CRISPR: Diverse Applications, Advancements, and Impact
Received: 01-May-2025 / Manuscript No. science-25-172972 / Editor assigned: 05-May-2025 / PreQC No. science-25-172972 (PQ) / Reviewed: 19-May-2025 / QC No. science-25-172972 / Revised: 22-May-2025 / Manuscript No. science-25-172972 (R) / Accepted Date: 29-May-2025 / Published Date: 29-May-2025
Abstract
CRISPR-Cas9 gene editing has revolutionized biology and medicine, enabling precise genetic modifications. Initial in-human
trials demonstrated its therapeutic potential for systemic diseases. Research focuses on optimizing delivery systems like AAVs and
expanding the gene editing toolbox with prime and base editors for enhanced precision. CRISPR/Cas9 is transforming disease modeling, agriculture, and cancer therapy, while also providing rapid diagnostic tools. Ethical considerations, particularly for germline
editing, remain a key area of discussion. Overall, in vivo genome editing is a powerful tool accelerating functional genomics and
translating into diverse disease treatments.
Keywords
CRISPR-Cas9; Gene Editing; In Vivo Delivery; Prime Editing; Base Editors; Disease Modeling; Cancer Therapy; Diagnostics; Transthyretin Amyloidosis; AAV; Ethics
Introduction
The realm of gene editing has seen remarkable progress, exemplified by the first in-human gene editing trial utilizing CRISPR-Cas9 to address transthyretin amyloidosis. This significant study showcased that a single intravenous infusion of an LNP-encapsulated CRISPR-Cas9 component effectively reduced circulating transthyretin protein levels, thereby illustrating the profound potential of in vivo gene editing for managing systemic diseases. This achievement represents a pivotal milestone, accelerating the trajectory towards widespread therapeutic CRISPR applications [1].
Building on these foundational advances, ongoing research actively refines the delivery mechanisms for Cas9 components in in vivo gene editing. A key area of focus involves the development and meticulous characterization of adeno-associated viruses (AAVs) engineered to express Cas9. This critical work is paramount for enhancing the overall efficiency and safety profile of CRISPR-based gene therapies, with findings strongly supporting the adoption of these optimized AAV vectors for a broad spectrum of therapeutic uses [2].
CRISPR/Cas9-mediated gene editing is fundamentally transforming approaches to disease modeling and therapeutic development. The technology's unparalleled ability to introduce precise genetic alterations into diverse cell and animal models is dramatically speeding up the elucidation of disease mechanisms. This capability is not only deepening our understanding but also charting new paths for innovative therapeutic strategies across an extensive range of human health conditions [3].
Amidst these scientific breakthroughs, the ethical landscape surrounding CRISPR, particularly concerning human germline gene editing, demands careful consideration. Discussions revolve around the compelling potential to prevent inherited diseases balanced against serious concerns like unforeseen consequences, the societal implications of 'designer babies,' and ensuring equitable access to these powerful technologies. It is imperative that this discourse fosters thoughtful societal dialogue and leads to the establishment of robust regulatory frameworks to guide its responsible application [4].
The gene editing toolbox has also seen significant expansion with the introduction of advancements in prime editing. This innovative work details improved systems designed for targeted gene integration and highly efficient in vivo delivery. Such improvements are vital for correcting a much broader array of genetic mutations with heightened precision and notably fewer off-target effects compared to conventional CRISPR-Cas9, thereby unlocking entirely new therapeutic avenues for complex genetic disorders [5].
Beyond therapeutic applications, CRISPR technology has revolutionized diagnostic tools, transitioning from initial laboratory research to practical, real-world applications. Various formats, including SHERLOCK and DETECTR, leverage CRISPR's capabilities, offering high sensitivity, specificity, and rapid detection for a multitude of targets, encompassing infectious diseases, cancer biomarkers, and genetic conditions. The continuous evolution of these tools holds immense promise for future point-of-care diagnostics, making rapid and accurate testing more accessible [6].
In the agricultural sector, CRISPR-Cas9 has demonstrated its transformative potential for genome editing in plants. This technology allows for precise gene modifications that significantly enhance crucial crop traits such as increased yield, improved nutritional value, and bolstered disease resistance. By addressing these key challenges, CRISPR-Cas9 plays a vital role in advancing global food security and offers promising future directions for plant genetic engineering, ensuring sustainable agricultural practices [7].
Recent advancements in CRISPR/Cas9-based cancer therapy represent another frontier in medical innovation. Research explores diverse strategies, including directly targeting oncogenes, correcting dysfunctional tumor suppressor genes, and engineering immune cells to boost their anti-tumor activity. Additionally, the development of oncolytic viruses is being pursued. Despite these promising developments, significant challenges remain in clinical translation, particularly regarding delivery efficiency and minimizing off-target effects, necessitating continued research to overcome these hurdles [8].
Further enhancing precision in genome editing, comprehensive reviews detail the intricate mechanisms and diverse applications of both base editors and prime editors. These sophisticated CRISPR tools enable direct conversion of single base pairs or highly targeted insertions and deletions without requiring problematic double-strand breaks. This capability markedly expands the range of treatable genetic disorders and considerably augments fundamental research capabilities, pushing the boundaries of what is possible in genetic manipulation [9].
The overarching field of in vivo genome editing is recognized as an exceptionally powerful tool, serving both functional genomics research and advanced gene therapy. Extensive overviews highlight various effective delivery methods and a wide spectrum of applications. This direct editing within living organisms is rapidly accelerating our fundamental understanding of gene function and is simultaneously being translated into groundbreaking new treatments for a broad spectrum of human diseases, signifying a new era in biomedical science [10].
Description
The landscape of gene editing has been irrevocably altered by CRISPR-Cas9 technology, transitioning from a conceptual framework to a tangible clinical reality. A landmark achievement was the first in-human gene editing trial, which successfully applied CRISPR-Cas9 to treat transthyretin amyloidosis. This pivotal study demonstrated that a single intravenous infusion of an LNP-encapsulated CRISPR-Cas9 component could effectively reduce circulating transthyretin protein levels [1]. This not only showcased the immense potential of in vivo gene editing for systemic diseases but also marked a profound step toward widespread therapeutic CRISPR applications. Concurrently, a critical focus remains on developing robust and safe delivery systems for Cas9 components. Ongoing research is specifically centered on engineering optimized adeno-associated viruses (AAVs) to express Cas9, a development vital for enhancing both the efficiency and safety profiles of CRISPR-based gene therapies, supporting their broad utility [2].
Beyond its direct therapeutic applications, CRISPR/Cas9 has emerged as a transformative force in both disease modeling and therapeutic development. The technology’s unparalleled capability to create precise genetic alterations in diverse cell and animal models is dramatically accelerating the understanding of complex disease mechanisms [3]. This capability is instrumental in paving new avenues for innovative therapeutic strategies across a comprehensive spectrum of human health conditions. Furthermore, CRISPR-Cas9-mediated genome editing is revolutionizing the agricultural sector. Through precise gene modifications, this technology holds significant promise for enhancing crucial crop traits such, as increasing yield, improving nutritional value, and bolstering disease resistance, directly addressing pressing global food security challenges and charting future directions for sustainable plant genetic engineering [7].
The gene editing toolbox continues to expand with sophisticated advancements like prime editing, which significantly improves targeted gene integration and in vivo delivery. These refined systems are proving indispensable for correcting a wider array of genetic mutations with heightened precision and markedly fewer off-target effects compared to earlier CRISPR-Cas9 iterations, thereby unlocking new therapeutic possibilities for various genetic disorders [5]. This pursuit of precision is further amplified by base editors and prime editors, which allow for direct conversion of single base pairs or highly targeted insertions/deletions without the need for problematic double-strand breaks. These advanced tools not only broaden the scope of treatable genetic disorders but also substantially enhance fundamental research capabilities [9]. In the specific context of cancer therapy, CRISPR/Cas9 is actively being explored for multiple strategies, including targeting oncogenes, correcting tumor suppressor genes, and engineering immune cells to boost their anti-tumor activity. Despite these promising developments, significant clinical translation challenges persist, particularly concerning delivery efficiency and minimizing off-target effects, necessitating continued research to overcome these hurdles and bring these therapies to patients [8].
CRISPR technology also extends its utility to powerful diagnostic capabilities. The evolution of CRISPR-based diagnostic tools, exemplified by formats like SHERLOCK and DETECTR, underscores their high sensitivity, specificity, and rapid detection for a myriad of targets, including infectious diseases, cancer biomarkers, and genetic conditions. These advancements hold considerable promise for future point-of-care diagnostics, making rapid and accurate testing more widely accessible and efficient [6]. However, the profound societal implications of CRISPR, especially concerning human germline gene editing, necessitate a thorough and ongoing examination of complex ethical questions. The balance between the compelling potential to prevent inherited diseases and concerns about unintended consequences, the concept of 'designer babies,' and ensuring equitable access to these powerful technologies demands thoughtful societal dialogue and the establishment of robust, adaptable regulatory frameworks to guide their responsible and ethical application [4].
Collectively, the field of in vivo genome editing stands as an exceptionally powerful instrument, serving a dual role in both functional genomics research and advanced gene therapy. Extensive reviews and studies highlight the increasing sophistication of various delivery methods and the expansive spectrum of its applications. This capacity for direct genetic modification within living organisms is rapidly accelerating our fundamental comprehension of gene function and is simultaneously being translated into groundbreaking new treatments for a broad array of human diseases. This signifies a new era in biomedical science, where the precise manipulation of genetic code promises to address some of the most challenging medical conditions [10].
Conclusion
CRISPR-Cas9 gene editing has emerged as a groundbreaking technology with wide-ranging applications. The first in-human trial demonstrated its potential to treat systemic diseases like transthyretin amyloidosis by reducing protein levels through in vivo gene editing. This paves the way for therapeutic CRISPR applications by showing the efficacy of LNP-encapsulated CRISPR-Cas9 components. To enhance its delivery for in vivo gene editing, research focuses on developing optimized adeno-associated viruses (AAVs) engineered to express Cas9, improving both efficiency and safety in gene therapies. Beyond direct therapeutic interventions, CRISPR/Cas9 is revolutionizing disease modeling by enabling precise genetic alterations in various cellular and animal models, accelerating our understanding of disease mechanisms and fostering novel therapeutic strategies. The technology also extends to agriculture, where CRISPR-Cas9 is used to enhance crop traits such as yield, nutritional value, and disease resistance, contributing to global food security. Furthermore, advanced CRISPR tools like base editors and prime editors are expanding the precision of genome editing, allowing for targeted base pair conversions or insertions/deletions without double-strand breaks. This capability significantly broadens the scope of treatable genetic disorders. CRISPR's utility isn't limited to therapeutics and research; it has also led to the development of highly sensitive and specific diagnostic tools for infectious diseases, cancer biomarkers, and genetic conditions. However, the ethical implications, particularly concerning human germline gene editing, require thoughtful societal dialogue and robust regulatory frameworks. In the realm of cancer therapy, CRISPR/Cas9 is being explored for targeting oncogenes, correcting tumor suppressor genes, and engineering immune cells, despite challenges in delivery and off-target effects. Overall, in vivo genome editing continues to be a powerful tool for functional genomics research, accelerating our understanding of gene function and translating into new treatments for a wide array of diseases.
References
- Julian DG, David B, Marianna F (2021) CRISPR-Cas9-mediated gene editing in patients with transthyretin amyloidosis.N Engl J Med 385:493-502.
Indexed at, Google Scholar, Crossref
- Xiaohui Y, Haoming Z, Yue H (2020) Development of Cas9-expressing AAVs for in vivo gene editing applications.Mol Ther 28:2207-2216.
Indexed at, Google Scholar, Crossref
- Yu Z, Xin W, Yifan Z (2020) CRISPR/Cas9-mediated gene editing: A new avenue for disease modeling and therapeutic development.J Cell Mol Med 24:6695-6709.
Indexed at, Google Scholar, Crossref
- Christine B, Zeynep GK, Mark JC (2020) The ethics of CRISPR: New questions in human germline gene editing.Trends Mol Med 26:749-762.
Indexed at, Google Scholar, Crossref
- Andrew VA, Jonathan RK, Joshua AL (2021) An expanded toolbox for targeted gene integration and in vivo delivery of prime editing.Nat Biotechnol 39:1109-1118.
Indexed at, Google Scholar, Crossref
- Faheem K, Syed H, Syed S (2021) CRISPR-based diagnostics: a review of the state-of-the-art and future prospects.Clin Chem Lab Med 60:1-13.
Indexed at, Google Scholar, Crossref
- Lokender A, Manoj K, Anil KS (2020) CRISPR-Cas9 mediated genome editing in plants: current status and future prospects.Crit Rev Biotechnol 40:311-329.
Indexed at, Google Scholar, Crossref
- Xiaoyan W, Wei L, Jinying Z (2021) CRISPR/Cas9-based cancer therapy: recent advances and future challenges.Cancer Lett 512:106-118.
Indexed at, Google Scholar, Crossref
- Nicola MG, Alexis CS, Jeffrey EK (2020) Precision genome editing with base editors and prime editors.Mol Cell 79:4-18.
Indexed at, Google Scholar, Crossref
- Mohammad T, Feng Z, W. MD (2023) In vivo genome editing: a tool for functional genomics and gene therapy.Nat Rev Genet 24:709-728.
Citation: Monteiro JL (2025) CRISPR: Diverse Applications, Advancements, and Impact. Arch Sci 09: 277.
Copyright: © 2025 Javier L. Monteiro This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permitsunrestricted use, distribution and reproduction in any medium, provided the original author and source are credited.
Select your language of interest to view the total content in your interested language
Share This Article
Open Access Journals
Article Usage
- Total views: 121
- [From(publication date): 0-0 - Dec 15, 2025]
- Breakdown by view type
- HTML page views: 101
- PDF downloads: 20
