Appearance
This report is written by MaltSci based on the latest literature and research findings
What are the applications of base editing?
Abstract
Base editing represents a revolutionary advancement in genome editing technology, allowing for precise alterations of DNA sequences at the single-base level without the introduction of double-strand breaks (DSBs) or the need for donor DNA templates. This innovative approach, developed by integrating the CRISPR/Cas9 system with deaminase enzymes, facilitates direct nucleotide conversions, making it a safer and more efficient alternative to traditional gene-editing methods. The significance of base editing is highlighted by its diverse applications across genetic research, therapeutic development, and agricultural enhancement. In medicine, base editing has shown promise in gene therapy, particularly for genetic disorders caused by point mutations, with the potential to correct pathogenic mutations associated with conditions like sickle cell disease and beta-thalassemia. Additionally, it plays a crucial role in cancer research by allowing for the modeling of cancer-driving mutations and the development of targeted therapies. In agriculture, base editing has been employed to enhance crop traits and improve disease resistance, showcasing its versatility beyond human health. Despite its exciting prospects, challenges such as off-target effects, delivery mechanisms, and ethical considerations surrounding human germline editing remain. This review synthesizes current literature and case studies to illuminate the transformative potential of base editing, emphasizing both its promise and the challenges that lie ahead. The ongoing refinement of base editing technologies and innovations in delivery methods are expected to further broaden its applications, paving the way for advancements in genetic medicine and sustainable agricultural practices.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Mechanisms of Base Editing
- 2.1 Overview of Base Editing Technology
- 2.2 Comparison with Other Genome Editing Techniques
- 3 Applications in Medicine
- 3.1 Gene Therapy for Genetic Disorders
- 3.2 Cancer Research and Treatment
- 4 Applications in Agriculture
- 4.1 Crop Improvement and Disease Resistance
- 4.2 Livestock Genetic Enhancement
- 5 Ethical Considerations and Challenges
- 5.1 Ethical Implications of Genome Editing
- 5.2 Regulatory Challenges and Public Perception
- 6 Future Directions and Innovations
- 6.1 Advancements in Base Editing Techniques
- 6.2 Potential for Broader Applications
- 7 Conclusion
1 Introduction
Base editing represents a transformative advancement in genome editing technology, allowing for precise alterations of DNA sequences at the single-base level without the introduction of double-strand breaks (DSBs) or the need for donor DNA templates. This innovative approach, developed through the integration of the CRISPR/Cas9 system with deaminase enzymes, facilitates the direct conversion of nucleotides, thereby offering a safer and more efficient alternative to traditional gene-editing methods. The significance of base editing is underscored by its potential applications across various domains, including genetic research, therapeutic development, and agricultural enhancement, making it a focal point of contemporary biomedical investigations[1][2].
The research landscape surrounding base editing has expanded rapidly since its inception, with a plethora of studies elucidating its mechanisms and applications. Base editors, particularly cytosine base editors (CBEs) and adenine base editors (ABEs), have been optimized to improve editing efficiency, specificity, and delivery methods, which are critical for their therapeutic use[3][4]. Recent advancements have demonstrated the ability of base editing to correct pathogenic mutations associated with various genetic disorders, highlighting its promise in gene therapy[2][5]. Moreover, the technology's applicability extends beyond human health; it has been effectively employed in agricultural settings to enhance crop traits and improve disease resistance, further emphasizing its versatility[6][7].
Despite the exciting prospects that base editing presents, several challenges and ethical considerations remain. Issues related to off-target effects, delivery mechanisms, and public perception of genome editing technologies pose significant hurdles that must be addressed to facilitate the successful translation of base editing into clinical and agricultural practices[4][8]. Additionally, the ethical implications of editing human germline cells and the potential for unintended consequences in ecosystems require careful consideration and regulatory frameworks[7].
This review aims to provide a comprehensive overview of the applications of base editing, organized into several key sections. The first section will delve into the mechanisms of base editing, offering an overview of the technology and a comparison with other genome editing techniques. Following this, we will explore the applications of base editing in medicine, focusing on gene therapy for genetic disorders and its role in cancer research and treatment. The subsequent section will address the agricultural applications of base editing, including crop improvement and livestock genetic enhancement. Ethical considerations and challenges will also be discussed, providing insight into the societal implications of genome editing technologies. Finally, we will outline future directions and innovations in base editing, emphasizing the advancements in techniques and the potential for broader applications across various fields.
Through a synthesis of current literature and case studies, this review seeks to highlight the transformative potential of base editing, elucidating both its promise and the challenges that lie ahead in its implementation. By doing so, we aim to foster a deeper understanding of this groundbreaking technology and its implications for the future of biomedical research and applications.
2 Mechanisms of Base Editing
2.1 Overview of Base Editing Technology
Base editing technology represents a significant advancement in the field of genome editing, primarily due to its precision and efficiency in altering genetic sequences without inducing double-strand breaks (DSBs). This innovative approach combines the CRISPR/Cas system with deaminases to facilitate targeted single-base substitutions in DNA or RNA, making it a valuable tool in various biomedical applications.
The applications of base editing are extensive and encompass several key areas:
Gene Therapy: Base editing holds great promise for treating genetic disorders caused by point mutations, which account for a substantial proportion of genetic diseases. The ability to precisely correct single nucleotide variants offers new avenues for therapeutic interventions in conditions such as sickle cell disease and beta-thalassemia, where specific mutations can be targeted and repaired without the risk of DSBs that traditional CRISPR/Cas9 methods entail (Huang et al., 2023) [2].
Disease Modeling: In the realm of research, base editing allows for the creation of accurate models of human diseases. By introducing specific mutations into the genomes of model organisms, researchers can study the effects of these mutations on disease progression and test potential therapeutic strategies (Liang et al., 2023) [1].
Directed Protein Evolution: Base editors can be employed to engineer proteins with enhanced functionalities. By facilitating precise amino acid substitutions through targeted nucleotide changes, scientists can optimize proteins for various applications, including therapeutic proteins and enzymes used in industrial processes (Yang et al., 2021) [9].
Genetic Lineage Tracing: Base editing technology enables the tagging of specific cells or tissues within organisms, allowing researchers to trace cellular lineages and understand developmental processes or disease progression in vivo. This application is crucial for elucidating the mechanisms underlying complex biological systems (Zheng et al., 2025) [7].
Crop Improvement: Beyond human health, base editing has been applied in agricultural biotechnology to enhance crop traits. By precisely modifying genes associated with desirable characteristics such as yield, disease resistance, and stress tolerance, base editing can contribute to sustainable agricultural practices (Yang et al., 2024) [6].
Biomedicine and Cancer Research: The potential of base editing in oncology is particularly noteworthy, as it can be used to investigate the role of specific mutations in cancer development and to develop targeted therapies aimed at correcting these mutations. The technology’s ability to deliver precise edits could lead to breakthroughs in cancer treatment strategies (Huang et al., 2023) [2].
The underlying mechanism of base editing involves the fusion of a catalytically impaired Cas9 protein with a deaminase enzyme. This configuration allows for the direct conversion of one nucleotide to another, such as converting cytosine to thymine or adenine to guanine, thereby achieving targeted genetic modifications without the complexities and potential hazards associated with traditional genome editing techniques (Molla & Yang, 2019) [10].
In summary, base editing technology is a transformative tool with a wide array of applications in gene therapy, disease modeling, protein engineering, agricultural enhancement, and cancer research. Its precision and efficiency in executing targeted genetic modifications make it a cornerstone of modern genetic engineering and a promising avenue for future therapeutic innovations.
2.2 Comparison with Other Genome Editing Techniques
Base editing technology, a groundbreaking advancement derived from the CRISPR/Cas9 system, facilitates precise and efficient modifications of genomic DNA. This technology allows for targeted alterations of individual nucleotides without inducing double-strand breaks (DSBs), thereby circumventing some of the challenges associated with traditional gene editing methods. The applications of base editing span various fields, including gene therapy, crop improvement, and functional genomics, among others.
In the realm of gene therapy, base editing has emerged as a promising tool for the treatment of genetic disorders. Its ability to make precise single-nucleotide modifications positions it as an effective strategy for correcting point mutations that underlie many genetic diseases. Recent reviews have highlighted the potential of cytosine base editors (CBEs) and adenine base editors (ABEs) in the therapeutic landscape, focusing on their principles, delivery methods, and applications in treating genetic diseases [5]. Furthermore, advancements in base editing have demonstrated its utility in correcting common single-base substitutions, thereby enhancing the prospects for developing targeted therapies [11].
In agricultural biotechnology, base editing is increasingly being applied to improve crop traits, such as disease resistance, yield, and nutritional quality. For instance, researchers have successfully employed base editing to facilitate genetic improvements in various crops, including fruit crops like tomatoes and other economically significant plants [12]. The technology allows for the introduction of desirable traits without the drawbacks associated with transgenic approaches, thereby providing a more acceptable method for genetic enhancement in agriculture [13].
Additionally, base editing has been instrumental in constructing model organisms for functional genomics and disease modeling. In zebrafish, for example, base editors have been utilized to study gene function and the genetic basis of diseases, leveraging the organism's rapid development and genetic similarity to humans [14]. This capability is vital for advancing our understanding of complex biological processes and developing therapeutic strategies.
When comparing base editing to other genome editing techniques, such as traditional CRISPR/Cas9 methods, TALENs, and ZFNs, several advantages become apparent. Base editing does not rely on DSBs, which significantly reduces the likelihood of unwanted insertions and deletions (indels) and off-target effects [10]. Moreover, base editing systems can achieve high editing efficiencies in various contexts, including cultured mammalian cells and in vivo applications, which is crucial for therapeutic efficacy [15].
The specific design of base editors allows for targeted base conversions—C•G to T•A or A•T to G•C—enabling the correction of the majority of pathogenic point mutations [8]. This precision sets base editing apart from traditional methods that often result in broader genomic alterations. Moreover, recent developments have expanded the targeting scope of base editors, allowing for greater flexibility in selecting target sites [16].
In summary, base editing is a versatile and powerful tool with applications spanning gene therapy, agriculture, and functional genomics. Its unique mechanism of action, characterized by precise nucleotide modifications without DSBs, provides significant advantages over traditional genome editing techniques, positioning it as a key player in the future of genetic engineering and therapeutic development.
3 Applications in Medicine
3.1 Gene Therapy for Genetic Disorders
Base editing, a revolutionary advancement in genome editing technology, has significant applications in the field of medicine, particularly in gene therapy for genetic disorders. This technology allows for precise modifications of DNA sequences without the need for double-strand breaks, thereby reducing the risk of unintended genetic alterations. The primary applications of base editing in gene therapy can be categorized into several key areas.
Firstly, base editing is particularly promising for treating rare monogenic disorders, which are caused by mutations in single genes with an incidence rate of less than 0.5%. Traditional therapeutic strategies often focus on managing symptoms rather than addressing the underlying genetic cause. In contrast, base editing offers a "one-and-done" therapeutic approach by enabling the precise correction of pathogenic mutations. For instance, cytosine base editors (CBE) and adenine base editors (ABE) can theoretically correct approximately 95% of pathogenic transition mutations cataloged in ClinVar, thus presenting a substantial opportunity for effective treatment of these disorders (Cabré-Romans & Cuella-Martin 2025) [17].
Moreover, base editing has shown considerable promise in treating genetic diseases characterized by point mutations, which are responsible for a significant portion of human genetic disorders. A recent study indicates that 62% of pathogenic single nucleotide variants (SNVs) can be effectively corrected using base editing techniques, with specific capabilities to address G>A and T>C transitions as well as C>T and A>G transitions (Dadush et al. 2024) [18]. This high correction rate underscores the potential of base editing to serve as a viable therapeutic strategy for a wide range of genetic diseases.
In addition to its applications in rare genetic disorders, base editing is also being explored for its potential in treating common diseases. For example, point mutations account for about 58% of disease-causing genetic mutations, and the ability of base editors to achieve single-base substitutions with precision positions them as critical tools in oncology and other therapeutic areas (Huang et al. 2023) [2]. This capability allows researchers to target specific mutations that contribute to disease progression, thereby offering new avenues for intervention.
Furthermore, the development of advanced delivery methods for base editors is enhancing their therapeutic potential. The use of adeno-associated viruses (AAV) for the delivery of base editing systems has emerged as a leading approach, enabling efficient and targeted gene editing in vivo (Kantor et al. 2020) [13]. This progress is crucial for translating base editing technology from laboratory research to clinical applications.
Base editing is also being investigated for its applications in the treatment of blood disorders, where traditional nuclease-based genome editing strategies have faced challenges due to the risks associated with double-strand breaks. The ability of base editing to introduce point mutations without generating such breaks presents a safer alternative for correcting mutations associated with these conditions (Antoniou et al. 2021) [19].
In summary, the applications of base editing in gene therapy for genetic disorders are vast and varied. By enabling precise corrections of genetic mutations, base editing holds the potential to transform the therapeutic landscape for both rare and common genetic diseases, offering hope for effective treatments that address the root causes of these conditions. As research progresses, the continued refinement of base editing technologies and delivery methods will further enhance their applicability and efficacy in clinical settings.
3.2 Cancer Research and Treatment
Base editing, a revolutionary genome editing technology derived from the CRISPR/Cas9 system, has emerged as a promising tool in cancer research and treatment due to its ability to introduce precise single-base substitutions in DNA without causing double-strand breaks. This precision is particularly advantageous given that point mutations are implicated in approximately 58% of disease-causing genetic mutations in humans, and they play a critical role in tumorigenesis. As a result, base editing offers significant potential for both modeling cancer and developing therapeutic strategies.
One of the primary applications of base editing in cancer research is the modeling of cancer-driving mutations. For instance, base editing has been employed to create complex tumor models in adult stem cell-derived organoids, demonstrating its efficacy in modeling mutations such as those in the CTNNB1 gene in hepatocyte organoids and PTEN in endometrial organoids. These models have not only facilitated the study of tumorigenicity but have also provided insights into drug sensitivity and the underlying mechanisms of tumorigenesis [20].
In addition to modeling, base editing has been utilized for therapeutic editing. The technology allows researchers to correct mutations associated with various cancers, thereby restoring normal gene function. For example, a base editing platform has been developed to investigate the functional impact of cancer hotspot mutations, such as TP53 mutations. This platform has shown that correcting these mutations can reinstate tumor-suppressive transcriptional programs, suggesting a pathway for targeted therapies [21].
Base editing's advantages extend to its application in therapeutic contexts, particularly in cancer immunotherapy. It has been reported that base editors can facilitate multiplex gene modifications in primary human T cells, which are essential for developing allogeneic CAR-T therapies. This approach minimizes the risk of unintended genomic alterations and enhances the expansion of modified T cells, showcasing base editing's potential to improve the efficacy of adoptive cellular therapies [22].
Furthermore, base editing has been explored for its ability to correct mutations in cancer-driving genes, thereby offering a novel strategy for investigating tumor biology and therapeutic vulnerabilities. The continuous improvement in base editing technology, focusing on editing efficiency and specificity, further enhances its applicability in clinical settings [2].
Overall, the applications of base editing in cancer research and treatment are vast and evolving. From modeling cancer mutations to developing targeted therapies and improving immunotherapy strategies, base editing represents a critical advancement in the fight against cancer, with the potential to significantly impact patient outcomes and our understanding of tumor biology.
4 Applications in Agriculture
4.1 Crop Improvement and Disease Resistance
Base editing technology has emerged as a transformative tool in agriculture, particularly for crop improvement and disease resistance. This innovative genome editing approach enables precise alterations of single nucleotides in plant genomes without the need for double-strand breaks or donor repair templates, which are common limitations in traditional CRISPR/Cas9 methods. The following sections outline the key applications of base editing in agriculture, emphasizing its role in crop improvement and disease resistance.
One of the primary applications of base editing is in enhancing crop traits such as stress resistance, yield, and quality. For instance, base editing has been utilized to create novel germplasm with improved agronomic traits, including herbicide resistance, disease resistance, and grain quality in staple crops like rice, wheat, and maize [23]. This precision allows for the targeted introduction of beneficial traits that can significantly improve crop performance under various environmental conditions.
In the context of disease resistance, base editing provides a powerful method for generating crops that can withstand various pathogens, including bacteria, fungi, and viruses. The technology facilitates the development of plants with enhanced resistance to diseases, which is crucial for maintaining food security in the face of increasing agricultural challenges [24]. The ability to make specific, targeted mutations that confer resistance to diseases can accelerate the breeding process and reduce the reliance on chemical pesticides, promoting more sustainable agricultural practices [25].
Moreover, base editing is being explored for its potential in creating climate-resilient crops. As climate change poses significant threats to agricultural productivity, the ability to enhance traits such as drought tolerance, heat resistance, and salinity tolerance through precise genetic modifications is increasingly vital [26]. For example, by targeting key regulatory genes involved in stress responses, researchers can improve crops' resilience to abiotic stresses, thus contributing to more sustainable agricultural systems [23].
The technology also enables the generation of specific alleles associated with desirable traits through artificial evolution techniques. Recent studies have demonstrated the use of base editing to create a diverse range of genetic variants in model plants like Arabidopsis, which can then be utilized to enhance crop traits in economically important species [27]. This approach allows for the rapid screening and selection of beneficial alleles, expediting the breeding process for crop improvement [6].
In summary, base editing holds significant promise for agricultural applications, particularly in crop improvement and disease resistance. By enabling precise genetic modifications, this technology facilitates the development of crops with enhanced traits, improving agricultural productivity and sustainability in the face of environmental challenges. The ongoing research and advancements in base editing technologies are expected to further broaden their applications in crop genetic improvement, ultimately contributing to global food security.
4.2 Livestock Genetic Enhancement
Base editing has emerged as a revolutionary tool in the field of genetic enhancement for livestock, offering precise methods for altering genetic traits without the complications associated with traditional genome editing techniques. This technology enables the direct modification of specific DNA bases, facilitating the development of livestock with improved traits such as disease resistance, enhanced growth rates, and better reproductive performance.
One significant application of base editing in livestock is the optimization of genetic traits through targeted modifications. For instance, Xu et al. (2022) reported the use of the base editor BE4max in chicken somatic cells, where the efficiency of this editor was improved by 10.4% ± 4.6 after optimization for chicken-specific conditions. Furthermore, by inhibiting the expression of the uracil DNA glycosylase-related gene methyl binding domain protein 4 (MBD4) using siRNA, they enhanced editing efficiency by an additional 4.43% ± 1.4. These advancements indicate that base editing can significantly impact poultry breeding studies by facilitating the introduction of desired genetic traits more efficiently than traditional methods[28].
In sheep, Zhou et al. (2020) demonstrated the potential of adenine base editors (ABE) to create specific point mutations that influence economically important traits. Their study achieved a mutation efficiency of 75% in newborn lambs for the FecBB mutation, which is known to affect fecundity. This highlights the capability of base editing to enhance reproductive traits in livestock, which is crucial for improving productivity in agricultural settings[29].
Additionally, base editing can be employed to generate disease-resistant livestock. Yuan et al. (2024) discussed the broader implications of genome editing, including base editing, in improving livestock health and production efficiency. They emphasized that genome editing technologies could help in developing livestock that are less susceptible to diseases, thereby reducing the need for antibiotics and enhancing overall animal welfare[30].
Moreover, base editing has shown promise in generating livestock models for research purposes. The ability to create specific genetic modifications allows researchers to study the genetic basis of traits and diseases in livestock, which can lead to better management practices and breeding strategies[6].
In summary, base editing offers a powerful approach to enhance livestock genetics by enabling precise modifications that can improve traits such as growth, reproduction, and disease resistance. The advancements in base editing technologies continue to provide new opportunities for genetic enhancement in livestock, with significant implications for agricultural productivity and sustainability.
5 Ethical Considerations and Challenges
5.1 Ethical Implications of Genome Editing
Base editing is a groundbreaking genome editing technology that has found applications across various fields, including basic research, medicine, agriculture, and biotechnology. Its unique ability to make precise single-nucleotide changes without inducing double-strand breaks (DSBs) has opened up numerous avenues for therapeutic and experimental use.
In the realm of medicine, base editing has significant potential for treating genetic diseases caused by point mutations. It enables the correction of specific genetic defects, such as those responsible for blood disorders, without the complications associated with traditional genome editing techniques. For instance, adenine base editors (ABEs) and cytosine base editors (CBEs) can introduce precise A•T-to-G•C or C•G-to-T•A conversions, which can address a vast majority of pathogenic single-nucleotide polymorphisms (SNPs) that lead to genetic disorders (Antoniou et al., 2021; Huang et al., 2021) [15][19].
In agriculture, base editing has been employed to enhance crop traits by enabling targeted modifications that improve disease resistance, nutritional value, and yield. The technology allows for the precise alteration of single bases in plant genomes, facilitating the development of crops with desirable traits such as blight resistance and improved fruit quality. This application is particularly advantageous because it can be conducted without the need for transgenic approaches, thus addressing regulatory concerns associated with genetically modified organisms (Azameti & Dauda, 2021; Mishra et al., 2020) [25][31].
Base editing has also been applied in various model organisms, including zebrafish and mice, to study gene function and disease mechanisms. The technology has proven to be an effective tool for generating animal models that mimic human diseases, thus advancing our understanding of genetic conditions and the development of potential therapies (Liu et al., 2019; Schatoff et al., 2019) [32][33].
Despite the promise of base editing, ethical considerations and challenges remain a significant concern. The ability to make precise edits in the genome raises questions about the potential for unintended consequences, such as off-target effects, which could lead to unforeseen genetic alterations. Studies have indicated that base editors can cause substantial off-target editing, independent of guide RNA, primarily due to the inherent promiscuity of the deaminase enzymes used in these systems (Park & Beal, 2019) [34].
Furthermore, the implications of applying base editing in humans, especially in germline editing, pose ethical dilemmas regarding consent, the potential for "designer babies," and the long-term impacts on human genetics. These concerns necessitate a robust ethical framework and public discourse to navigate the implications of such powerful technologies responsibly.
In summary, base editing presents a multitude of applications in medicine, agriculture, and research, yet it also brings forth critical ethical considerations that must be addressed to ensure its responsible use in society.
5.2 Regulatory Challenges and Public Perception
Base editing, a groundbreaking gene-editing technology that combines the CRISPR/Cas system with deaminases, has emerged as a highly precise tool for making targeted modifications in DNA and RNA. Its applications span various fields, particularly in biomedicine, where it holds significant promise for advancing therapeutic strategies.
In the realm of biomedicine, base editing is employed for gene function investigation, directed protein evolution, genetic lineage tracing, disease modeling, and gene therapy. Specifically, it enables precise single-base substitutions without generating double-strand breaks (DSBs), which can lead to severe genomic damage, thereby offering a more secure alternative to conventional gene-editing methods like CRISPR/Cas9 (Liang et al., 2023) [1]. The technology has led to the development of over 100 optimized base editors, enhancing their efficiency, precision, specificity, and delivery capabilities in vivo, thereby significantly broadening their application potential (Liang et al., 2023) [1].
In therapeutic contexts, base editing has been utilized to address genetic disorders caused by point mutations, which account for a significant proportion of disease-causing mutations in humans. For instance, the technology has shown potential in treating blood disorders by enabling precise edits in hematopoietic stem cells (Zeng et al., 2020) [35]. Moreover, advancements in base editing have made it possible to correct mutations associated with conditions like sickle cell disease and β-thalassemia, demonstrating its applicability in clinical settings (Zeng et al., 2020) [35].
Base editing also plays a role in the development of agricultural crops and microbial metabolic engineering, showcasing its versatility beyond human health applications. The technology has been successfully applied to improve crop traits and enhance the metabolic pathways of industrial microorganisms, thus contributing to advancements in synthetic biology (Zheng et al., 2025) [7].
Despite its transformative potential, the deployment of base editing raises ethical considerations and challenges. The precision of base editing minimizes unintended genetic alterations, yet concerns regarding off-target effects persist, necessitating thorough evaluations of its safety and efficacy before clinical application (Huang et al., 2023) [2]. Furthermore, the public perception of gene editing technologies is often influenced by ethical debates surrounding genetic modification, particularly in human embryos and germline editing. Addressing these concerns through transparent communication and ethical guidelines is crucial for fostering public trust and acceptance of base editing technologies.
Regulatory challenges also play a significant role in the advancement of base editing applications. As the technology evolves, regulatory frameworks must adapt to ensure the safety and efficacy of base editing therapies. This includes establishing clear guidelines for clinical trials, ethical standards for genetic modifications, and monitoring long-term effects in treated individuals (Carlaw et al., 2020) [36].
In conclusion, base editing represents a promising frontier in genetic engineering with diverse applications in biomedicine and agriculture. However, its ethical implications, regulatory hurdles, and public perception must be carefully navigated to maximize its benefits while minimizing potential risks.
6 Future Directions and Innovations
6.1 Advancements in Base Editing Techniques
Base editing, a sophisticated gene-editing technology derived from the CRISPR/Cas system, has emerged as a transformative tool in various fields of biomedicine, agriculture, and genetic research. Its ability to enable precise single-base substitutions in DNA or RNA without generating double-strand breaks (DSBs) or requiring donor DNA templates significantly enhances its application potential. The applications of base editing can be broadly categorized into several key areas:
Gene Therapy: Base editing has shown great promise in the treatment of genetic disorders, particularly those caused by point mutations. For instance, it has been utilized to correct mutations associated with blood disorders such as sickle cell disease and β-thalassemia by targeting specific genomic loci, like the BCL11A enhancer, leading to the induction of fetal hemoglobin (HbF) in erythroid progeny from patient-derived hematopoietic stem and progenitor cells (HSPCs) [35]. This highlights the potential of base editing as a viable therapeutic strategy to permanently remedy genetic diseases.
Disease Modeling: The technology is also instrumental in creating accurate disease models. By introducing specific mutations into model organisms, researchers can study the effects of these mutations on biological processes and disease progression, which aids in understanding the underlying mechanisms of various genetic disorders [1].
Directed Protein Evolution: Base editing facilitates the engineering of proteins by enabling targeted mutations that can enhance or modify protein function. This application is critical in the development of therapeutic proteins and antibodies, where precision is essential [1].
Genetic Lineage Tracing: The ability to mark specific cell lineages through precise genomic alterations allows for the tracking of cell fate and lineage in developmental biology and regenerative medicine [1].
Crop Improvement: In agriculture, base editing has been applied to enhance crop traits, such as disease resistance and yield. By precisely modifying single nucleotides in plant genomes, researchers can develop crops with improved characteristics without the risks associated with traditional genetic modification techniques [6].
Synthetic Biology: Base editing is being integrated into synthetic biology applications, where it can be used to construct novel genetic circuits and metabolic pathways, facilitating advancements in bioengineering and biotechnology [7].
As for future directions, advancements in base editing technology continue to focus on improving its efficiency, specificity, and delivery mechanisms. Innovations such as the development of novel base editor variants and optimization strategies are expected to broaden the scope of targetable sequences, including those previously deemed inaccessible due to protospacer adjacent motif (PAM) limitations [16]. Additionally, ongoing research aims to enhance the safety profile of base editing, addressing potential off-target effects and improving the fidelity of edits [8].
The continuous evolution of base editing technologies, including the introduction of prime editing, which expands the editing capabilities beyond point mutations to include insertions and deletions, signifies a promising horizon for therapeutic applications [11]. As these technologies advance, they are likely to play a crucial role in the future of genetic medicine, offering innovative solutions for the treatment of genetic disorders and the development of new biotechnological applications.
6.2 Potential for Broader Applications
Base editing technology, a sophisticated gene-editing tool derived from the CRISPR/Cas9 system, has shown remarkable potential across various applications in biomedicine. Its unique capability to facilitate precise single-base substitutions in DNA or RNA without inducing double-strand breaks (DSBs) positions it as a safer alternative to traditional editing methods. The applications of base editing can be categorized into several key areas:
Gene Function Investigation: Base editors enable researchers to study the roles of specific genes by inducing targeted mutations. This allows for a better understanding of gene function and regulation, which is crucial for unraveling the complexities of genetic pathways involved in various diseases (Liang et al. 2023) [1].
Disease Modeling: By creating specific mutations associated with human diseases, base editing serves as a powerful tool for generating accurate disease models. These models can be utilized to investigate disease mechanisms, evaluate potential therapeutic strategies, and assess drug efficacy (Huang et al. 2023) [2].
Gene Therapy: One of the most promising applications of base editing lies in its potential for gene therapy. It can be used to correct pathogenic mutations that lead to genetic disorders. For instance, base editing has been shown to effectively induce fetal hemoglobin expression in hematopoietic stem cells from patients with sickle cell disease and β-thalassemia, demonstrating its therapeutic promise (Zeng et al. 2020) [35].
Directed Protein Evolution: Base editing can be employed to create specific mutations in proteins, facilitating the directed evolution of enzymes and other proteins. This application is particularly relevant in biotechnology, where engineered proteins with enhanced functionalities can be developed for industrial applications (Liang et al. 2023) [1].
Genetic Lineage Tracing: By marking specific cells with unique genetic alterations, base editing allows researchers to trace the lineage of cells over time. This technique can provide insights into developmental biology and the dynamics of cell populations in various contexts (Liang et al. 2023) [1].
Therapeutic Applications in Cancer: Base editing holds promise for cancer treatment by targeting and correcting mutations that drive tumorigenesis. Its precision reduces the risk of off-target effects commonly associated with other genome editing technologies, making it a valuable tool in the development of oncotherapy strategies (Huang et al. 2023) [2].
Broadening Targeting Scope: Recent innovations have focused on expanding the targeting capabilities of base editors, allowing them to reach previously inaccessible genetic loci. This advancement enhances the versatility of base editing and broadens its applicability in various genetic contexts (Yu et al. 2022) [16].
In summary, base editing represents a transformative advancement in genetic engineering with applications spanning from fundamental research to therapeutic interventions. As the technology continues to evolve, its potential for broader applications in treating genetic disorders, advancing cancer therapies, and enhancing biotechnological processes will likely expand, paving the way for innovative solutions in medicine and beyond. The ongoing development of optimized base editors and novel delivery methods will further enhance the efficiency and specificity of this powerful tool, making it a focal point for future biomedical research and therapeutic strategies (Kaplan et al. 2025) [37].
7 Conclusion
Base editing has emerged as a transformative technology in genome editing, with significant applications across medicine, agriculture, and genetic research. Its ability to make precise single-base modifications without introducing double-strand breaks offers distinct advantages over traditional editing methods, such as CRISPR/Cas9. The primary findings indicate that base editing holds substantial promise in gene therapy, particularly for monogenic disorders, and in cancer research, where it can correct mutations associated with tumorigenesis. Furthermore, its applications in agriculture for crop improvement and livestock genetic enhancement highlight its versatility. However, the rapid advancement of this technology is accompanied by ethical considerations and regulatory challenges that must be addressed to ensure its responsible use. Future research directions will likely focus on enhancing the efficiency and specificity of base editing techniques, broadening their applications, and developing robust delivery methods. As the technology evolves, it is poised to play a pivotal role in shaping the future of genetic medicine and sustainable agriculture, ultimately contributing to improved health outcomes and food security.
References
- [1] Yanhui Liang;Fangbing Chen;Kepin Wang;Liangxue Lai. Base editors: development and applications in biomedicine.. Frontiers of medicine(IF=3.5). 2023. PMID:37434066. DOI: 10.1007/s11684-023-1013-y.
- [2] Lu Huang;Chao Yang;Yan Chen;Han Deng;Zhi Liao;Hongtao Xiao. CRISPR-Mediated Base Editing: Promises and Challenges for a Viable Oncotherapy Strategy.. Human gene therapy(IF=4.0). 2023. PMID:37276175. DOI: 10.1089/hum.2023.045.
- [3] Bei Yang;Li Yang;Jia Chen. Development and Application of Base Editors.. The CRISPR journal(IF=4.0). 2019. PMID:30998092. DOI: 10.1089/crispr.2019.0001.
- [4] Elizabeth M Porto;Alexis C Komor;Ian M Slaymaker;Gene W Yeo. Base editing: advances and therapeutic opportunities.. Nature reviews. Drug discovery(IF=101.8). 2020. PMID:33077937. DOI: 10.1038/s41573-020-0084-6.
- [5] Fang Xu;Caiyan Zheng;Weihui Xu;Shiyao Zhang;Shanshan Liu;Xiaopeng Chen;Kai Yao. Breaking genetic shackles: The advance of base editing in genetic disorder treatment.. Frontiers in pharmacology(IF=4.8). 2024. PMID:38510648. DOI: 10.3389/fphar.2024.1364135.
- [6] Xuefei Yang;Ping Zhu;Jinshan Gui. Advancements of CRISPR-Mediated Base Editing in Crops and Potential Applications in Populus.. International journal of molecular sciences(IF=4.9). 2024. PMID:39125884. DOI: 10.3390/ijms25158314.
- [7] Yi Zheng;Fei Du;Yiwen Hang;Wang Ma;Guang Yang;Na Wu;Xiaoman Sun. Recent Advances in Genome Base Editing Technology and Its Applications in Industrial Microorganism.. Biotechnology journal(IF=3.1). 2025. PMID:40490981. DOI: 10.1002/biot.70052.
- [8] Elizabeth M Porto;Alexis C Komor. In the business of base editors: Evolution from bench to bedside.. PLoS biology(IF=7.2). 2023. PMID:37043430. DOI: 10.1371/journal.pbio.3002071.
- [9] Li Yang;Jing Tang;Xuelei Ma;Yuan Lin;Guorong Ma;Minghai Shan;Libin Wang;Yanhui Yang. Progression and application of CRISPR-Cas genomic editors.. Methods (San Diego, Calif.)(IF=4.3). 2021. PMID:33774156. DOI: 10.1016/j.ymeth.2021.03.013.
- [10] Kutubuddin A Molla;Yinong Yang. CRISPR/Cas-Mediated Base Editing: Technical Considerations and Practical Applications.. Trends in biotechnology(IF=14.9). 2019. PMID:30995964. DOI: 10.1016/j.tibtech.2019.03.008.
- [11] Gregory A Newby;David R Liu. In vivo somatic cell base editing and prime editing.. Molecular therapy : the journal of the American Society of Gene Therapy(IF=12.0). 2021. PMID:34509669. DOI: 10.1016/j.ymthe.2021.09.002.
- [12] Junhui Zhou;Dongdong Li;Guoming Wang;Fuxi Wang;Merixia Kunjal;Dirk Joldersma;Zhongchi Liu. Application and future perspective of CRISPR/Cas9 genome editing in fruit crops.. Journal of integrative plant biology(IF=9.3). 2020. PMID:30791200. DOI: 10.1111/jipb.12793.
- [13] Ariel Kantor;Michelle E McClements;Robert E MacLaren. CRISPR-Cas9 DNA Base-Editing and Prime-Editing.. International journal of molecular sciences(IF=4.9). 2020. PMID:32872311. DOI: 10.3390/ijms21176240.
- [14] Yuwen Liu;Chao Li;Yiren Qiu;Sihong Chen;Yijun Luo;Donghua Xiong;Jun Zhao;Jianmin Ye;Xuegeng Wang;Wei Qin;Fang Liang. Base editors in zebrafish: a new era for functional genomics and disease modeling.. Frontiers in genome editing(IF=4.4). 2025. PMID:40469890. DOI: 10.3389/fgeed.2025.1598887.
- [15] Tony P Huang;Gregory A Newby;David R Liu. Precision genome editing using cytosine and adenine base editors in mammalian cells.. Nature protocols(IF=16.0). 2021. PMID:33462442. DOI: 10.1038/s41596-020-00450-9.
- [16] Si-Yue Yu;Alexandra Birkenshaw;Tyler Thomson;Tiffany Carlaw;Lin-Hua Zhang;Colin J D Ross. Increasing the Targeting Scope of CRISPR Base Editing System Beyond NGG.. The CRISPR journal(IF=4.0). 2022. PMID:35238621. DOI: 10.1089/crispr.2021.0109.
- [17] Júlia-Jié Cabré-Romans;Raquel Cuella-Martin. CRISPR-dependent base editing as a therapeutic strategy for rare monogenic disorders.. Frontiers in genome editing(IF=4.4). 2025. PMID:40242216. DOI: 10.3389/fgeed.2025.1553590.
- [18] Ariel Dadush;Rona Merdler-Rabinowicz;David Gorelik;Ariel Feiglin;Ilana Buchumenski;Lipika R Pal;Shay Ben-Aroya;Eytan Ruppin;Erez Y Levanon. DNA and RNA base editors can correct the majority of pathogenic single nucleotide variants.. NPJ genomic medicine(IF=4.8). 2024. PMID:38409211. DOI: 10.1038/s41525-024-00397-w.
- [19] Panagiotis Antoniou;Annarita Miccio;Mégane Brusson. Base and Prime Editing Technologies for Blood Disorders.. Frontiers in genome editing(IF=4.4). 2021. PMID:34713251. DOI: 10.3389/fgeed.2021.618406.
- [20] Maarten H Geurts;Shashank Gandhi;Matteo G Boretto;Ninouk Akkerman;Lucca L M Derks;Gijs van Son;Martina Celotti;Sarina Harshuk-Shabso;Flavia Peci;Harry Begthel;Delilah Hendriks;Paul Schürmann;Amanda Andersson-Rolf;Susana M Chuva de Sousa Lopes;Johan H van Es;Ruben van Boxtel;Hans Clevers. One-step generation of tumor models by base editor multiplexing in adult stem cell-derived organoids.. Nature communications(IF=15.7). 2023. PMID:37591832. DOI: 10.1038/s41467-023-40701-3.
- [21] Pascal Wang;Rituparno Sen;Frank Buchholz;Shady Sayed. A base editing platform for the correction of cancer driver mutations unmasks conserved p53 transcription programs.. Genome biology(IF=9.4). 2025. PMID:40696461. DOI: 10.1186/s13059-025-03667-7.
- [22] Beau R Webber;Cara-Lin Lonetree;Mitchell G Kluesner;Matthew J Johnson;Emily J Pomeroy;Miechaleen D Diers;Walker S Lahr;Garrett M Draper;Nicholas J Slipek;Branden A Smeester;Klaus N Lovendahl;Amber N McElroy;Wendy R Gordon;Mark J Osborn;Branden S Moriarity. Highly efficient multiplex human T cell engineering without double-strand breaks using Cas9 base editors.. Nature communications(IF=15.7). 2019. PMID:31745080. DOI: 10.1038/s41467-019-13007-6.
- [23] Hao Peng;Jiajun Li;Kehui Sun;Huali Tang;Weihong Huang;Xi Li;Surong Wang;Ke Ding;Zhiyang Han;Zhikun Li;Le Xu;Ke Wang. Advances and Applications of Plant Base Editing Technologies.. International journal of molecular sciences(IF=4.9). 2025. PMID:41096720. DOI: 10.3390/ijms26199452.
- [24] Kangquan Yin;Jin-Long Qiu. Genome editing for plant disease resistance: applications and perspectives.. Philosophical transactions of the Royal Society of London. Series B, Biological sciences(IF=4.7). 2019. PMID:30967029. DOI: 10.1098/rstb.2018.0322.
- [25] Rukmini Mishra;Raj Kumar Joshi;Kaijun Zhao. Base editing in crops: current advances, limitations and future implications.. Plant biotechnology journal(IF=10.5). 2020. PMID:31365173. DOI: 10.1111/pbi.13225.
- [26] R L Chavhan;S G Jaybhaye;V R Hinge;A S Deshmukh;U S Shaikh;P K Jadhav;U S Kadam;J C Hong. Emerging applications of gene editing technologies for the development of climate-resilient crops.. Frontiers in genome editing(IF=4.4). 2025. PMID:40129518. DOI: 10.3389/fgeed.2025.1524767.
- [27] Xiang Wang;Wenbo Pan;Chao Sun;Hong Yang;Zhentao Cheng;Fei Yan;Guojing Ma;Yun Shang;Rui Zhang;Caixia Gao;Lijing Liu;Huawei Zhang. Creating large-scale genetic diversity in Arabidopsis via base editing-mediated deep artificial evolution.. Genome biology(IF=9.4). 2024. PMID:39123212. DOI: 10.1186/s13059-024-03358-9.
- [28] Tianpeng Xu;Jing Zhong;Zhenwen Huang;Lintian Yu;Jitan Zheng;Long Xie;Lingling Sun;Xingting Liu;Yangqing Lu. Optimization of the base editor BE4max in chicken somatic cells.. Poultry science(IF=4.2). 2022. PMID:36240636. DOI: 10.1016/j.psj.2022.102174.
- [29] Shiwei Zhou;Yige Ding;Jiao Liu;Yao Liu;Xiaoe Zhao;Guanwei Li;Chenguang Zhang;Chao Li;Ying Wang;Peter Kalds;Yawei Gao;Bo Zong;Xiaoyu Huang;Shuhong Huang;Honghao Yu;Qifang Kou;Bjoern Petersen;Xingxu Huang;Xiaolong Wang;Baohua Ma;Yulin Chen. Highly efficient generation of sheep with a defined FecBB mutation via adenine base editing.. Genetics, selection, evolution : GSE(IF=3.1). 2020. PMID:32611306. DOI: 10.1186/s12711-020-00554-6.
- [30] Yu-Guo Yuan;Song-Zi Liu;Muhammad Farhab;Mei-Yun Lv;Ting Zhang;Shao-Xiao Cao. Genome editing: An insight into disease resistance, production efficiency, and biomedical applications in livestock.. Functional & integrative genomics(IF=3.1). 2024. PMID:38709433. DOI: 10.1007/s10142-024-01364-5.
- [31] Mawuli K Azameti;Wadzani Palnam Dauda. Base Editing in Plants: Applications, Challenges, and Future Prospects.. Frontiers in plant science(IF=4.8). 2021. PMID:34386023. DOI: 10.3389/fpls.2021.664997.
- [32] Kaili Liu;Cassidy Petree;Teresa Requena;Pratishtha Varshney;Gaurav K Varshney. Expanding the CRISPR Toolbox in Zebrafish for Studying Development and Disease.. Frontiers in cell and developmental biology(IF=4.3). 2019. PMID:30886848. DOI: 10.3389/fcell.2019.00013.
- [33] Emma M Schatoff;Maria Paz Zafra;Lukas E Dow. Base editing the mammalian genome.. Methods (San Diego, Calif.)(IF=4.3). 2019. PMID:30836137. DOI: 10.1016/j.ymeth.2019.02.022.
- [34] SeHee Park;Peter A Beal. Off-Target Editing by CRISPR-Guided DNA Base Editors.. Biochemistry(IF=3.0). 2019. PMID:31433621. DOI: 10.1021/acs.biochem.9b00573.
- [35] Jing Zeng;Yuxuan Wu;Chunyan Ren;Jasmine Bonanno;Anne H Shen;Devlin Shea;Jason M Gehrke;Kendell Clement;Kevin Luk;Qiuming Yao;Rachel Kim;Scot A Wolfe;John P Manis;Luca Pinello;J Keith Joung;Daniel E Bauer. Therapeutic base editing of human hematopoietic stem cells.. Nature medicine(IF=50.0). 2020. PMID:32284612. DOI: 10.1038/s41591-020-0790-y.
- [36] Tiffany M Carlaw;Lin-Hua Zhang;Colin J D Ross. CRISPR/Cas9 Editing: Sparking Discussion on Safety in Light of the Need for New Therapeutics.. Human gene therapy(IF=4.0). 2020. PMID:32586150. DOI: 10.1089/hum.2020.111.
- [37] Eleanor G Kaplan;Ryan J Steger;Spencer T Shah;Laura M Drepanos;Audrey L Griffith;Ganna Reint;John G Doench. Activity-based selection for enhanced base editor mutational scanning.. Nature genetics(IF=29.0). 2025. PMID:41087678. DOI: 10.1038/s41588-025-02366-0.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
Base Editing · Gene Therapy · Agricultural Enhancement
© 2025 MaltSci
