that are not known a priori”. This means AI can identify “non-intuitive circuit design rules” or propose “innovative pathways that may not be immediately obvious to human researchers” [21,22].
The ability of AI to discern these subtle, hidden patterns allows synthetic biologists to explore and optimize design spaces that would otherwise be inaccessible through traditional, hypothesis-driven approaches. By moving beyond the limitations of human intuition and explicit mechanistic models, AI enables the discovery and creation of novel biological entities and functions that might not have been conceived through conventional means. In this sense, AI shifts the role of the researcher from designer to curator, evaluating and validating machine-proposed architectures that may extend beyond human foresight. This represents a critical shift in the design paradigm, where AI’s pattern recognition capabilities directly lead to the unlocking of unprecedented biological designs [21,22]. Looking ahead, explainable AI frameworks will be essential to ensure that these non-intuitive discoveries are not only effective but also interpretable, thereby building trust in AI-assisted design.

2.5. The Virtuous Cycle of Data and Design

The integration of ML/DL into synthetic biology establishes a virtuous cycle of data and design, fundamentally transforming the DBTL process. Synthetic biology experiments, especially those employing high-throughput techniques, are capable of “producing large data sets for training models”. This ever-increasing volume of biological data serves as the fuel for AI algorithms (Table 1). Crucially, the quality, diversity, and standardization of these datasets determine the performance ceiling of ML/DL models, underscoring the importance of robust data curation strategies.
In return, ML/DL models are not merely passive analytical tools; they actively “inform design” and “advise unrivaled experiments to perform”. This means that the data generated from one iteration of the DBTL cycle is fed into AI models, which then learn from these observations to provide rapid, data-driven feedback for the next design iteration. This iterative process, where AI leverages growing datasets from high-throughput biological experiments, effectively “reshapes” and “accelerates” the entire DBTL cycle. Over time, this creates a “learning ecosystem” in which synthetic biology continuously refines its own design capabilities, with AI acting as both analyst and architect. The outcome is a self-improving system where each experiment generates data that makes the AI smarter, leading to more efficient, precise, and effective designs in subsequent cycles. Such feedback-driven integration is a stepping stone toward autonomous, closed-loop experimental platforms, where AI dynamically designs, executes, and evaluates biological systems with minimal human intervention. This continuous feedback loop is a crucial causal relationship, demonstrating how the two fields mutually reinforce each other’s progress, driving innovation at an unprecedented pace [21,22].
In summary, the integration of AI, ML, and DL into synthetic biology is not merely an incremental improvement but a paradigm shift. By addressing biological complexity, accelerating the DBTL cycle, creating reciprocal data-model synergies, enabling non-intuitive design, and establishing self-reinforcing feedback loops, these computational approaches collectively transform synthetic biology from an empirical, trial-and-error practice into a predictive, scalable and increasingly autonomous discipline. Looking forward, the maturation of explainable AI, integration of multi-omics datasets, and development of closed-loop experimental platforms will be pivotal in realizing the full potential of this convergence, ultimately fostering a new era of rational and automated biological design.

2.6. Integration Process of AI/ML/DL in the Synthetic Biology Workflow

The integration of AI/ML/DL transforms the synthetic biology workflow by shifting the iterative Design-Build-Test-Learn (DBTL) cycle from a largely heuristic, manual process into a data-driven, closed-loop system [23]. This strategic integration is not uniform; rather, specific computational architectures are deployed at each stage to address distinct challenges, enabling automation, prediction, and optimization across the entire biological engineering pipeline (Figure 2). In the design stage, AI’s primary role is to rationalize hypothesis generation and explore the vast sequence and pathway space. Generative models, particularly, Deep Learning (DL) architectures like transformers (for sequence data) and Graph Neural Networks (GNNs) (for network data), are central. Transformers, utilizing their self-attention mechanisms, are trained on immense datasets of known functional DNA, RNA, or protein sequences. They learn long-range dependencies and complex regulatory grammar, allowing them to generate de novo sequences for high-performing genetic parts (e.g., strong promoters, optimized RBS). GNNs are applied to model existing metabolic or regulatory networks, predicting the impact of adding or deleting specific nodes (genes/enzymes) on the system’s flux or stability [24,25,26]. The output is an optimized, high-confidence biological blueprint, effectively replacing ad hoc design with rational prediction, significantly increasing the probability of in silico success.
The build stage focuses on the efficient, reliable physical construction of the designed biological parts. Classical AI algorithms and ML are integrated with laboratory automation. Bayesian optimization and ML-based control systems are utilized to manage and optimize robotic assembly platforms. These algorithms take parameters from previous successful builds (e.g., reagent concentrations, reaction times, temperature profiles) and intelligently adjust the liquid handling and DNA assembly protocols to maximize the yield and purity of the final genetic construct. This ensures that the physical realization of the AI-designed construct is executed with minimal error and resource waste resulting in high-throughput, precise, and cost-effective construction, enabling the rapid creation of large, diverse genetic libraries necessary for the testing phase.
The test stage generates enormous, high-dimensional datasets (microscopy, genomics, and proteomics) that require immediate, objective analysis using DL [27]. CNNs and image recognition DL models are trained to autonomously analyze complex visual data from High-Content Screening (HCS), such as classifying cellular phenotypes or quantifying fluorescent reporter levels from microscopy images. Similarly, ML is used for rapid processing and feature extraction from omics data, identifying subtle, non-linear patterns that correlate sequence design with functional output. This integration allows for real-time, high-throughput phenotyping, transforming raw experimental data into actionable performance metrics needed for the subsequent learn stage.
The learn stage is where the cycle becomes “intelligent,” utilizing the data from the test stage to refine the models and dictate the next course of action. This is the stage that truly closes the DBTL loop. Reinforcement Learning (RL) and active learning (AL) models are employed. RL frameworks treat the synthetic organism and the experimental setup as an environment, learning an optimal policy to achieve a defined goal (e.g., maximum yield) through simulated or real-world experimentation. AL is used to prioritize future experiments by calculating which unsampled region of the design space would provide the most information gain for the predictive model, thereby minimizing unnecessary wet-lab work. This integration results in a self-optimizing system that accelerates convergence toward the optimal biological design, minimizing the number of expensive, time-consuming experimental cycles required for discovery. This comprehensive, stage-specific integration of AI/ML/DL creates a powerful synergy, realizing the potential for autonomous biological engineering.

2.7. Real-World Applications of AI in Synthetic Biology

The integration of AI is rapidly transitioning from theoretical promise to laboratory-validated practical application in synthetic biology. These real-world successes underscore a shift from simple prediction toward AI-driven co-design and autonomous experimentation. One of the most impactful applications lies in optimizing gene and protein components. Yan et al. (2024) utilizes a DL workflow to solve the crucial genetic engineering problem of optimizing the N-terminal Coding Sequence (NCS) that impacts the translation initiation rate to maximize gene expression [15]. Traditional methods for optimizing the NCS are labor-intensive and yield only small improvements. Using Green Fluorescent Protein (GFP) in Bacillus subtilis as a reporter, the model generated an optimal NCS variant (MLD62) that achieved a 5.41 fold increase in GFP expression. The engineered NCS (MLD62) was successfully demonstrated to boost the production of the valuable chemical N-acetylneuraminic acid by enhancing the expression of the crucial rate-limiting GNA1 gene. The study demonstrates a powerful method for enhancing gene expression regulation using AI, highlighting the utility of few-shot learning in overcoming data sparsity challenges common in synthetic biology. Similarly, transformer-based architectures have been used for de novo promoter design and protein variant generation, demonstrating the growing maturity of DL models in producing experimentally verifiable outputs [28,29]. These generative models are crucial for navigating the vast sequence spaces that define biological function.
AI is also fundamentally restructuring metabolic engineering for the production of valuable chemicals, biofuels, and pharmaceuticals. The MINN (Multi-omics Integrated Neural Network) model represents a significant advance by integrating diverse multi-omics data directly into Genome-Scale Metabolic Models (GEMs) to accurately predict metabolic fluxes [30]. This predictive power guides researchers to the most effective genetic edits for maximizing product yield. Furthermore, GNNs are proving essential for analyzing the relational structure of biological systems. Studies have utilized GNNs in conjunction with GEMs for tasks like predicting gene essentialities, identifying which genes are critical for cell viability—a vital consideration for industrial strain stability [31]. Specialized GNN variants, such as the Directed Message Passing Neural Network (D-MPNN) and Graph Attention Networks (G-Attn), have been successfully applied to predict the complex properties of biofuel-related species and chemical molecules, thus accelerating the development of sustainable production platforms [32].
The ultimate integration is the realization of closed-loop, autonomous experimentation. Recent studies have successfully demonstrated systems where RL/AL models are coupled with robotic platforms to complete the entire DBTL cycle without human intervention. These systems leverage ML models to analyze test results, learn from them, and propose the next most informative experiment, effectively self-optimizing genetic circuits or metabolic pathways in real-time [15]. In a study, RL is applied to control the microbial communities in bioreactors, where AI bioprocess development simulator learned to dynamically adjust nutrient flow rates to maximize productivity while maintaining cultural species in balance something traditional proportional-integral-derivative controllers failed to achieve [33]. This capability marks the culmination of AI integration, moving the field beyond mere prediction toward autonomous discovery. Collectively, these cases confirm a decisive transition: AI in synthetic biology is no longer a theoretical pursuit but an indispensable tool that is actively reshaping biological experimentation. While these successes underscore the growing maturity of AI-driven bio-design, they simultaneously highlight persistent challenges regarding model interpretability, generalization across different biological hosts, and the necessity for higher quality, standardized training data.

3. Advanced Applications of ML/DL in Synthetic Biology

3.1. Rational Design and Engineering of Biological Systems

The integration of AI has moved synthetic biology from a discipline of iterative tinkering to one predictive design. By leveraging vast datasets, ML/DL models now provide a computational scaffold for rationally engineering biological systems, significantly accelerating the design process and enhancing the predictability of outcomes.

3.1.1. Genetic Circuit Design and Optimization

AI algorithms, particularly machine learning models, are pivotal in the rational design and optimization of genetic circuits. These models analyze data from previous experiments to predict the behavior of new genetic designs, enabling highly effective predictive modeling. ML significantly enhances synthetic gene circuit engineering, addressing challenges from individual components to complex circuit-level interactions. AI-powered modeling engines integrate biological knowledge with machine learning to provide real-time design feedback, allowing researchers to predict protein expression levels, anticipate off-target effects or crosstalk, estimate metabolic burden on host cells, and identify potential failure points before physical fabrication [34,35]. Furthermore, advanced ML paradigms are critical for navigating the vast and complex design spaces of genetic circuits. RL models can train agents to explore these spaces and learn optimal actions for desired circuit designs. Active learning and sequential model-based optimization methods iteratively refine design exploration while minimizing the need for extensive data acquisition, focusing on the most informative experiments. This systematic, AI-guided approach overcomes the limitations of traditional, often inefficient, ad hoc tinkering, leading to more precise and predictable circuit behaviors [36,37].

3.1.2. De Novo Design of Novel Biological Components and Parts

Beyond optimizing existing designs, ML/DL models are increasingly employed for the de novo design of entirely novel biological components and parts. These models have moved beyond passive analysis to active generation, directly creating new parts with desired properties. A new generation of generative models has demonstrated the capability to design proteins with desired functions from scratch. The emergence of foundation models (FMs) is particularly transformative in this area. These large-scale, self-supervised models, leveraging powerful analogies between natural language and biological sequences, directly support complex biological sequence-design tasks. This includes the engineering of novel proteins, the design of small molecules, and the generation of genomic sequences with specific properties. Such generative capabilities are fundamentally expanding the frontiers of synthetic biology, enabling the rational creation of biological entities devoid of natural precedent [38,39,40].

3.1.3. Protein Engineering and Enzyme Optimization

ML is a powerful computational tool revolutionizing enzyme engineering, selection, and design. It facilitates the identification of enzyme functions, optimizes crucial experimental properties such as the enzyme environment and reactants, and enables the design of novel enzymes. DL models, exemplified by breakthroughs like AlphaFold, have achieved unprecedented accuracy in protein structure prediction by training on thousands of publicly available protein structures, significantly outperforming historical methods. These advanced methods are also applied to predict complex bimolecular interactions among proteins, nucleic acids, small molecules, and ions, and to design de novo proteins with specific desired functions [41,42,43].
ML-guided platforms integrate various high-throughput experimental techniques, such as cell-free DNA assembly, gene expression, and functional assays, to rapidly map complex fitness landscapes across vast protein sequence spaces (Table 1). This allows for the efficient optimization of enzymes for multiple, distinct chemical reactions. Recent advancements include ML-aided design and screening that have led to the targeted engineering of proteins for industrial and biomedical applications, even enabling previously unforeseen emergent functions like spatiotemporal self-organization within synthetic cells. This capability significantly accelerates the development of specialized biocatalysts and novel protein functions [41,42,43].

3.1.4. Metabolic Pathway Design and Optimization

ML is fundamentally transforming the design and optimization of metabolic pathways for the prediction of biofuels, pharmaceutical, and commodity chemicals. ML algorithms rapidly analyze vast genomic and proteomic datasets to accurately predict enzyme functions, and substrate specificities, thereby facilitating the design of novel and non-native metabolic pathways. Beyond prediction, ML excels at optimizing these pathways to enhance yield and efficiency, superseding laborious trial-and-error methods through its ability to identify intricate patterns in experimental data and recommend specific, impactful modifications. Techniques like RL are particularly powerful, autonomous refining pathway configurations for optimal output. Furthermore, ML enables the discovery of entirely new synthesis routes for target molecules by integrating diverse datasets, including genomic sequences, chemical properties, and metabolic databases, to propose innovative, often more efficient or sustainable, pathways that may elude conventional discovery. These predictive capabilities also extend to simulating the metabolic consequences of genetic edits within metabolic pathways, enabling researchers to prioritize beneficial modifications and mitigate unintended cellular outcomes, such as toxicity or reduced fitness, thereby fostering greener manufacturing processes and reducing reliance on traditional chemical synthesis [44,45,46].

3.1.5. DNA Sequence Optimization and Gene Editing

ML/DL have become indispensable for optimizing DNA sequences and enhancing the precision of gene editing. ML-based optimization engines can analyze a comprehensive range of factors crucial for gene expression and function including codon usage, mRNA secondary structure, regulatory element configuration, as well as the strengths of promoters, ribosome binding sites, and host-specific genomic features (Table 1, Figure 3). By training these models on large experimental datasets, AI tools can suggest high-performing genetic constructs with a significantly higher probability of proper expression and functionality, thereby streamlining the genetic engineering workflows. Beyond sequence optimization, neural networks and DL power the design of precise gene circuits and enhance the targeting efficacy of advanced gene editing technologies like CRISPR-Cas9 by predicting guide RNA activity and off-target effects. This computational guidance renders genetic manipulation more accurate, efficient, and predictable, representing a cornerstone of modern synthetic biology [47,48].

3.2. Predictive Modeling and Simulation of Biological Behavior

The capacity for predictive modeling represents a cornerstone of AI’s value to synthetic biology, enabling a shift from descriptive to perspective science. ML algorithms leverage historical and high-throughput data to forecast how new genetic designs with increasing accuracy. This predictive power is crucial for enhancing the reliability, scalability, and efficacy of synthetic biology applications, as it empowers researchers to simulate system performance under diverse environmental and genetic conditions. These AI-driven simulations enable the forecasting of intricate biological interactions, thereby guiding experimental design and accelerating the innovation cycles. Specifically, ML models can accurately predict protein expression levels, anticipate cross-talk and off-target effects and estimate the metabolic burden imposed by synthetic constructs. This computational foresight allows for the identification of bottlenecks and failure points at the in silico stage, long before physical fabrication, thereby minimizing costly, iterative experimental rounds. Ultimately this capability is foundational to transforming the design process from a craft of empirical discovery to an engineering discipline of rational prediction, significantly improving efficiency and success rates [46,49,50].

3.3. High-Throughput Data Analysis and Knowledge Extraction

The advent of high-throughput technologies has generated an unprecedented deluge of biological data, creating a critical demand for ML/DL applications to mine this information for novel insights. High-throughput DNA sequencing, for instance, has revolutionized synthetic biology by enabling the rapid generation of genetic information, which forms the fundamental basis for designing and constructing synthetic systems. ML tools are now deeply integrated with these high-throughput experimental approaches within the DBTL process, not only accelerating circuit design but also efficiently processing and interpreting the resulting multimodal data (Table 1). ML is uniquely equipped for distilling complex patterns within the data-rich outputs provided by system biology, moving beyond correlation to uncover causal relationships. ML algorithms are adept at extracting meaningful biological insights from massive omics datasets, thereby leading to a more comprehensive understanding of cellular function and informing more rational design decisions. Furthermore, automated analysis of microscopy data exemplifies significant achievement at the intersection of engineering biology and ML/DL, streamlining the high-content evaluation of cellular responses and phenotypes. This collective capability to rapidly analyze and extract actionable knowledge from vast, heterogeneous biological datasets is the engine that drives the iterative improvements central to synthetic biology [12,51,52].

3.4. Automation of Laboratory Processes and Experimental Design

The integration of ML/DL is profoundly transforming laboratory operations, ushering in an era of unprecedented and intelligent automation. AI orchestrates can be applied across the entire scientific research and development (R&D) process, from the precise design of laboratory experiments to the creation of protocols to run specific laboratory equipment, thereby democratizing complex procedures and enhancing reproducibility. The synergistic application of AI, robotics and automation is projected to radically accelerate synthetic biology timelines. Experts suggest that this convergence could enable the creation of a new commercially viable molecule in approximately six months, a dramatic reduction from the traditional ten years. Automation of the assembly process within the DBTL framework directly reduces the time, labor, and cost associated with generating multiple constructs, thereby increasing throughput and shortening overall development cycles [53,54].
AI serves as the central nervous system for these automated workflows, enabling the development of integrated software platforms that collect data seamlessly from lab automation systems and sensors. These platforms leverage ML to identify trends, outliers, and optimal parameters, thereby subsequently recommending design improvements for the next iteration of experiments. This creates a closed-loops, self-optimizing system that becomes “smarter with every experiment,” dramatically enhancing cost-effectiveness and accelerating the path to successful outcomes. This is exemplified by AI-driven solutions such as microbial colony picking systems, High-Content Screening systems for cellular analysis, and single-cell dispensers that provide rapid and traceable monoclonality proof. Collectively, this automation acts as a force multiplier, industrializing the entire DBTL cycle and propelling the pace of discovery [53,54].

3.5. The Role of Foundation Models and Generative AI

The landscape of AI in synthetic biology is being radically reshaped by the emergence of Generative AI and Foundation Models (FMs). Generative AI distinguishes itself by generating novel, functional outputs based on patterns it detects, rather than merely predicting or classifying existing data. This capability is predominantly built upon transformer architectures, which excel at synthesizing complex data patterns. FMs are defined by their ability to learn from massive, task-agnostic pre-training, making them highly adaptable to a wide range of downstream tasks with minimal fine-tuning. These models are poised to transform AI research, particularly in biological design applications, by exploiting the inherent linguistic analogies between natural language and biological sequences. This allows FMs to directly tackle complex biological sequence-design tasks, including de novo protein engineering, small molecule design, and genomic sequence design [12,38].
Large Language Models (LLMs), a prominent class of generative AI, have achieved remarkable performance in natural language tasks. Their success is founded on the transformer architecture and is now being translated to biological sequences, framing DNA, RNA, and proteins as “biological languages” to enable new approaches to biological design. For instance, models like Evo, trained on millions of prokaryotic genomes [55], are emerging as early-generation foundation models capable of making pan-genomic functional predictions, thus serving as valuable community resources for gene circuit design. This represents a significant leap towards AI systems that can not only analyze but also autonomously design novel biological entities with desired properties, heralding a new era of generative biology.

3.6. From Prediction to Generation: The Evolution of AI’s Impact

The evolution of AI’s impact in synthetic biology marks a significant and transforming progression from passively merely predicting biological phenomena to actively designing and generating novel biological entities. Early applications of ML/DL primarily focused on “predictive modeling”, aiming to forecast system behavior or to identify optimal parameters within existing systems. This predictive capability was, and remains, crucial for streamlining the DBTL cycle and reducing experimental iterations. However, a paradigm-shifting leap has occurred with the advent of generative AI. This advanced form of AI generates novel, functional outputs based on the patterns it detects, moving beyond analysis to de novo creation. This includes the ability to design novel enzymes with desired functions and to create entirely new biological entities that may not exist in nature or be discoverable through traditional means. The development of foundation models, which exploit powerful analogies between natural language and biological sequences, is the driving force behind this shift, enabling the rational design of proteins, small molecules, and genomic sequences from scratch. This evolution fundamentally changes the scope of synthetic biology from a field primarily focused on understanding and modifying existing biological systems to one that actively engineers and creates new biological functions and forms. The transition from prediction to generation signifies a profound expansion of synthetic biology’s capabilities, enabling true biological engineering [39,56,57].

3.7. AI-Driven Automation as a Force Multiplier for Discovery

The DBTL cycle, while fundamental to synthetic biology, is inherently iterative and can be laborious, creating significant operational bottlenecks that traditionally hamstring the pace of discovery and optimization. AI-driven automation serves as a direct antidote to these constraints, significantly reducing the time, labor, and cost associated with biological experimentation 5 and dramatically increasing throughput. The ambitious goal of creating a commercially viable molecule in approximately six months instead of roughly ten years vividly illustrates the dramatic acceleration enabled by this powerful convergence. AI’s role in this transformation extends far beyond the mere orchestration of robotic systems. It involves the creation of sophisticated software platforms that autonomously recommend design improvements for the next iteration. This capability effectively establishes a self-improving feedback loop that becomes smarter with every experiment, creating a closed-loop, self-optimizing laboratory environment. This virtuous cycle leads to continuous and autonomous improvement, minimizing human intervention. This deep integration of AI’s analytical and predictive power with automated laboratory workflows acts as a powerful force multiplier for discovery. It transforms the pace of biological engineering from a human-limited endeavor, constrained by manual processes and empirical iteration, to an AI-accelerated process capable of exploring vast design spaces with unprecedented efficiency and speed [58].
In summary, the advanced applications of AI/ML span the entire synthetic biology workflow, creating a profound paradigm shift, progressing from using AI for predictive modeling of biological behavior to generative design of novel components, and fully to the full automation of the DBTL cycle. This evolution, from prediction to generation to autonomous discovery, is not incremental but transformative. The convergence of generative AI, foundation models, and intelligent robotics is no longer merely a tool but is becoming a co-pilot in biological engineering, fundamentally expanding the scope and accelerating the pace of what is possible in designing and programming living systems. While these advances underscore the transformative potential of AI in reshaping synthetic biology, they also surface challenges, ranging from data quality and model interpretability to ethical and biosafety considerations, that must be addressed to ensure reliable, responsible, and sustainable integration.

4. Critical Analysis of ML/DL in Synthetic Biology

4.1. Performance Disparities in ML/DL Approaches

The effectiveness of particular ML architectures in synthetic biology applications is not about their universal superiority, but rather their inherent capacity to discern and leverage domain-specific features [59]. GNNs, for instance, excel in task like metabolic pathway prediction and modeling protein–protein interactions. This is because GNNs are designed to explicitly encode the relational structures that are fundamental to biological systems. Their “message-passing” mechanisms perfectly align with how information flows within biochemical networks, enabling them to learn representations that accurately reflect genuine biological patterns [60,61]. On the other hand, Transformers truly shine in sequence-based tasks, such as designing promoters or predicting regulatory elements. Their powerful self-attention mechanisms allow them to capture long-range dependencies across vast genomic sequences, providing a representational depth that surpasses the locality-limited convolutional approaches [62]. It is crucial to remember, however, that the superior performance of these models heavily relies on the availability of high-quality, structured training data that accurately represents the biological domain they are targeting. Without this, the most sophisticated architecture will struggle to deliver optimal results. Thus, the architecture-data interplay must be viewed as symbiotic, where model choice is only as powerful as the biological data foundation on which it is trained.

4.2. Conditional Failures and Success of DL Models

Contemporary DL models, despite their power, often exhibit predictable failure modes directly tied to specific synthetic biology context [12]. For instance, GNNs typically falter when applied to poorly annotated or incomplete biological networks. Missing edges and nodes in these scenarios fundamentally alter the graph topology, rendering the learned representations unreliable. GNNs also struggle with novel synthetic circuits that significantly deviate from the naturally occurring network motifs present in their training data. Similarly, transformers, while incredibly powerful for sequence modeling, show diminished performance on short sequences where contextual information is limited. They can also generate biologically implausible sequences if their training dataset lack sufficient negative examples. Furthermore, these models often exhibit domain-specific brittleness when transferred across different organism types or synthetic biology platforms. This suggests that their learned representations might be more organism-specific than initially anticipated. To achieve successful outcomes with these models, several conditions are crucial. These include access to comprehensive training dataset, meticulous feature engineering that accurately reflects genuine biological constraints, and robust validation protocols designed to test model robustness across diverse experimental conditions. Future progress will likely hinge on hybrid approaches that combine domain-informed constraints with data-driven architectures, thereby reducing brittleness while improving generalizability [12].

4.3. Limitations in Current Benchmarking Practices

Existing benchmarks have fundamental design flaws that limit their usefulness for evaluating and comparing models [63]. Most of them focus on narrow, well-defined tasks, like predicting protein stability or estimating metabolic flux. Nevertheless, they often miss the mark by not capturing the multi-objective, constraint-heavy nature of real-world design challenges (Table 2, Figure 4). The datasets used in benchmarks frequently have major imbalances. Successful products are vastly under-represented compared to failed attempts. This can lead to models that look good statistically but are not very practical. On top of that, many benchmarks rely on simulated or retrospective data. This data might not accurately reflect the complexity and noise found in actual experimental work.
Another big miss is the neglect of the temporal dimension. Models often train on static datasets, ignoring the iterative and adaptive nature of the design cycle. This means our current benchmarks are not really pushing models to solve the complex, real-world problems. As a result, many models that perform impressively on these standardized tests but struggle to transfer that performance to new challenges. Redesigning benchmarks to capture temporal dynamics, multi-objective optimization, and failure-rich datasets will be essential to produce truly translational AI for synthetic biology.

4.4. The Accuracy–Interpretability Trade-Off

The community’s singular emphasis on predictive accuracy has inadvertently created a critical void in both model interpretability and readiness for regulatory compliance. While accuracy metrics offer quantifiable measures of model performance, they provide limited insight into the underlying biological rationale for predictions. This opacity presents significant impediments to achieving regulatory approval and successful clinical translation [64]. The intricate non-linear transformations inherent in DL models generate predictions that are, in essence, “black boxes”. This makes it exceedingly difficult to discern “why” specific design choices are recommended or how to appropriately interpret model confidence. Such as lack of transparency is particularly problematic during model failures, as practitioners cannot readily identify the source of errors or effectively adjust their design strategies. Moreover, the absence of interpretability hinders the crucial integration of domain expertise, preventing researchers (mainly biologists) from effectively combining their nuanced understanding of biological principles with model outputs.
Regulatory agencies, especially for therapeutic applications, mandate clear mechanistic explanations for synthetic biology products. Yet, current computational models offer minimal insight into the biological pathways and molecular interactions that drive their predictions, posing a formidable challenge to their broader adoption and impact. This reality underscores the urgent need for explainable AI (xAI) solutions tailored to biological contexts, where interpretability is not a luxury but a prerequisite for safety, adoption, and regulatory approval [64,65]. The implication is that simply achieving high predictive accuracy is often insufficient for widespread adoption in a scientific discipline that emphasizes fundamental biological insights. Without a clear understanding of “why” a model makes a particular prediction, it becomes challenging for researchers to trust its outputs, troubleshoot unexpected behaviors, or derive new biological knowledge. Bridging the accuracy–interpretability divide will require a cultural shift in model development, with equal weight given to mechanistic transparency and predictive performance.

4.5. Dataset Standardization and Reproducibility Concerns

The lack of standardized datasets and evolution protocols has become a major roadblock for reproducible research. Currently, individual research groups often create their own datasets using different experimental methods, data preprocessing techniques, and annotation standards. This makes it incredibly difficult to compare results across studies or even reproduce published findings [66,67,68]. This fragmentation is made worse by the fact that many industrial synthetic biology datasets are proprietary. This severely limits the amount of high quality, diverse training data available to academic researchers. The problem of non-standardization is not just about data collection; it also extends to how success is measured, the variety of experimental conditions used, and how missing or uncertain data is handled (Table 2). These inconsistencies mean that reported performance improvements in the literature might just be due to differences in the datasets used, rather than true advances in algorithms.
The issue is further complicated by the limited availability of negative control data. Failed synthetic biology experiments are rarely reported or shared. This creates training datasets that might not accurately represent the full range of possible outcomes, leading to models that are less robust in real-world applications. Addressing this gap will require community-driven initiatives to establish shared data repositories, standardized reporting practices, and incentives for publishing negative results, all of which are critical to building trustworthy and generalizable AI models in synthetic biology.

4.6. Fundamental Limitations in Current DL Approaches

Despite impressive performance on benchmark tasks, DL models exhibit several fundamental limitations that compromise their real-world utility. For one, training data biases, often a result of historical experimental focuses and publication trends, can lead these models to simply perpetuate existing design prejudices instead of uncovering genuinely novel synthetic biology solutions [69,70]. Overfitting remains a persistent challenge, especially considering how small most synthetic biology datasets are compared to the vast parameter space of modern DL architectures. These models frequently show poor transferability across different experimental conditions, organism types, or synthetic biology platforms. This suggests they might be learning specific quirks of the datasets rather than general biological principles.
Perhaps more critically, the interpretability deficit makes it incredibly difficult to tell whether models are truly capturing biological mechanisms or just memorizing statistical patterns in the training data. This is a significant concern given the safety-critical nature of many synthetic biology applications, where model failures could have serious consequences for human health or environmental safety (biosafety and biosecurity). Taken together, these limitations highlight a central paradox: models that appear powerful in silico often collapse under the weight of real-world complexity, underscoring the need for fundamentally new approaches that embed biological priors into AI systems.

4.7. The Implementation Gap in Real-World Applications

A significant disconnect exists between the sophisticated ML models published in academic literature and their actual implementation in industrial pipelines. While academic studies frequently report impressive applications of DL to industrial challenges, remarkably few of these models have been successfully integrated into commercial drug discovery or biotechnology workflows [71,72,73,74]. This implementation gap stems from several practical hurdles rarely addressed in academic publications. Industrial synthetic biology demands models capable of operating efficiently under tight time and resources constraints, providing reliable uncertainty quantification, and seamlessly integrating with existing laboratory automation systems (Table 2). Many published models, however, necessitates extensive computational resources or specialized expertise that are unavailable in industrial environments.
Furthermore, these models frequently lack the robustness and reliability essential for high-stakes commercial applications, where inaccuracies can lead to substantial financial losses or significant regulatory setbacks. The existing regulatory landscape further complicates deployment, as synthetic biology products must meet stringent safety and efficacy standards that current models are not inherently designed to address. This fundamental misalignment between academic focus and industrial requirements calls for a new research paradigm centered on “deployment-readiness” rather than proof-of-concept performance.

4.8. Are DL Models Truly Learning Biological Principles?

A fundamental question arises: are current DL models genuinely learning biological principles or merely memorizing statistical patterns with their training datasets? This distinction is crucial, where the aim is to design novel biological systems that go beyond existing natural or synthetic examples [75,76,77]. Evidence suggests that many models might be operating closer to the memorizing end of the spectrum. Their poor performance on out-of-distribution samples and their inability to generalize to new synthetic biology contexts clearly demonstrate this. The limited diversity in most synthetic biology datasets, coupled with high dimensionality of biological sequence and structure spaces, creates conditions where memorization can easily be mistaken for successful learning.
For instance, models trained on protein sequences might learn to recognize sequence motifs linked to specific functions without grasping the underlying biochemical mechanisms driving those connections. This memorization-based learning becomes a real problem when models encounter novel synthetic biology designs that differ from their training examples. They simply lack the mechanistic understanding needed to make reliable predictions about biological systems they have never seen before. Moving forward, progress will depend on hybrid approaches that integrate data-driven learning with explicit mechanistic modeling, ensuring that AI captures causality rather than correlation.

4.9. Feasibility of AI-Generated Synthetic Biology Products

The practical feasibility of AI-generated products, successfully navigating real-world development pipelines, is highly questionable given current technological limitations. While computational models can generate designs that meet in silico optimization criteria, the leap from digital prediction to functional biological system involves numerous challenges that current AI approaches simply do not adequately address [78,79,80]. Typically, these models optimize for single objectives or overly simplified multi-objective functions (Table 2). They often fail to capture the full complexity of biological systems, including context-dependent interactions, evolutionary pressure, and emergent properties that arise from system-level integration. Crucially, manufacturing constraints, regulatory requirements, and economic considerations are rarely factored into AI-driven design processes. This often leads to products that might be theoretically optimal but are practically infeasible to produce or deploy.
Furthermore, current static modeling approaches do a poor job of capturing the temporal dynamics of biological systems, such as adaptation, evolution, and degradation over time. Until AI models can incorporate temporal, evolutionary, and systems-level constraints, their outputs will remain more aspirational than actionable for synthetic biology innovation.

4.10. Prerequisites for AI Integration in Synthetic Biology

Before AI can truly become a standard tool in synthetic biology design, there is a need to overcome several critical challenges [81,82]. Firstly, interpretable AI models should be developed. These models must offer mechanistic insights into their predictions, which is vital for both regulatory compliance and scientific understanding. They should be able to explain their reasoning in terms that synthetic biologists can readily grasp and validate through experiments. Secondly, comprehensive standardization of datasets, experimental protocols, and evaluation metrics is essential (Table 2). This will enable reproducible research and allow for meaningful comparisons between different models. Thirdly, multi-objective optimization frameworks must be integrated. These frameworks need to simultaneously consider biological function, manufacturability, safety, and economic factors to be truly useful in practical applications. Fourth, developing robust uncertainty quantification methods is crucial. These methods will help practitioners understand when models are operating within their reliable prediction domains, adding a layer of confidence to AI-driven designs. Finally, establishing regulatory frameworks that can accommodate AI-generated synthetic biology products while maintaining appropriate safety standards is indispensable for commercial viability (Table 2). Taken together, these prerequisites define a roadmap for transforming AI from an experimental curiosity into a foundational design partner for synthetic biology.

4.11. Evolution of Datasets for Multi-Objective Optimization

The future evolution of synthetic biology datasets is paramount to supporting the intricate, multi-objective optimization challenges inherent in real-world applications. Current datasets predominantly focus on single objectives, such as protein stability or metabolic flux. However, practical synthetic biology design necessitates simultaneous optimization across numerous, often conflicting, objectives including biological function, manufacturability, safety, environmental impact, and economic viability [83,84,85,86,87]. Future datasets must incorporate comprehensive experimental metadata, capturing not only successful designs but also failed attempts, partial successes, and the specific conditions under which diverse outcomes were observed. Including “negative results” and context-specific metadata will be critical for avoiding biased training and enabling more robust predictions. The temporal dimension also needs explicit inclusion, with longitudinal data tracking the performance and stability of synthetic biology systems over time. Furthermore, datasets should integrate information regarding regulatory constraints, manufacturing limitations, and market requirements to enable models capable of generating designs with genuine commercial potential.
Developing standardization ontologies and annotation frameworks will be crucial for facilitating data sharing and integration across various research groups and industrial partners (Table 2). Here, the establishment of community-driven, FAIR compliant (Findable, Accessible, interoperable, and Reusable) datasets should be emphasized as a practical next step. Ultimately, establishing large-scale collaborative initiatives, akin to those in genomics and structural biology, will be essential to generate the comprehensive, high-quality datasets required for robust multi-objective optimization in synthetic biology. Such initiatives would transform data from fragmented silos into shared infrastructure, enabling AI models to move beyond narrow tasks toward holistic, multi-objective design frameworks.

4.12. Outlook: Hybrid Models, Advanced Automation and Societal Impact

The future of ML/DL in synthetic biology is poised for continued transformative growth, driven by several key developments. A promising direction involves the development of hybrid approaches that combine the strengths of ML with mechanistic modeling. These models can leverage the high predictive power of data-driven AI while simultaneously providing greater interpretability and prescriptive ability derived from mechanism-based models. This dual approach directly addresses the accuracy–interpretability trade-off and could provide the mechanistic grounding necessary for regulatory acceptance [88,89,90]. Advanced automation will continue to scale up the entire DBTL cycle through robotics-assisted laboratory automation. This includes the implementation of closed-loop pipelines for tasks like protein engineering and the deployment of agent- and -RL based learning approaches for fully autonomous algorithm ensembles that can navigate vast, high-dimensional design spaces. Integrating automation with AI ensures not just speed but also reproducibility, a critical bottleneck in current practice. Such automation promises to dramatically accelerate discovery timelines, potentially reducing the time to develop new biological products from years to months [89,90].
Efforts will also focus on addressing data limitations, particularly by developing new methods to compensate for shortcomings in data quality and quantity. Accessible fine-tuning and transfer learning approaches, leveraging established foundation models, will enable research groups without extensive high-throughput capabilities to tackle data-sparse problems more effectively. Furthermore, the advent of quantum computing holds the potential to solve massive, complex optimization problems much faster, which could significantly accelerate AI optimization in synthetic biology. This suggests that computational infrastructure itself, not just biological insight, will become a decisive factor in research competitiveness.
From a broader societal impact perspective, AI-driven optimization is expected to lead the charge in resource-efficient innovation and sustainability. Governments, such as the U.S. government, have already underscored their commitment to accelerating synthetic biology research, positioning it as a cornerstone for sustainability strategies. This includes significant investments aimed at developing new medicines, commodities, reducing waste, and advancing sustainable farming practices, all while mitigating climate change impacts. Framing AI-synthetic biology convergence as a societal and economic imperative, not just a scientific trend, will strengthen the relevance of this research to policy, funding, and global innovation ecosystems.

5. Conclusions

AI and its subtypes are catalyzing a paradigm shift in synthetic biology, transforming it from an empirical discipline into a predictive engineering science. Traditional DBTC cycles have been hampered by the inherent complexity and non-linear dynamics of biological systems, resulting in iterative, resource-intensive approaches with limited predictability. AI-driven methodologies address these fundamental limitations by capturing complex, non-linear patterns and latent variables that conventional biophysical models cannot adequately represent. This enhanced predictive capacity enables rational design of genetic circuits, de novo protein engineering, and optimized metabolic pathway construction. Crucially, in silico modeling capabilities reduce experimental iterations by forecasting biological behavior and guiding targeted experimental design. The synergy between synthetic biology and AI creates a self-reinforcing cycle: synthetic biology can generate high-quality/quantity datasets essential for training sophisticated AI models, while AI provides computational intelligence to optimize biological designs. This iterative refinement process, amplified by generative AI and laboratory automation, accelerates discovery timelines and expands the scope of engineerable biological functions.
Yet, major challenges remain. Limitations in data quality, completeness, and bias still constrain model reliability; the “black box “nature of deep models impedes mechanistic interpretability; and discrepancies between computational predictions and empirical outcomes underscore the need for rigorous validation. Ethical, safety, and regulatory frameworks have also lagged behind technological advances, raising critical questions about responsible governance and societal trust. Looking forward, progress will likely hinge on the development of hybrid models that integrate data-driven AI with mechanistic insight, ensuring both predictive accuracy and interpretability. Advances in laboratory automation, foundation models, and even quantum computing promise to reshape DBTL workflows and unlock multi-objective design at scale. Ultimately, the convergence of AI and synthetic biology is not merely a scientific milestone but a strategic imperative, offering transformative pathways to address global challenges in healthcare, agriculture, industrial biotechnology, and sustainability. Harnessing this convergence responsibly will not only accelerate biological discovery but also define the next era of innovation at the interface of life and technology.

Author Contributions

Conceptualization, A.M., N.Q. (Nabila Qayyum), R.R., N.Q. (Naila Qayyum) and S.I.; methodology, A.M., N.Q. (Nabila Qayyum), R.R., N.Q. (Naila Qayyum) and S.I.; validation, A.M., N.Q. (Nabila Qayyum), R.R., N.Q. (Naila Qayyum) and S.I.; formal analysis, A.M., N.Q. (Nabila Qayyum), R.R., N.Q. (Naila Qayyum) and S.I.; investigation, A.M., N.Q. (Nabila Qayyum), R.R., N.Q. (Naila Qayyum) and S.I.; resources, A.M., N.Q. (Nabila Qayyum), R.R., N.Q. (Naila Qayyum) and S.I.; data curation, A.M., N.Q. (Nabila Qayyum), R.R., N.Q. (Naila Qayyum) and S.I.; writing—original draft preparation, A.M., N.Q. (Nabila Qayyum), R.R., N.Q. (Naila Qayyum) and S.I.; writing—review and editing, A.M., N.Q. (Nabila Qayyum), R.R., N.Q. (Naila Qayyum) and S.I.; visualization, A.M., N.Q. (Nabila Qayyum), R.R., N.Q. (Naila Qayyum) and S.I.; supervision, S.I.; project administration A.M., and S.I. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

During the preparation of this manuscript, the author(s) used Gemini 2.5 Flash, for the purposes of generating “Graphical Abstract”. All authors have consented to the acknowledgement. The authors have reviewed and edited the output and take full responsibility for the content of this publication.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AIArtificial Intelligence
CNNsConvolutional Neural Networks
DBTLDesign-Build-Test-Learn
DLDeep Learning
D-MPNNDirected Message Passing Neural Network
FMsFoundation Models
GANsGenerative Adversarial Networks
GEMsGenome-Scale Metabolic Models
GNNsGraph Neural Networks
Graph AttentionG-Attn
LLMsLarge Language Models
MINNMulti-omics Integrated Neural Network
MLMachine Learning
NLPNatural Language processing
R&DResearch and Development
RBSRibosomal Binding Site
RNNsRecurrent Neural Networks

References

  1. Krohs, U.; Bedau, M.A. Interdisciplinary Interconnections in Synthetic Biology. Biol. Theory 2013, 8, 313–317. [Google Scholar] [CrossRef][Green Version]
  2. Calvert, J. Synthetic Biology: Constructing Nature? Sociol. Rev. 2010, 58, 95–112. [Google Scholar] [CrossRef]
  3. Bensaude-Vincent, B. Building Multidisciplinary Research Fields: The Cases of Materials Science. In Nanotechnology and Synthetic Biology; Springer: Cham, Switzerland, 2016; pp. 45–60. [Google Scholar]
  4. Ilyas, S.; Lee, J.; Hwang, Y.; Choi, Y.; Lee, D. Deciphering Cathepsin K Inhibitors: A Combined QSAR, Docking and MD Simulation Based Machine Learning Approaches for Drug Design. SAR QSAR Environ. Res. 2024, 35, 771–793. [Google Scholar] [CrossRef]
  5. Ogliore, T. Heme Is Not Just for Impossible Burgers-The Source-WashU. Available online: https://source.washu.edu/2021/05/heme-is-not-just-for-impossible-burgers/ (accessed on 28 July 2025).
  6. Le Feuvre, R.A.; Scrutton, N.S. A Living Foundry for Synthetic Biological Materials: A Synthetic Biology Roadmap to New Advanced Materials. Synth. Syst. Biotechnol. 2018, 3, 105–112. [Google Scholar] [CrossRef]
  7. Shapira, P.; Kwon, S.; Youtie, J. Tracking the Emergence of Synthetic Biology. Scientometrics 2017, 112, 1439–1469. [Google Scholar] [CrossRef]
  8. Mao, N.; Aggarwal, N.; Poh, C.L.; Cho, B.K.; Kondo, A.; Liu, C.; Yew, W.S.; Chang, M.W. Future Trends in Synthetic Biology in Asia. Adv. Genet. 2021, 2, e10038. [Google Scholar] [CrossRef]
  9. Rai, K.; Wang, Y.; O’Connell, R.W.; Patel, A.B.; Bashor, C.J. Using Machine Learning to Enhance and Accelerate Synthetic Biology. Curr. Opin. Biomed. Eng. 2024, 31, 100553. [Google Scholar] [CrossRef]
  10. Jeon, S.; Sohn, Y.J.; Lee, H.; Park, J.Y.; Kim, D.; Lee, E.S.; Park, S.J. Recent Advances in the Design-Build-Test-Learn (DBTL) Cycle for Systems Metabolic Engineering of Corynebacterium glutamicum. J. Microbiol. 2025, 63, e2501021. [Google Scholar] [CrossRef] [PubMed]
  11. Ali, M.; Dewan, A.; Sahu, A.K.; Taye, M.M. Understanding of Machine Learning with Deep Learning: Architectures, Workflow, Applications and Future Directions. Computers 2023, 12, 91. [Google Scholar] [CrossRef]
  12. Goshisht, M.K. Machine Learning and Deep Learning in Synthetic Biology: Key Architectures, Applications, and Challenges. ACS Omega 2024, 9, 9921–9945. [Google Scholar] [CrossRef]
  13. Freemont, P.S. Synthetic Biology Industry: Data-Driven Design Is Creating New Opportunities in Biotechnology. Emerg. Top. Life Sci. 2019, 3, 651. [Google Scholar] [CrossRef] [PubMed]
  14. Lei, X.; Wang, X.; Chen, G.; Liang, C.; Li, Q.; Jiang, H.; Xiong, W. Combining Diffusion and Transformer Models for Enhanced Promoter Synthesis and Strength Prediction in Deep Learning. mSystems 2025, 10, e0018325. [Google Scholar] [CrossRef] [PubMed]
  15. Yan, Z.; Chu, W.; Sheng, Y.; Tang, K.; Wang, S.; Liu, Y.; Wong, W.-F. Integrating Deep Learning and Synthetic Biology: A Co-Design Approach for Enhancing Gene Expression via N-Terminal Coding Sequences. ACS Synth. Biol. 2024, 13, 2960–2968. [Google Scholar] [CrossRef]
  16. Lutz, I.D.; Wang, S.; Norn, C.; Courbet, A.; Borst, A.J.; Zhao, Y.T.; Dosey, A.; Cao, L.; Xu, J.; Leaf, E.M.; et al. Top-down Design of Protein Architectures with Reinforcement Learning. Science 2023, 380, 266–273. [Google Scholar] [CrossRef] [PubMed]
  17. Tang, T.; Fu, L.; Guo, E.; Zhang, Z.; Wang, Z.; Ma, C.; Zhang, Z.; Zhang, J.; Huang, J.; Si, T. Automation in Synthetic Biology Using Biological Foundries. Chin. Sci. Bull. 2021, 66, 300–309. [Google Scholar] [CrossRef]
  18. Radivojević, T.; Costello, Z.; Workman, K.; Garcia Martin, H. A Machine Learning Automated Recommendation Tool for Synthetic Biology. Nat. Commun. 2020, 11, 4879. [Google Scholar] [CrossRef]
  19. Pandi, A.; Diehl, C.; Yazdizadeh Kharrazi, A.; Scholz, S.A.; Bobkova, E.; Faure, L.; Nattermann, M.; Adam, D.; Chapin, N.; Foroughijabbari, Y.; et al. A Versatile Active Learning Workflow for Optimization of Genetic and Metabolic Networks. Nat. Commun. 2022, 13, 3876. [Google Scholar] [CrossRef]
  20. Arboleda-Garcia, A.; Stiebritz, M.; Boada, Y.; Picó, J.; Vignoni, A. DBTL Bioengineering Cycle for Part Characterization and Refactoring. IFAC-PapersOnLine 2024, 58, 7–12. [Google Scholar] [CrossRef]
  21. Helmy, M.; Smith, D.; Selvarajoo, K. Systems Biology Approaches Integrated with Artificial Intelligence for Optimized Metabolic Engineering. Metab. Eng. Commun. 2020, 11, e00149, Erratum in Metab. Eng. Commun. 2021, 13, e00186. [Google Scholar] [CrossRef]
  22. Groff-Vindman, C.S.; Trump, B.D.; Cummings, C.L.; Smith, M.; Titus, A.J.; Oye, K.; Prado, V.; Turmus, E.; Linkov, I. The Convergence of AI and Synthetic Biology: The Looming Deluge. npj Biomed. Innov. 2025, 2, 20. [Google Scholar] [CrossRef]
  23. Sieow, B.F.-L.; De Sotto, R.; Seet, Z.R.D.; Hwang, I.Y.; Chang, M.W. Synthetic Biology Meets Machine Learning. Methods Mol. Biol. 2023, 2553, 21–39. [Google Scholar] [CrossRef]
  24. Cuperlovic-Culf, M.; Nguyen-Tran, T.; Bennett, S.A.L. Machine Learning and Hybrid Methods for Metabolic Pathway Modeling. Methods Mol. Biol. 2023, 2553, 417–439. [Google Scholar] [CrossRef]
  25. Cuperlovic-Culf, M. Machine Learning Methods for Analysis of Metabolic Data and Metabolic Pathway Modeling. Metabolites 2018, 8, 4. [Google Scholar] [CrossRef]
  26. Azrag, M.A.K.; Kadir, T.A.A.; Kabir, M.N.; Jaber, A.S. Large-Scale Kinetic Parameters Estimation of Metabolic Model of Escherichia Coli. Int. J. Mach. Learn. Comput. 2019, 9, 160–167. [Google Scholar] [CrossRef]
  27. Li, Y.; Wu, F.-X.; Ngom, A. A Review on Machine Learning Principles for Multi-View Biological Data Integration. Brief. Bioinform. 2018, 19, 325–340. [Google Scholar] [CrossRef] [PubMed]
  28. Wu, Z.; Johnston, K.E.; Arnold, F.H.; Yang, K.K. Protein Sequence Design with Deep Generative Models. Curr. Opin. Chem. Biol. 2021, 65, 18–27. [Google Scholar] [CrossRef] [PubMed]
  29. Dahiya, G.S.; Bakken, T.I.; Fages-Lartaud, M.; Lale, R. From Context to Code: Rational De Novo DNA Design and Predicting Cross-Species DNA Functionality Using Deep Learning Transformer Models. bioRxiv 2023. [Google Scholar] [CrossRef]
  30. Tazza, G.; Moro, F.; Ruggeri, D.; Teusink, B.; Vidács, L. MINN: A Metabolic-Informed Neural Network for Integrating Omics Data into Genome-Scale Metabolic Modeling. Comput. Struct. Biotechnol. J. 2025, 27, 3609–3617. [Google Scholar] [CrossRef] [PubMed]
  31. Hasibi, R.; Michoel, T.; Oyarzún, D.A. Integration of Graph Neural Networks and Genome-Scale Metabolic Models for Predicting Gene Essentiality. npj Syst. Biol. Appl. 2024, 10, 24. [Google Scholar] [CrossRef]
  32. Han, X.; Jia, M.; Chang, Y.; Li, Y.; Wu, S. Directed Message Passing Neural Network (D-MPNN) with Graph Edge Attention (GEA) for Property Prediction of Biofuel-Relevant Species. Energy AI 2022, 10, 100201. [Google Scholar] [CrossRef]
  33. Treloar, N.J.; Fedorec, A.J.H.; Ingalls, B.; Barnes, C.P. Deep Reinforcement Learning for the Control of Microbial Co-Cultures in Bioreactors. PLoS Comput. Biol. 2020, 16, e1007783. [Google Scholar] [CrossRef] [PubMed]
  34. Patron, N.J. Beyond Natural: Synthetic Expansions of Botanical Form and Function. New Phytol. 2020, 227, 295–310. [Google Scholar] [CrossRef]
  35. Palacios, S.; Collins, J.J.; Del Vecchio, D. Machine Learning for Synthetic Gene Circuit Engineering. Curr. Opin. Biotechnol. 2025, 92, 103263. [Google Scholar] [CrossRef]
  36. Müller, M.M.; Arndt, K.M.; Hoffmann, S.A. Genetic Circuits in Synthetic Biology: Broadening the Toolbox of Regulatory Devices. Front. Synth. Biol. 2025, 3, 1548572. [Google Scholar] [CrossRef]
  37. Prasad, K.; Cross, R.S.; Jenkins, M.R. Synthetic Biology, Genetic Circuits and Machine Learning: A New Age of Cancer Therapy. Mol. Oncol. 2023, 17, 946–949. [Google Scholar] [CrossRef]
  38. Chang, J.; Ye, J.C. Bidirectional Generation of Structure and Properties Through a Single Molecular Foundation Model. Nat. Commun. 2023, 15, 2323. [Google Scholar] [CrossRef] [PubMed]
  39. Loeffler, H.H.; He, J.; Tibo, A.; Janet, J.P.; Voronov, A.; Mervin, L.H.; Engkvist, O. Reinvent 4: Modern AI–Driven Generative Molecule Design. J. Cheminform. 2024, 16, 20. [Google Scholar] [CrossRef] [PubMed]
  40. Feng, H.; Wu, L.; Zhao, B.; Huff, C.; Zhang, J.; Wu, J.; Lin, L.; Wei, P.; Wu, C. Benchmarking DNA Foundation Models for Genomic Sequence Classification. bioRxiv 2024, bioRxiv:2024.08.16.608288. [Google Scholar] [CrossRef]
  41. Thomas, N.; Belanger, D.; Xu, C.; Lee, H.; Hirano, K.; Iwai, K.; Polic, V.; Nyberg, K.D.; Hoff, K.G.; Frenz, L.; et al. Engineering Highly Active Nuclease Enzymes with Machine Learning and High-Throughput Screening. Cell Syst. 2025, 16, 101236. [Google Scholar] [CrossRef]
  42. Kouba, P.; Kohout, P.; Haddadi, F.; Bushuiev, A.; Samusevich, R.; Sedlar, J.; Damborsky, J.; Pluskal, T.; Sivic, J.; Mazurenko, S. Machine Learning-Guided Protein Engineering. ACS Catal. 2023, 13, 13863–13895. [Google Scholar] [CrossRef]
  43. Liu, S.-H.; Bai, L.; Wang, X.-D.; Wang, Q.-Q.; Wang, D.-X.; Bornscheuer, U.T.; Ao, Y.-F. Machine Learning-Guided Protein Engineering to Improve the Catalytic Activity of Transaminases under Neutral PH Conditions. Org. Chem. Front. 2025, 12, 4788–4793. [Google Scholar] [CrossRef]
  44. van Lent, P.; Schmitz, J.; Abeel, T. Simulated Design-Build-Test-Learn Cycles for Consistent Comparison of Machine Learning Methods in Metabolic Engineering. ACS Synth. Biol. 2023, 12, 2588–2599. [Google Scholar] [CrossRef]
  45. Zhou, K.; Ng, W.; Cortés-Peña, Y.; Wang, X. Increasing Metabolic Pathway Flux by Using Machine Learning Models. Curr. Opin. Biotechnol. 2020, 66, 179–185. [Google Scholar] [CrossRef]
  46. Cheng, Y.; Bi, X.; Xu, Y.; Liu, Y.; Li, J.; Du, G.; Lv, X.; Liu, L. Machine Learning for Metabolic Pathway Optimization: A Review. Comput. Struct. Biotechnol. J. 2023, 21, 2381–2393. [Google Scholar] [CrossRef]
  47. Tabane, E.; Mnkandla, E.; Wang, Z. Optimizing DNA Sequence Classification via a Deep Learning Hybrid of LSTM and CNN Architecture. Appl. Sci. 2025, 15, 8225. [Google Scholar] [CrossRef]
  48. Li, J.; Wu, P.; Cao, Z.; Huang, G.; Lu, Z.; Yan, J.; Zhang, H.; Zhou, Y.; Liu, R.; Chen, H.; et al. Machine Learning-Based Prediction Models to Guide the Selection of Cas9 Variants for Efficient Gene Editing. Cell Rep. 2024, 43, 113765. [Google Scholar] [CrossRef] [PubMed]
  49. Einarson, D.; Frisk, F.; Klonowska, K.; Sennersten, C. A Machine Learning Approach to Simulation of Mallard Movements. Appl. Sci. 2024, 14, 1280. [Google Scholar] [CrossRef]
  50. Okoro, O.V.; Hippolyte, D.E.C.; Nie, L.; Karimi, K.; Denayer, J.F.M.; Shavandi, A. Machine Learning-Based Predictive Modeling and Optimization: Artificial Neural Network-Genetic Algorithm vs. Response Surface Methodology for Black Soldier Fly (Hermetia Illucens) Farm Waste Fermentation. Biochem. Eng. J. 2025, 218, 109685. [Google Scholar] [CrossRef]
  51. Smith, G.D.; Ching, W.H.; Cornejo-Páramo, P.; Wong, E.S. Decoding Enhancer Complexity with Machine Learning and High-Throughput Discovery. Genome Biol. 2023, 24, 116. [Google Scholar] [CrossRef]
  52. Alcantar, M.A.; English, M.A.; Valeri, J.A.; Collins, J.J. A High-Throughput Synthetic Biology Approach for Studying Combinatorial Chromatin-Based Transcriptional Regulation. Mol. Cell 2024, 84, 2382–2396.e9. [Google Scholar] [CrossRef]
  53. Rapp, J.T.; Bremer, B.J.; Romero, P.A. Self-Driving Laboratories to Autonomously Navigate the Protein Fitness Landscape. Nat. Chem. Eng. 2024, 1, 97–107. [Google Scholar] [CrossRef]
  54. Martin, H.G.; Radivojevic, T.; Zucker, J.; Bouchard, K.; Sustarich, J.; Peisert, S.; Arnold, D.; Hillson, N.; Babnigg, G.; Marti, J.M.; et al. Perspectives for Self-Driving Labs in Synthetic Biology. Curr. Opin. Biotechnol. 2023, 79, 102881. [Google Scholar] [CrossRef]
  55. Nguyen, E.; Poli, M.; Durrant, M.G.; Kang, B.; Katrekar, D.; Li, D.B.; Bartie, L.J.; Thomas, A.W.; King, S.H.; Brixi, G.; et al. Sequence Modeling and Design from Molecular to Genome Scale with Evo. Science 2024, 386, eado9336. [Google Scholar] [CrossRef]
  56. Tang, B.; Ewalt, J.; Ng, H.-L. Generative AI Models for Drug Discovery. In Topics in Medicinal Chemistry; Springer: Berlin/Heidelberg, Germany, 2021; Volume 37, pp. 221–243. [Google Scholar]
  57. Seo, E.; Choi, Y.-N.; Shin, Y.R.; Kim, D.; Lee, J.W. Design of Synthetic Promoters for Cyanobacteria with Generative Deep-Learning Model. Nucleic Acids Res. 2023, 51, 7071–7082. [Google Scholar] [CrossRef]
  58. Kitano, S.; Lin, C.; Foo, J.L.; Chang, M.W. Synthetic Biology: Learning the Way toward High-Precision Biological Design. PLoS Biol. 2023, 21, e3002116. [Google Scholar] [CrossRef] [PubMed]
  59. Park, J.H.; Han, R.; Jang, J.; Kim, J.; Paik, J.; Heo, J.; Lee, Y.; Park, J. MetaboGNN: Predicting Liver Metabolic Stability with Graph Neural Networks and Cross-Species Data. J. Cheminform. 2025, 17, 140. [Google Scholar] [CrossRef] [PubMed]
  60. Zhou, J.; Cui, G.; Hu, S.; Zhang, Z.; Yang, C.; Liu, Z.; Wang, L.; Li, C.; Sun, M. Graph Neural Networks: A Review of Methods and Applications. AI Open 2020, 1, 57–81, Erratum in AI Open 2024. [Google Scholar] [CrossRef]
  61. Zhang, X.M.; Liang, L.; Liu, L.; Tang, M.J. Graph Neural Networks and Their Current Applications in Bioinformatics. Front. Genet. 2021, 12, 690049. [Google Scholar] [CrossRef]
  62. Yasmeen, E.; Wang, J.; Riaz, M.; Zhang, L.; Zuo, K. Designing Artificial Synthetic Promoters for Accurate, Smart, and Versatile Gene Expression in Plants. Plant Commun. 2023, 4, 100558. [Google Scholar] [CrossRef]
  63. Orzechowski, P.; Moore, J.H. Generative and Reproducible Benchmarks for Comprehensive Evaluation of Machine Learning Classifiers. Sci. Adv. 2022, 8, 4747. [Google Scholar] [CrossRef] [PubMed]
  64. Hassija, V.; Chamola, V.; Mahapatra, A.; Singal, A.; Goel, D.; Huang, K.; Scardapane, S.; Spinelli, I.; Mahmud, M.; Hussain, A. Interpreting Black-Box Models: A Review on Explainable Artificial Intelligence. Cogn. Comput. 2024, 16, 45–74. [Google Scholar] [CrossRef]
  65. Adadi, A.; Berrada, M. Peeking Inside the Black-Box: A Survey on Explainable Artificial Intelligence (XAI). IEEE Access 2018, 6, 52138–52160. [Google Scholar] [CrossRef]
  66. Semmelrock, H.; Ross-Hellauer, T.; Kopeinik, S.; Theiler, D.; Haberl, A.; Thalmann, S.; Kowald, D. Reproducibility in Machine-learning-based Research: Overview, Barriers, and Drivers. AI Mag. 2025, 46, e70002. [Google Scholar] [CrossRef]
  67. Semmelrock, H.; Kopeinik, S.; Theiler, D.; Ross-Hellauer, T.; Kowald, D. Reproducibility in Machine Learning-Driven Research. AI Mag. 2023, 46, e70002. [Google Scholar]
  68. Kapoor, S.; Narayanan, A. Leakage and the Reproducibility Crisis in Machine-Learning-Based Science. Patterns 2023, 4, 100804. [Google Scholar] [CrossRef]
  69. Devi, N.B.; Sen, S.; Pakshirajan, K. Artificial Intelligence in Synthetic Biology. In Artificial Intelligence and Biological Sciences; CRC Press: Boca Raton, FL, USA, 2025; pp. 278–300. [Google Scholar]
  70. Adewumi, O.O.; Oladele, E.O.; Gbenle, O.A.; Taiwo, I.A. Artificial Intelligence in Bioinformatics: Cutting-Edge Techniques and Future Prospects. Bull. Nat. Appl. Sci. 2025, 1, 79–91. [Google Scholar] [CrossRef]
  71. Schmitt, F.-J.; Golüke, M.; Budisa, N. Bridging the Gap: Enhancing Science Communication in Synthetic Biology with Specific Teaching Modules, School Laboratories, Performance and Theater. Front. Synth. Biol. 2024, 2, 1337860. [Google Scholar] [CrossRef]
  72. Brooks, S.M.; Alper, H.S. Applications, Challenges, and Needs for Employing Synthetic Biology beyond the Lab. Nat. Commun. 2021, 12, 1390. [Google Scholar] [CrossRef] [PubMed]
  73. Davies, J.A. Real-World Synthetic Biology: Is It Founded on an Engineering Approach, and Should It Be? Life 2019, 9, 6. [Google Scholar] [CrossRef] [PubMed]
  74. Kelley, N.J.; Whelan, D.J.; Kerr, E.; Apel, A.; Beliveau, R.; Scanlon, R. Engineering Biology to Address Global Problems: Synthetic Biology Markets, Needs, and Applications. Ind. Biotechnol. 2014, 10, 140–149. [Google Scholar] [CrossRef]
  75. Garner, K.L. Principles of Synthetic Biology. Essays Biochem. 2021, 65, 791–811. [Google Scholar] [CrossRef]
  76. Ching, T.; Himmelstein, D.S.; Beaulieu-Jones, B.K.; Kalinin, A.A.; Do, B.T.; Way, G.P.; Ferrero, E.; Agapow, P.-M.; Zietz, M.; Hoffman, M.M.; et al. Opportunities and Obstacles for Deep Learning in Biology and Medicine. J. R. Soc. Interface 2018, 15, 20170387. [Google Scholar] [CrossRef]
  77. Beardall, W.A.V.; Stan, G.-B.; Dunlop, M.J. Deep Learning Concepts and Applications for Synthetic Biology. GEN Biotechnol. 2022, 1, 360–371. [Google Scholar] [CrossRef]
  78. Kuiken, T. Artificial Intelligence in The Biological Sciences: Uses, Safety, Security, and Oversight; R47849; Congressional Research Service (CRS) Reports & Issue Briefs: Washington, DC, USA, 2023. [Google Scholar]
  79. Mirchandani, I.; Khandhediya, Y.; Chauhan, K. Review on Advancement of AI in Synthetic Biology. Methods Mol. Biol. 2025, 2952, 483–490. [Google Scholar] [CrossRef]
  80. Hynek, N. Synthetic Biology/AI Convergence (SynBioAI): Security Threats in Frontier Science and Regulatory Challenges. AI Soc. 2025, 1–18. [Google Scholar] [CrossRef]
  81. Iram, A.; Dong, Y.; Ignea, C. Synthetic Biology Advances towards a Bio-Based Society in the Era of Artificial Intelligence. Curr. Opin. Biotechnol. 2024, 87, 103143. [Google Scholar] [CrossRef] [PubMed]
  82. Ameyaw, S.A.; Boateng, J.; Afari, D.A. Artificial Intelligence Algorithms for Designing Synthetic Biological Systems and Predicting Their Behavior. OSF Prepr. 2025. [Google Scholar] [CrossRef]
  83. Gaeta, A.; Zulkower, V.; Stracquadanio, G. Design and Assembly of DNA Molecules Using Multi-Objective Optimization. Synth. Biol. 2021, 6, ysab026. [Google Scholar] [CrossRef] [PubMed]
  84. Tian, Y.; Si, L.; Zhang, X.; Cheng, R.; He, C.; Tan, K.C.; Jin, Y. Evolutionary Large-Scale Multi-Objective Optimization: A Survey. ACM Comput. Surv. 2022, 54, 1–34. [Google Scholar] [CrossRef]
  85. Collins, T.K.; Zakirov, A.; Brown, J.A.; Houghten, S. Single-Objective and Multi-Objective Genetic Algorithms for Compression of Biological Networks. In Proceedings of the 2017 IEEE Conference on Computational Intelligence in Bioinformatics and Computational Biology, CIBCB, Manchester, UK, 23–25 August 2017. [Google Scholar] [CrossRef]
  86. Patane, A.; Santoro, A.; Costanza, J.; Carapezza, G.; Nicosia, G. Pareto Optimal Design for Synthetic Biology. IEEE Trans. Biomed. Circuits Syst. 2015, 9, 555–571. [Google Scholar] [CrossRef]
  87. Boada, Y.; Reynoso-Meza, G.; Picó, J.; Vignoni, A. Multi-Objective Optimization Framework to Obtain Model-Based Guidelines for Tuning Biological Synthetic Devices: An Adaptive Network Case. BMC Syst. Biol. 2016, 10, 27. [Google Scholar] [CrossRef] [PubMed]
  88. Braun, M.; Fernau, S.; Dabrock, P. (Re-)Designing Nature? An Overview and Outlook on the Ethical and Societal Challenges in Synthetic Biology. Adv. Biosyst. 2019, 3, 1800326. [Google Scholar] [CrossRef] [PubMed]
  89. Stephenson, A.; Lastra, L.; Nguyen, B.; Chen, Y.-J.; Nivala, J.; Ceze, L.; Strauss, K. Physical Laboratory Automation in Synthetic Biology. ACS Synth. Biol. 2023, 12, 3156–3169. [Google Scholar] [CrossRef] [PubMed]
  90. Anh Tuan, D.; Uyen, P.V.N.; Masak, J. Hybrid Quorum Sensing and Machine Learning Systems for Adaptive Synthetic Biology: Toward Autonomous Gene Regulation and Precision Therapies. Preprints 2024, 2024101551. [Google Scholar] [CrossRef]
Figure 1. Comprehensive analysis and challenges of AI/ML/DL integration in synthetic biology workflows. (A) Biological complexity challenges addressed by AI including non-linear interactions, high-dimensional systems, and interconnected biosystems. (B) Transformation of the traditional DBTL cycle through AI integration, reducing timeline from ~10 years to ~6 months for commercial molecule development. (C) Synergistic relationship between synthetic biology and AI/ML/DL showing mutual reinforcement through data generation and design optimization. (D) AI as a “predictive powerhouse” overcoming human limitations in complex biological design spaces.
Figure 1. Comprehensive analysis and challenges of AI/ML/DL integration in synthetic biology workflows. (A) Biological complexity challenges addressed by AI including non-linear interactions, high-dimensional systems, and interconnected biosystems. (B) Transformation of the traditional DBTL cycle through AI integration, reducing timeline from ~10 years to ~6 months for commercial molecule development. (C) Synergistic relationship between synthetic biology and AI/ML/DL showing mutual reinforcement through data generation and design optimization. (D) AI as a “predictive powerhouse” overcoming human limitations in complex biological design spaces.
Synbio 03 00017 g001
Figure 2. The integrated AI-driven design-build-tests-learn (DBTL) cycle. The synthetic biology workflow is accelerated and made autonomous through the synergistic integration of specialized AI/ML/DL algorithms at each stage. The cycle is closed by a data-rich AI-driven feedback loop flowing from learn to design, enabling continuous self-optimization.
Figure 2. The integrated AI-driven design-build-tests-learn (DBTL) cycle. The synthetic biology workflow is accelerated and made autonomous through the synergistic integration of specialized AI/ML/DL algorithms at each stage. The cycle is closed by a data-rich AI-driven feedback loop flowing from learn to design, enabling continuous self-optimization.
Synbio 03 00017 g002
Figure 3. The broad landscape of synthetic biology applications. Synthetic biology acts as a foundational technology, generating products like optimized pathways, engineered cells, and novel biomolecules that radiate impact across the major industrial and societal sectors.
Figure 3. The broad landscape of synthetic biology applications. Synthetic biology acts as a foundational technology, generating products like optimized pathways, engineered cells, and novel biomolecules that radiate impact across the major industrial and societal sectors.
Synbio 03 00017 g003
Figure 4. Critical analysis of ML applications in synthetic biology: current state, challenges, and future directions. A comprehensive overview of ML/DL applications in synthetic biology. (A) Current workflow from data collection to real-world implementation showing major bottlenecks. (B) Comparative performance analysis of different ML architectures across synthetic biology tasks. (C) Major challenges limiting practical adoption including dataset limitations, interpretability issues, and regulatory compliance. (D) Proposed solutions and future directions for addressing current limitations.
Figure 4. Critical analysis of ML applications in synthetic biology: current state, challenges, and future directions. A comprehensive overview of ML/DL applications in synthetic biology. (A) Current workflow from data collection to real-world implementation showing major bottlenecks. (B) Comparative performance analysis of different ML architectures across synthetic biology tasks. (C) Major challenges limiting practical adoption including dataset limitations, interpretability issues, and regulatory compliance. (D) Proposed solutions and future directions for addressing current limitations.
Synbio 03 00017 g004
Table 1. AI/ML/DL contributions across the synthetic biology DBTL cycle.
Table 1. AI/ML/DL contributions across the synthetic biology DBTL cycle.
DBTL StageTraditional Approach/ChallengeAI/ML/DL ContributionImpact/BenefitRepresentative Studies
DesignAd hoc, intuition-driven methods:
Designers often rely on trial-and-error due to the inherent complexity and non-linear interactions within biological systems. This approach is time-consuming and lacks predictability in output behavior. Difficulty arises in predicting the precise behavior of novel genetic circuits or pathways.
Predictive modeling & generative AI:
Predictive modeling of genetic circuits, metabolic pathways, and cellular responses allows in silico validation before experimental work.
De novo sequence generation for proteins, RNA, and DNA elements (e.g., promoters, RBS using generative models (e.g., GANs, VAEs) that adhere to biological constraints.
Optimization of DNA sequences for expression, stability, or specific functionalities.
AI-driven recommendations for optimal experimental designs and component selections, exploring vast design spaces far beyond human capacity.
Accelerated design & enhanced success rates:
Significantly reduced design iterations, minimizing costly and time-consuming experimental cycles.
Higher probability of successful outcomes due to in silico validation and optimized component selection.
Enhanced precision and predictability in desired biological functions.
Exploration and discovery of non-intuitive or counter-intuitive designs that would be inaccessible through traditional, heuristic approaches.
Expedited time-to-market for novel biological products and therapies.
DL for gene/promoter design: DL models (transformers, CNNs) accurately predict gene expression and optimize sequences in silico. De novo rational design; transformer models for synthetic promoter design [14]. CNNs to design N-terminal coding sequences, resulting in increase in protein expression [15]. RL for protein design [16]
BuildManual and labor-intensive assembly:
The physical construction of genetic constructs is often performed manually, making it labor-intensive, prone to human error, and limited in throughput. This constraint severely restricts the diversity of genetic libraries that can be synthesized and screened.
Automated synthesis & robotic assembly:
Automated DNA synthesis and cloning platforms leveraging high-throughput robotics for constructing gene circuits and pathways.
Robotic assembly of genetic constructs (e.g., Golden Gate, Gibson Assembly) minimizes manual intervention and improves consistency.
AI-driven scheduling and optimization of robotic workflows to maximize efficiency and minimize material waste.
Integration with liquid handling systems for precise reagent dispensing and reaction setup.
Increased throughput & reduced costs:
Substantially faster assembly times for complex genetic constructs.
Significant reduction in labor costs and human-induced errors, leading to higher reliability.
Vastly increased throughput and library diversity, enabling the exploration of a much broader design space.
Improved consistency and reproducibility of built components, leading to more reliable experimental outcomes.
ML-guided design–build automation: Bayesian optimization and RL integrate with robotic platforms to automatically determine and execute the most efficient DNA construction protocols. Minimizes human error and labor; accelerates construction time for large genetic libraries by intelligently optimizing assembly protocols [17].
TestLaborious and low-throughput screening:
Experimental testing involves laborious screening processes and often manual analysis of microscopy or flow cytometry data. This creates a limited capacity for handling large datasets and extracting meaningful insights efficiently.
High-throughput data analysis & automated phenotyping:
High-throughput data analysis (e.g., genomics, transcriptomics, proteomics, metabolomics data) using advanced ML/DL algorithms for pattern recognition and feature extraction.
Automated image and microscopy data analysis for rapid quantification of cellular phenotypes, morphology, and protein localization.
Rapid identification of subtle patterns and anomalies in large, complex datasets that are imperceptible to human analysis.
Real-time monitoring and anomaly detection in bioreactors and cell cultures.
Rapid insights & efficient evaluation:
Accelerated insights from experimental results, enabling faster decision-making and iteration.
Massively increased experimental throughput, allowing for parallel testing of numerous designs.
More efficient and accurate evaluation of phenotypes and functional outputs.
Early identification of successful or problematic designs, minimizing resources spent on unproductive pathways.
Discovery of unexpected relationships within experimental data.
DL-enabled high-throughput analysis: CNN and GNN models analyze massive datasets (microscopy, multi-omics) to rapidly quantify phenotypes, classify cell states, and extract performance metrics [18]. Accelerates data-to-insight; enables rapid and objective evaluation of vast numbers of constructs; uncovers subtle or novel phenotypes otherwise missed by manual inspection.
LearnLimited model-based refinement:
Iterative refinement is often based on limited empirical models, leading to unpredictable performance and a high number of design-build-test cycles. The lack of a robust feedback loop slows down the overall discovery process.
AL and RL for iterative improvement:
AL strategies guide subsequent experiments by selecting the most informative data points to optimize model training and reduce experimental burden.
RL frameworks enable autonomous optimization of experimental parameters and processes based on observed outcomes.
AI-driven feedback loops that automatically analyze test data, update design models, and recommend parameters for the next iteration.
Identification of underlying trends, optimal parameters, and governing rules from vast experimental data, facilitating true biological understanding.
Accelerated discovery & optimized cycles:
Significantly accelerated discovery cycles by intelligently guiding subsequent experiments.
Smarter, data-driven feedback loops replace manual guesswork.
Optimized experimental conditions and resource allocation.
Minimized trial-and-error, leading to more efficient resource utilization and faster convergence to desired designs.
Continuous improvement of synthetic biology platforms and processes through automated knowledge extraction.
AI-driven closed-loop optimization: RL and AL systematically retrain models on new results to propose the most informative subsequent experiment, iteratively refining designs [19]. AL accelerates metabolic pathway optimization. Minimizes trial-and-error; guides the search space intelligently, leading to a faster convergence on optimal designs; accelerates scientific discovery by automating the hypothesis generation step.
DBTL integrationNot applicable as a traditional stage/challenge, but the lack of true autonomy is the underlying issue.End-to-end AI frameworks coupling all DBTL stages: Comprehensive closed-loop AI pipelines orchestrate and connect the automated design, build, and test steps, creating self-optimizing bioengineering systemsAchieves autonomous science: Realizes the full potential of high-throughput bioengineering; Dramatically reduces cycle time and cost for industrial-scale pathway and product development.AI integrated DBTL platforms for autonomous genetic circuit optimization [20]. This dramatically reduces cycle time and cost for industrial-scale pathway and product development.
Where RL (Reinforcement Learning); AL (Active Learning); RBS (Ribosome Binding Sites).
Table 2. Prerequisites for effective AI integration in synthetic biology.
Table 2. Prerequisites for effective AI integration in synthetic biology.
Challenge AreaDescriptionImplicationsCurrent Gaps/Needs
Interpretable AI modelsAI models must provide mechanistic, explainable predictions that align with biological reasoning.Enhances scientific understanding and regulatory transparency.Lack of models with biologically meaningful interpretability; limited tools for hypothesis generation.
Data & protocol standardizationHarmonization of datasets, experimental protocols, and performance metrics across the field.Enables reproducibility, benchmarking, and fair evaluation of AI models.Fragmented datasets, inconsistent formats, lack of agreed-upon evaluation metrics.
Multi-objective optimizationIntegration of frameworks that balance biological performance with manufacturability, safety, and cost-effectiveness.Supports practical translation of AI designs into real-world applications.Limited optimization tools that handle diverse biological and engineering constraints simultaneously.
Uncertainty quantificationDevelopment of AI tools that assess the confidence or reliability of predictions within their operational domain.Increases user trust and identifies when models are making extrapolative or risky suggestions.Absence of robust, domain-specific uncertainty quantification methods in synthetic biology contexts.
Regulatory frameworksEstablish policies and standards that can assess and approve AI-designed biological products without compromising safety.Critical for commercialization, public trust, and long-term viability of AI-enabled products.Regulatory ambiguity; lack of precedent and guidelines for AI-derived biological constructs.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Manan, A.; Qayyum, N.; Ramachandran, R.; Qayyum, N.; Ilyas, S. Digital to Biological Translation: How the Algorithmic Data-Driven Design Reshapes Synthetic Biology. SynBio 2025, 3, 17. https://doi.org/10.3390/synbio3040017

AMA Style

Manan A, Qayyum N, Ramachandran R, Qayyum N, Ilyas S. Digital to Biological Translation: How the Algorithmic Data-Driven Design Reshapes Synthetic Biology. SynBio. 2025; 3(4):17. https://doi.org/10.3390/synbio3040017

Chicago/Turabian Style

Manan, Abdul, Nabila Qayyum, Rajath Ramachandran, Naila Qayyum, and Sidra Ilyas. 2025. "Digital to Biological Translation: How the Algorithmic Data-Driven Design Reshapes Synthetic Biology" SynBio 3, no. 4: 17. https://doi.org/10.3390/synbio3040017

APA Style

Manan, A., Qayyum, N., Ramachandran, R., Qayyum, N., & Ilyas, S. (2025). Digital to Biological Translation: How the Algorithmic Data-Driven Design Reshapes Synthetic Biology. SynBio, 3(4), 17. https://doi.org/10.3390/synbio3040017

Article Metrics

Back to TopTop