Previous Article in Journal
Genetic Association Between TMPRSS2 Polymorphisms and COVID-19 Severity in Brazilian Health Workers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CRISPR Treatments for AI-Designed Synthetic Viruses: Rapid Programmable Countermeasures for Emerging and Engineered Viruses

by
Douglas P. Gladue
* and
Alison O’Mahony
Seek Labs, 350 W 800 N Suite 220, Salt Lake City, UT 84103, USA
*
Author to whom correspondence should be addressed.
Viruses 2025, 17(12), 1588; https://doi.org/10.3390/v17121588
Submission received: 7 November 2025 / Revised: 4 December 2025 / Accepted: 4 December 2025 / Published: 5 December 2025
(This article belongs to the Section General Virology)

Abstract

The convergence of artificial intelligence and synthetic biology is innovating and accelerating the design of novel viral genomes, expanding both therapeutic opportunities and dual-use risk. This review articulates a countermeasure strategy for emerging and engineered viruses leveraging the programmable CRISPR modality. Building on mounting in vitro and in vivo evidence that Cas9 degrades DNA viruses (e.g., Orthopoxviruses, HSV-1, ASFV), while Cas13 targets RNA viral genomes (e.g., Influenza A, Dengue, RSV), both leading to reduced viremia, diminished disease burden, and alleviated symptoms. Here, we outline a rapid-response pipeline to position CRISPR-based countermeasures in translational and pandemic-response frameworks, linking real-time sequencing to AI-assisted gRNA selection and multiplexed cassette design to achieve viral targeting efficacy. To minimize resistance and off-target risk, we emphasize multi-gRNA cocktails, continuous genomic surveillance, and adaptive gRNA rotation. We also propose governance mechanisms, such as pre-cleared gRNA repositories, transparent design logs, standardized off-target/safety screening, and alignment with evolving nucleic-acid-synthesis screening frameworks to enable emergency deployment while preserving security. Furthermore, compressing the time from sequence to treatment and complementary to vaccines and small-molecule antivirals, CRISPR represents a technologically agile and strategically essential capability to combat both natural outbreaks and AI-enabled biothreats. Collectively, programmable CRISPR antivirals represent an auditable, rapidly adaptable foundation for next-generation biodefense preparedness.

1. Introduction

The accelerating convergence of artificial intelligence and synthetic biology (synbio) has expanded the capacity to design, generate, and potentially deploy novel viruses at unprecedented speed. Advances in these technologies offer substantial societal benefits but also carry inherent risks of misuse or unintended harm. For instance, Stanford researchers recently reported using AI models to engineer novel bacteriophage variants from existing lab strains. These variants incorporate safeguards to ensure specificity for bacteria (without harming humans, animals, or plants), serving as an antibiotic alternative and potentially saving lives in cases of drug-resistant bacterial infections. On the risk side, such advances could create viruses that bypass existing biosafety protocols, increasing the potential for bioterrorism or accidental pandemics. For instance, a recent Microsoft study demonstrated an AI approach to generate novel genetic sequences encoding harmful proteins, such as cell death inducers [1]. Of concern was the finding that even with software patches, about 3% of biothreat designs still evaded the screening software used by biotech DNA synthesis suppliers [1]. The potential dual-use capabilities heighten the urgent and critical need for equally adaptable and accelerated countermeasures.
This review proposes a rapid-response pipeline from AI-gRNA design to emergency deployment and outlines governance mechanisms, including pre-cleared gRNA repositories for new and emerging priority pathogens, along with streamlined AI-predicted emergency-use gRNAs for optimal off-target and safety potential. Integrating these elements provides an actionable path towards delivering safe, effective, and scalable programmable antivirals suitable for rapid outbreak response.

2. AI in Synthetic Virology and the Expanding Threat Landscape

Large-context genome language models have now generated viral genomes with coherent coding architectures and viable phenotypes, including host tropism and replication competence in bacteria [2]. While AI applications have thus far focused on bacteriophages, they mark a qualitative shift where generative models can design viral genomes beyond simple motif shuffling and reconstruction of particles, toward functional novelty and even new organisms. Although this capability expands the potential for new viral designs to help with therapeutic applications, it also opens the door to the development of novel biothreats. While AI lowers the design barrier, significant wet-lab constraints (assembly, rescue, and biosafety) still exist and would require more than computational and technical expertise to design a new lifeform or bioweapon that poses a threat to humans. Nonetheless, this development represents a future reality that may not be that far off and compels the reassessment of technical countermeasures that can be rapidly deployed. In the case of an emergency outbreak or pandemic situation, re-programming CRISPR antivirals with novel gRNA sequences precisely fills the niche where current antivirals simply do not fit. Historical countermeasures, including vaccines, small molecule antivirals, and monoclonal antibodies, require considerable time, effort, and expense to develop as deployable therapeutics. Moreover, only a small percentage of infectious diseases can be successfully managed with these conventional treatments. In contrast, CRISPR-based modalities represent a programmable, rapid-response approach with several advantages over traditional approaches (Table 1).
Generative AI did not create the potential for synthetic viral biothreats on its own; indeed, there have been several historical milestones that shaped today’s synthetic viral landscape (Table 2). In 2002, the first demonstration of de novo synthesis of poliovirus from oligonucleotides was achieved, although at the time, this involved the complicated, error-prone, and laborious stitching together of small pieces of the viral genome [3]. This was followed only one year later by a whole-genome assembly of bacteriophage φX174 also from synthetic oligos [4]. Since then, other labs have followed similar methods for re-constructing different viruses; however, in all cases, whole genome assembly involved a virus that already existed in various laboratories. In 2018, researchers assembled an extinct relative of a smallpox virus, called horsepox virus, simply from purchased DNA fragments. At the time, the researchers were heavily criticized for publishing a step-by-step procedure that could be used to reconstruct smallpox, which raised global biosecurity risks [5]. In 2005, researchers reconstructed and characterized the 1918 influenza A virus that caused the infamous Spanish Flu pandemic by recovering the sequence from historical tissue samples and using a plasmid-based system for all eight segments of influenza. While this was performed under strict biosecurity measures, including laboratory containment and personal safety considerations, the researchers did re-create what had been an extinct version of influenza [6]. Infectious cDNA clones and reverse-genetics systems have existed for decades and effectively generate a wide range of viruses. Most recently, for SARS-CoV and SARS-CoV-2, they enabled reconstruction and variant engineering for research purposes [7,8]. Although for more difficult segmented dsRNA viruses (e.g., bluetongue), reverse genetics required longer development efforts, it now fully supports robust recovery and bioengineering [9]. Technology has also advanced for large dsDNA viruses—for example, yeast-based synthetic genomics methods have enabled genome-scale manipulations of HSV-1 [10], and with the help of a helper virus, recovery of synthetically developed Vaccinia virus [11] and African swine fever virus [12] now exists. Collectively, these efforts definitively prove that sequence-to-virus pipelines are possible across many virus families and are not just limited to bacteriophages that were recently recreated by AI [2]. Moreover, the time to synthesize novel sequences has accelerated exponentially with desktop DNA synthesizers, giving the potential to develop new infectious clones of viruses rapidly. To address the evolving threat landscape of AI-engineered viruses, CRISPR-based antivirals offer a uniquely programmable and rapid-response solution, further discussed below.

3. CRISPR Antivirals: A Programmable Firewall Against Synthetic Biology Threats

Unlike small molecules, antivirals, or therapeutic monoclonal antibodies that require a deep understanding of protein function, target selectivity, and biological context-dependent mechanism of action (MoA), CRISPR activity in any mammalian cell relies only on the presence of a short strand of RNA complementary to a short sequence of the target virus genome. This direct MoA enables sequence-specific, multiplexed targeting of viral genes, disrupting replication and lowering disease burden with the unique ability to be rapidly reprogrammed to target and cleave a different virus simply with new gRNAs. CRISPR/Cas13 effectors can degrade RNA viral genomes and transcripts across diverse viruses, including human infectious pathogens and zoonotic strains (e.g., influenza A, coronaviruses, dengue) [13,14,15,16,17,18]. We have recently shown that DNA-targeting Cas9-based multiplexed CRISPR can ablate African swine fever virus to facilitate survival of pigs infected with an otherwise lethal pathogen [19], highlighting the potential to treat other large DNA viruses such as Smallpox and Mpox.
Applications of single-gRNA CRISPR antiviral modalities could create selective pressure that could allow for the emergence of escape variants over time. Consequently, multi-gRNA cassettes designed against highly conserved, functionally constrained target regions raise the mutational hurdle and reduce the likelihood of pressure-based escape variants. For RNA viruses, simultaneous targeting and cleavage at multiple conserved loci often imposes severe fitness costs compared to point mutations that can overcome conventional drugs with a single molecule MoA. Although AI-designed viruses could distribute synonymous changes to avoid CRISPR targeting gRNAs, they cannot circumvent newly designed gRNAs, a capability by which CRISPR systems can keep pace with virus evolution or AI-designed viruses—even in the face of continuously changing biothreats—and represents a unique opportunity to design and deploy a treatment at the speed of an infection.
To prioritize stability and durability, replication-competent DNA-based CRISPR antivirals represent a compelling option, as DNA offers lower cost and greater reliability over RNA platforms, while also allowing for both the Cas and multiplex gRNAs to be delivered as a single payload. DNA-based systems also have the potential for lyophilization and stockpile treatments over the cold-chain that is required for RNA-based delivery systems. However, there is no single delivery mechanism that will fit all biothreats. Different viruses have different tissue tropisms and require different therapeutic strategies. We recently demonstrated the effectiveness of a Cas9 therapeutic using Lipid-nanoparticle delivery for African swine fever, where both the virus and therapeutic were able to interact in the blood, likely in circulating macrophage cells that serve as the primary site of replication for ASFV [19]. However, for respiratory diseases, intranasal or intratracheal administration would likely be required to deliver the treatment to infected respiratory tissues. An alternative delivery option could be Adeno-associated viruses (AAV) to achieve multi-week airway expression of Cas and gRNAs. Other viral vectors could be used for potentially larger payloads, such as helper-dependent adenovirus (HD-Ad), and robust acute expression or non-replicating HSV amplicons with large payload capacity could also be used as a delivery system. Regardless, all delivery platforms will have to be matched to the characteristics of the targeted disease, and multi-site gRNA cassettes remain essential. Safety considerations still need to be determined even in the case of rapid response for emergency outbreaks, such as vector biodistribution, vector immunogenicity (AAV/Ad), off-target, and collateral effects (Cas13). All CRISPR treatments will likely favor non-integrating vectors and time-limited promoters or delivery systems [20] (Table 3). Taken together, we will discuss below the feasibility of CRISPR antivirals for rapid response to new or emerging viruses.

4. CRISPR-Enabled Rapid-Response Potential

Upon first attribution of symptoms to a novel pathogen, next-generation sequencing (NGS) platforms must be capable of identifying both known and unknown viral sequences, rather than relying solely on similarity to established viral signatures. The recent emergence of AI-generated baculovirus-like genomes illustrates this challenge: several synthetic sequences were not recognized as viral DNA by conventional bioinformatic filters [2]. This underscores the importance of recognizing that previously unseen genomic signatures may represent AI-engineered biothreats. In most outbreaks involving highly pathogenic viruses, large viral burdens within the host enable early detection through clinical symptoms or post-mortem analysis. Once a unique nucleotide signature is identified, CRISPR-based targeting sequences (gRNAs) can be computationally designed within minutes, so long as the appropriate informatics infrastructure is in place.
Advances in de novo DNA synthesis now allow for commercial and benchtop systems to produce short nucleic acid fragments in near-real time. These gRNA sequences can be rapidly assembled into delivery backbones, such as viral vectors or nanoparticle formulations. The urgency and scale of production would naturally be dictated by disease morbidity and mortality. Despite the theoretical feasibility of this “minutes-to-therapeutic” model, several major challenges remain. These include scalable manufacturing infrastructure, continuous genomic surveillance to detect escape mutants, and automated regeneration of gRNA cocktails guided by AI-assisted analysis for both safety and efficacy. Synthetic sequences should be screened under established synthesis-monitoring frameworks [28,29]. Moreover, AI-enabled gRNA design tools must incorporate risk assessments for host off-target effects, predictive modeling for cleavage efficacy, and conserved-region prioritization to ensure robust resistance to viral evolution [30,31].

Computational Resources for CRISPR gRNA Design

A growing ecosystem of computational resources supports rapid, accurate CRISPR gRNA design across nucleases and host systems. It is useful to distinguish resources that distribute pre-vetted gRNA from those that enable de novo design. PAC-MAN (Prophylactic Antiviral CRISPR in huMAN cells) refers to a Cas13-based antiviral strategy rather than a standalone design algorithm. Following that study, public portals, such as the Stanford CRISPR PAC-MAN site, provided curated, “family-covering” crRNA pools (e.g., for Coronaviridae) intended for immediate experimental use, emphasizing breadth and prior validation rather than custom design [32]. In parallel, the New York Genome Center’s Cas13Design supports custom gRNA selection for Cas13d, allowing users to tailor crRNAs to new or evolving RNA viral genomes [33]. In practice, a rapid pipeline often combines these assets, drawing on curated panels for speed while invoking custom design when coverage gaps or emerging variants demand novel gRNA.
For the development of rapid response pipelines, Seek Labs uses BioSeeker™ and to create a disease atlas of curated genomic targets spanning the majority of clinically relevant infectious viruses, which can accelerate selection of viral targeting gRNAs by pre-mapping a set of gRNAs that pan-target all isolates of a particular virus. Complementing this, BioSeeker generates “safe-gRNA” from multi-host genomic data to minimize off-target risks, providing candidate sets that are both efficacious and deployment-ready in diverse biological contexts.
Beyond these, several widely used platforms provide design, analysis, and workflow integration. Tools such as CHOPCHOP [34], EuPaGDT [35], and TIGER [36] offer web-based gRNA design and annotation across multiple nucleases and organisms, incorporating common scoring and off-target heuristics. Commercial and developer ecosystems (e.g., IDT, Synthego, Benchling) can add capabilities for synthesis-ready formatting and cloning strategy output. New research-driven resources from the Arc Institute [37] and others continue to refine predictive models and benchmarking datasets. Together, these assets span the spectrum from curated, plug-and-play gRNA pools to fully custom, model-driven design, enabling teams to match turnaround time and rigor to the demands of each project.

5. Governance and ELSI for AI-Enabled Synthetic Virology: DURC to Digital Biosecurity

Synthetic biology (SynBio) lies at the cutting edge of bioengineering advances, enabling the design and creation of custom genetic systems and novel organisms to address complex challenges, such as targeted therapies and ultra-sensitive biosensors [38]. The integration of AI supercharges these efforts with accelerated sequence predictions, optimized designs, and scaled workflows, paving the way for powerful defensive tools to counter biothreats [39,40]. To mitigate the misuse of SynBio and AI-bioengineering, risk assessments are run to inform the Biological Weapons Convention (BWC) on potential threats [14,19,20,28,41]. Existing oversight from the Dual Use Research of Concern (DURC), Enhanced Pathogens with Pandemic Potential (ePPP/PEPP) policies, and sequence-data governance (e.g., NIH GDS, GDPR, OECD guidance) all emphasize an integrated, holistic approach involving government, academia, and industry [30,41,42,43,44,45,46,47,48,49]. However, when innovation and capabilities expand rapidly—including digital diffusion of models generating novel sequences or when parts of a workflow occur outside traditional oversight structures—risks can outpace effective responses [30,39,40,44,47].

5.1. What Current Frameworks Cover—And Where They Fall Short

Oversight agencies such as DURC and ePPP require institutions to assess whether legitimate R&D experiments could be misused to create, enhance, or disseminate dangerous pathogens [42,43,44]. Furthermore, they rely on these same institutions to manage risks through review, mitigation, or prohibition [42,43]. These entities work best when projects are bounded (defined organism, methods, materials) and when risk is confined to physical lab spaces [30,44]. They often fall short when design, optimization, and initial screening are in silico, when cloud-based or third-party providers execute steps, and when the AI models’ access, prompts, or outputs are not explicitly governed or do not have robust stewardship programs in place [30,39,47]. Data access or sharing rules can reduce indiscriminate sharing, but few policies comprehensively address data quality, security, compliance (i.e., data integrity, access/control points, metadata management, prompt filters, audits and monitoring, etc.) [40,43,47], or supply-chain screening for sequence-to-physical translation [45,46,50].

5.2. Historical Stress Tests and What They Changed

A series of landmark studies have shaped the Synthetic biology space: 1918 influenza reconstruction [51]; H5N1 airborne transmission in ferrets [52]; and synthetic horsepox from DNA fragments [5]. Together, these studies demonstrated that high scientific value could coexist with substantive risk in allowing an extinct virus to be recreated. More recent AI-enabled sequence design [2], coupled with inexpensive benchtop or ready-to-order DNA synthesis suppliers, has exposed gaps in policies and procedures required to advance research and also minimize risk. Table 4 summarizes representative cases, specific DURC/ePPP concerns, and observable policy outcomes that highlight the critical and urgent need to extend oversight from wet-lab activities to digital design and synthesis capabilities.

5.3. Digital Biosecurity: The AI Risk Surface

AI systems can (i) generate novel or optimized sequences with pathogenic traits, (ii) design constructs that slip past naïve sequence filters, (iii) translate literature and programming code into step-by-step protocols, and (iv) distribute capability beyond strictly regulated environments. Without model-level controls, the same tools that accelerate beneficial design can markedly reduce barriers to misuse. This expanding digital risk surface argues for governance that reaches upstream of research lab oversight to AI modeling, data access, prompt and output filtering, and auditability. Additionally, rigorous screening of nucleotide synthesis orders is required for a policy that keeps pace with the AI design of potential biothreats.

6. Examples of Existing Viral Pathogens for CRISPR Antivirals

6.1. Avian and Pandemic Strains of Influenza

Seasonal Influenza cases in the US over the past 3 years (2022–2025) have shown increasing severity, and the most recent 2024–2025 season was classified by the CDC as “high severity” with the highest estimated disease burden in the past decade based on outpatient visits, hospitalizations, and deaths [57,58]. Globally, the WHO estimates 290,000–650,000 annual respiratory deaths from seasonal influenza, with recent seasons exacerbated by dominant A(H1N1) pdm09 and A(H3N2) strains for which the CDC reported moderate overall effectiveness (VE) at 30–60% [59,60]. Coupled with this is the ongoing global outbreak of the highly pathogenic avian influenza A(H5N1) clade2.3.4.4b, which began in 2020 and has intensified from 2022 onward, affecting wild birds, domestic poultry, livestock, and humans [61]. As of February 2025, it has spread to every continent except Australia [61]. While no sustained mammal-to-mammal or human-to-human transmission has been reported, risk surveillance is high due to mammalian adaptations [62]. Together, these incidents serve as compelling reminders that existing respiratory viral diseases continue to pose a significant threat, where CRISPR modalities could be a potential preparedness treatment option.
CRISPR antivirals offer a rapid design, easily reprogrammable, sequence-directed way to target the influenza genome, disrupt viral replication, and limit cross-species spread [12,14]. By designing multiplexing Cas13 gRNAs to target and cleave conserved RNA genome segments, we can rapidly update gRNAs and reprogram the CRISPR system as genome sequences shift, an approach that not only mediates direct viral ablation but could also be used to complement vaccines and small molecule polymerase/neuraminidase inhibitors [12,14]. CRISPR antivirals remain agnostic to antigenic drift/shift and could function in multiple biological contexts. Translationally, delivery strategies can utilize upper-airway routes already validated for nucleic acid-based modalities: intranasally administered lipid nanoparticles (or inhaled aerosols) that deliver payload to airway epithelium and immune cells, enabling post-exposure treatment or potential ring prophylaxis in farms and healthcare settings [63,64]. The expanding mammalian footprint of Influenza A underscores a need for both commercially available therapeutics along with emergency-use gRNA stockpiles, which would not have evolutionary pressure if not in use. Additionally, CRISPR could decrease the risk from AI-designed viruses that could evade treatment. By using a platform approach, new gRNAs could be finalized within days of a new sequence or AI-designed variant.
Mounting evidence supports this strategy. Freije et al. (CARVER) demonstrated robust suppression of influenza A replication using human cell-based in vitro models and the CRISPR/Cas13b system with multiplexed gRNAs to target the conserved NP (nucleoprotein) and M (matrix) viral genes [21]. This study was independently validated using the PAC-MAN framework to take a more prophylactic CRISPR deployment in human lung epithelial cells, targeting conserved IAV gene regions, particularly in H1N1, with an eye toward broader, pan-influenza strategies [32]. Alternatively, we used our proprietary AI platform, Bioseeker, using a multiplex approach where all gRNAs achieve pan-targeting CRISPR for influenza A with predicted high efficacy. Translationally, mRNA-encoded Cas13a with PB1/PB2-targeting gRNAs, delivered to mice via nebulization post-infection, was shown to reduce influenza A gene expression consistent with therapeutic application [13]. Additional studies report site-specific genomic RNA degradation of IAV using mRNA-Cas13a and continued efficacy signals, supporting an intranasal or inhaled LNP/polymer administration as a viable route for treating acute flu [15]. Newer preclinical work also suggests cell-selective or targeted LNP (tLNP) delivery formulations of Cas13d can achieve large reductions in viral load, leading to complete survival in otherwise lethal mouse models, highlighting the potential for airway-targeted formulations that improve potency and safety as a selective antiviral treatment [16]. Taken together, these results support the promise of a response-ready, multiplexed Cas13 therapeutic for the current H5N1 panzootic [13] that could also be adapted for newly emerging strains or AI-designed biothreats.

6.2. Smallpox and Monkeypox

With recent outbreaks of mpox and the ongoing biothreat potential of smallpox, a CRISPR-Cas9 therapeutic for orthopoxviruses could be used to target conserved viral DNA using multiplexed gRNAs to inhibit replication, which could complement tecovirimat, which now shows both on-therapy and transmitted resistance [65,66]. Proof-of-concept with vaccinia (a surrogate for other orthopoxviruses) demonstrated that Cas9 gRNAs against conserved regions markedly reduced viral titers in human cells, including when delivered by AAV vectors [67]. For deployment, nonviral formulations enable stockpile-ready manufacturing and lesion-proximal administration: lipid nanoparticles or dissolvable microneedle patches can localize editors to skin and mucosa, offering intradermal/topical options for mpox lesions and systemic options for severe disease [68]. Together, Cas9 with multiplexed, pre-vetted gRNA sequences against conserved orthopoxviruses targets provides a programmable antiviral that can be retargeted within days as new mpox lineages, including potential tecovirimat resistance [66,69], emerge or potential AI-engineered variants that circumvent current countermeasures to orthopoxviruses. While CRISPR systems have been used experimentally to target both RNA and DNA viral genomes, the limitations for the broad use of CRISPR antivirals will be covered in the next section.

7. Current Limitations for CRISPR Antivirals

7.1. Targeted Tissue Delivery

Current CRISPR delivery methods for treatment strategies largely rely on two systems with very different risk profiles: lipid nanoparticles (LNP) and adeno-associated viruses (AAV) vectors [70]. LNPs encapsulate payloads comprising a Cas effector and gRNAs delivered as DNA plasmids, RNPs, or mRNA-gRNA and support transient expression, scalable manufacturing, and repeat dosing. However, LNPs often have poor tissue targeting outside the liver, variable endosomal escape, and may even be associated with lipid-driven toxicity. AAVs facilitate more durable expression in non-dividing cells but have a size-limited capacity, can drive strong population-level immunity, and carry host-genome integration risks. For dual-use concerns, a key point is that both delivery systems can constrain permanent host genome impact; LNPs limit persistence and tissue reach, while AAVs have restricted cargo size and can be blocked by common antibodies. Efforts to expand tropism, improve escape, or increase cargo capacity for therapeutic benefit can be monitored.

7.2. Potential Immunogenicity of CRISPR System Components

As with all large molecule/biological modalities, as well as cell and gene therapies, there is a risk of adverse events arising from pre-existing adaptive immunity in humans—driven by prior exposure to similar bacterial proteins manifesting as anti-Cas antibodies and cytotoxic T-cells that neutralize the nuclease, eliminate Cas-expressing cells, and impair efficacy, particularly with persistent viral delivery, like AAVs [71]. Additional risks include innate inflammatory responses to bacterial genetic or protein matter, potential systemic toxicity, anaphylaxis upon redosing, and reduced therapeutic efficacy in tissues. To mitigate these risks, bioengineering strategies to reduce immunogenicity include epitope masking (e.g., R338G substitution in SaCas9 [72] or using orthologs from non-pathogenic bacteria to evade recognition). Deploying LNPs as transient non-viral delivery systems for short-lived expression, transient immunosuppression treatments, and localized administration can all help limit systemic exposure to reduce the risk of immune-related adverse event (IRAE) [73].

7.3. Limitations to Sample-to-Formulation

The characterization of CRISPR antivirals as enabling “sample-to-formulation” within hours to days is grounded in emerging empirical experience, although the exact timeframe depends on the platform and indication. For RNA viruses, computational gRNA design and in vitro screening can now be completed within hours of sequence availability, as demonstrated repeatedly during the SARS-CoV-2 pandemic, where Cas13 gRNA sets were designed and validated against new variants on sub-week timescales. In practice, the dominant bottlenecks are (i) gRNA synthesis and scale-up, and (ii) fit-for-purpose delivery formulation. Chemical synthesis or in vitro transcription of gRNAs (or gRNA-encoding mRNA) is typically achieved within 1–3 days in a research or rapid-response setting, after which LNP or viral vector formulation can proceed using modular, pre-optimized platforms that require only substitution of the nucleic acid payload. Formulation optimization may extend timelines when novel tissue targets, routes of administration, or safety margins are required, but this is increasingly mitigated by using standardized LNP compositions, high-throughput formulation screens, and prior Chemistry, Manufacturing, and Controls (CMC) packages that can be rapidly updated rather than built de novo. Thus, while real-world deployment still depends on manufacturing and regulatory constraints, the molecular design-to-candidate-formulation phase for CRISPR antivirals is no longer intrinsically rate-limiting and can reasonably occur on the order of hours to a few days under prepared operational conditions.

8. Conclusions and Outlook

The convergence of large language models for genomic and transcriptomic sequences, synthetic biology, and CRISPR antivirals is reshaping preparedness strategies for viral pathogens. As these generative AI models begin to produce functional novel viral genomes, the field is moving rapidly from recombining known motifs to exploring innovative approaches in viral construction. Given the relative ease of execution, the potential for misuse is high and requires viral defense approaches are equally agile.
Across experimental and translational studies, CRISPR antivirals have emerged as highly compelling, sequence-reprogrammable countermeasures that can be deployed at the speed of infections; Cas9 platforms can combat DNA viruses, while Cas13 and variants target RNA viruses. In vivo data across HSV-1 [24], influenza, and SARS-CoV-2 show meaningful disease suppression or viral clearance using AAV, LNP, or mRNA delivery. However, significant challenges remain, including off-target risks, vector immunogenicity, delivery constraints, incomplete viral clearance leading to persistence, or potential evolutionary escape, particularly with single gRNA delivery systems.
Nevertheless, the potential for programmable CRISPR to serve as an appropriate countermeasure is highly compelling, and a path toward feasibility can be mapped using near-term priorities:
  • Standardized CRISPR benchmarking against the most common viruses to determine adequate antiviral effectiveness and tolerability in vivo.
  • Multiplex gRNA designs to prevent viral evolutionary escape along with probabilistic escape modeling, deep multiplexing in conserved regions, and adaptive gRNA rotation coupled with surveillance screening.
  • Targeted delivery for improved tissue tropism, repeat-dose capability, and mitigation of anti-vector immunogenicity.
  • Governance and auditability with pre-cleared “safe-gRNAs” repositories, open-source safety/off-target tools, traceable design logs, regulated access to predictive models, standardized metrics for data quality, etc.
  • Regulatory pathways ready with EUA-like mechanisms for programmable endonucleases, comparability rules for iterative gRNA updates, and adaptive rapid trials.
While many unknowns remain, especially around long-term safety, host off-target or collateral activities, immunogenicity effects, and real-world escape dynamics, the direction for programmable therapies is clear: CRISPR holds significant promise as an adaptable, rapid-response foundation for antiviral preparedness. With the advent of AI-made viruses, biothreat risk is escalated, and the threat window is markedly shorter; thus the defensive timeline must be equally streamlined, scaled, and accelerated. With integrated biology, engineering, policy, and governance, CRISPR antivirals can move from laboratory modalities to a trusted public-health strategy.
It is important to mention that, while treatment modalities such as CRISPR-based antivirals programmed with target-specific guides hold promise as precise and rapid-response countermeasures against dual-use synthetic viruses engineered or AI-scripted as a biothreat, by cleaving the viral genomes directly, some major constraints would need to be addressed to make this a viable strategy in the near future. Firstly, the manufacturing of the CRISPR/Cas-gRNA payload is expensive, time-intensive, and subject to variability in the end product. As detailed below, LNP and AAV delivery approaches have limitations and risks. Rapid safety evaluation in an emergency carries the risk of missing off-target activity or immune/inflammatory reactions leading to adverse outcomes. AI-enabled virus design also remains a new development, and while the EVO system [2] did achieve functional bacteriophage genomes, modeling human infection, viral transmission, and immune evasion is still beyond current capabilities. So while the capability to create a viable, bioengineered human virus is now technically possible in principle, there remain significant gaps in expertise and execution needed to create an imminent AI-scripted biothreat.

Author Contributions

D.P.G. and A.O. conceptualized, wrote the first draft, and edited the final draft. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Portions of language polishing were assisted by a generative AI tool; all scientific claims, citations, and text were verified and edited by the authors, who are responsible for the content.

Conflicts of Interest

All authors are employed by Seek Labs, Inc., which holds patents for CRISPR-based therapeutics. Seek Labs is pursuing its PTAP platform powered by Bioseeker for programmable therapeutics using CRISPR-based systems.

References

  1. Wittmann, B.J.; Alexanian, T.; Bartling, C.; Beal, J.; Clore, A.; Diggans, J.; Flyangolts, K.; Gemler, B.T.; Mitchell, T.; Murphy, S.T.; et al. Strengthening nucleic acid biosecurity screening against generative protein design tools. Science 2025, 390, 82–87. [Google Scholar] [CrossRef] [PubMed]
  2. King, S.; Driscoll, C.; Li, D.; Guo, D.; Merchant, A.; Brixi, G.; Wilkinson, M.; Hie, B. Generative design of novel bacteriophages with genome language models. bioRxiv 2025. [Google Scholar] [CrossRef]
  3. Cello, J.; Paul, A.V.; Wimmer, E. Chemical synthesis of poliovirus cDNA: Generation of infectious virus in the absence of natural template. Science 2002, 297, 1016–1018. [Google Scholar] [CrossRef]
  4. Smith, H.O.; Hutchison, C.A., III; Pfannkoch, C.; Venter, J.C. Generating a synthetic genome by whole genome assembly: phix174 bacteriophage from synthetic oligonucleotides. Proc. Natl. Acad. Sci. USA 2003, 100, 15440–15445. [Google Scholar] [CrossRef] [PubMed]
  5. Noyce, R.S.; Lederman, S.; Evans, D.H. Construction of an infectious horsepox virus vaccine from chemically synthesized DNA fragments. PLoS ONE 2018, 13, e0188453. [Google Scholar] [CrossRef]
  6. Tumpey, T.M.; Basler, C.F.; Aguilar, P.V.; Zeng, H.; Solórzano, A.; Swayne, D.E.; Cox, N.J.; Katz, J.M.; Taubenberger, J.K.; Palese, P.; et al. Characterization of the reconstructed 1918 spanish influenza pandemic virus. Science 2005, 310, 77–80. [Google Scholar] [CrossRef]
  7. Yount, B.; Curtis, K.M.; Fritz, E.A.; Hensley, L.E.; Jahrling, P.B.; Prentice, E.; Denison, M.R.; Geisbert, T.W.; Baric, R.S. Reverse genetics with a Full-Length infectious cDNA of severe acute respiratory syndrome coronavirus. Proc. Natl. Acad. Sci. USA 2003, 100, 12995–13000. [Google Scholar] [CrossRef]
  8. Thao, T.T.N.; Labroussaa, F.; Ebert, N.; V’Kovski, P.; Stalder, H.; Portmann, J.; Kelly, J.; Steiner, S.; Holwerda, M.; Kratzel, A.; et al. Rapid reconstruction of SARS-CoV-2 using a synthetic genomics platform. Nature 2020, 582, 561–565. [Google Scholar] [CrossRef]
  9. Celma, C.C.P.; Roy, P. Development of reverse genetics systems for bluetongue virus: Recovery of infectious virus from synthetic RNA transcripts. J. Virol. 2008, 82, 8339–8348. [Google Scholar] [CrossRef]
  10. Oldfield, L.M.; Grzesik, P.; Voorhies, A.A.; Alperovich, N.; MacMath, D.; Najera, C.D.; Chandra, D.S.; Prasad, S.; Noskov, V.N.; Montague, M.G.; et al. Genome-wide engineering of an infectious clone of herpes simplex virus type 1 using synthetic genomics assembly methods. Proc. Natl. Acad. Sci. USA 2017, 114, E8885–E8894. [Google Scholar] [CrossRef] [PubMed]
  11. Domi, A.; Moss, B. Engineering of a vaccinia virus bacterial artificial chromosome in Escherichia coli by bacteriophage λ–based recombination. Nat. Methods 2005, 2, 95–97. [Google Scholar] [CrossRef]
  12. Fuchs, W.; Assad-Garcia, N.; Abkallo, H.M.; Xue, Y.; Oldfield, L.M.; Fedorova, N.; Hübner, A.; Kabuuka, T.; Pannhorst, K.; Höper, D.; et al. A synthetic genomics-based African swine fever virus engineering platform. Sci. Adv. 2025, 11, eadu7670. [Google Scholar] [CrossRef]
  13. Blanchard, E.L.; Vanover, D.; Bawage, S.S.; Tiwari, P.M.; Rotolo, L.; Beyersdorf, J.; Peck, H.E.; Bruno, N.C.; Hincapie, R.; Michel, F.; et al. Treatment of influenza and SARS-CoV-2 infections via mRNA-encoded Cas13a in rodents. Nat. Biotechnol. 2021, 39, 717–726. [Google Scholar] [CrossRef] [PubMed]
  14. Teng, F.; Li, J.; Cui, B.; Wu, J.; Toischer, K.; Hasenfuß, G.; Xu, X. Crispr/cas13-based Anti-RNA viral approaches. Genes 2025, 16, 875. [Google Scholar] [CrossRef]
  15. Chaves, L.C.S.; Orr-Burks, N.; Vanover, D.; Mosur, V.V.; Hosking, S.R.; Kumar, E.K.P.; Jeong, H.; Jung, Y.; Assumpção, J.A.F.; Peck, H.E.; et al. mRNA-encoded Cas13 treatment of Influenza via site-specific degradation of genomic RNA. PLoS Pathog. 2024, 20, e1012345. [Google Scholar] [CrossRef]
  16. Wu, Z.; Zhao, C.; Ai, H.; Wang, Z.; Chen, M.; Lyu, Y.; Tong, Q.; Liu, L.; Sun, H.; Pu, J.; et al. A Susceptible Cell-Selective Delivery (SCSD) of mRNA-Encoded Cas13d Against Influenza Infection. Adv. Sci. 2025, 12, e2414651. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  17. Kukushkin, V.; Ivanov, R.; Reshetnikov, V. Progress in CRISPR/Cas13-Mediated suppression of influenza a virus. Biochemistry 2025, 90, 786–803. [Google Scholar] [CrossRef]
  18. Basu, M.; Zurla, C.; Auroni, T.T.; Vanover, D.; Chaves, L.C.S.; Sadhwani, H.; Pathak, H.; Basu, R.; Beyersdorf, J.P.; Amuda, O.O.; et al. mRNA-encoded Cas13 can be used to treat dengue infections in mice. Nat. Microbiol. 2024, 9, 2160–2172. [Google Scholar] [CrossRef] [PubMed]
  19. Verma, N.; O’Mahony, A.; Mohammad, R.; Keiser, D.; Mosman, C.W.; Holden, D.; Starr, K.; Bauer, J.; Bauer, B.; Suntisukwattana, R.; et al. The First CRISPR-Based Therapeutic (SL_1.52) for African Swine Fever Is Effective in Swine. Viruses 2025, 17, 1504. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  20. Zahedipour, F.; Zahedipour, F.; Zamani, P.; Jaafari, M.R.; Sahebkar, A. Harnessing CRISPR technology for viral therapeutics and vaccines: From bench to bedside. Drug Discov. Today 2024, 29, 103748. [Google Scholar] [CrossRef]
  21. Abbott, T.R.; Dhamdhere, G.; Liu, Y.; Lin, X.; Goudy, L.; Zeng, L.; Chemparathy, A.; Chmura, S.; Heaton, N.S.; Debs, R.; et al. Development of CRISPR as an Antiviral Strategy to Combat SARS-CoV-2 and Influenza. Cell 2020, 181, 865–876.e12. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  22. Chen, P.; Chen, M.; Chen, Y.; Jing, X.; Zhang, N.; Zhou, X.; Li, X.; Long, G.; Hao, P. Targeted inhibition of Zika virus infection in human cells by CRISPR-Cas13b. Virus Res. 2022, 312, 198707. [Google Scholar] [CrossRef]
  23. Saayman, S.; Ali, S.A.; Morris, K.V.; Weinberg, M.S. The therapeutic application of CRISPR/Cas9 technologies for HIV. Expert. Opin. Biol. Ther. 2015, 15, 819–830. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  24. Amrani, N.; Luk, K.; Singh, P.; Shipley, M.; Isik, M.; Donadoni, M.; Bellizzi, A.; Khalili, K.; Sariyer, K.; Neumann, D.; et al. CRISPR-Cas9-mediated genome editing delivered by a single AAV9 vector inhibits HSV-1 reactivation in a latent rabbit keratitis model. Mol. Ther. Methods Clin. Dev. 2024, 32, 101303. [Google Scholar] [CrossRef]
  25. Owens, C.M.; Diner, B.; Fusco, R.; King, E.; Friedland, A.; Singhal, P.; Gogi, K.; Harbinski, F.; Shen, S.; Stefanidakis, M.; et al. CRISPR/Cas9 Targeted Disruption of Herpes Simplex Virus type 1 in a Rabbit Latency Model Reduces Viral Reactivation and Associated Corneal Pathology. Investig. Ophthalmol. Vis. Sci. 2018, 59, 374. [Google Scholar]
  26. Xiao, J.; Deng, J.; Zhang, Q.; Ma, P.; Lv, L.; Zhang, Y.; Li, C.; Zhang, Y. Targeting human cytomegalovirus IE genes by CRISPR/Cas9 nuclease effectively inhibits viral replication and reactivation. Arch. Virol. 2020, 165, 1827–1835. [Google Scholar] [CrossRef]
  27. Yi, J.; Lei, X.; Guo, F.; Chen, Q.; Chen, X.; Zhao, K.; Zhu, C.; Cheng, X.; Lin, J.; Yin, H.; et al. Co-delivery of Cas9 mRNA and guide RNAs edits hepatitis B virus episomal and integration DNA in mouse and tree shrew models. Antivir. Res. 2023, 215, 105618. [Google Scholar] [PubMed]
  28. ASPR/NSTC Nucleic Acid Synthesis Screening Framework (Full PDF). 2024. Available online: https://aspr.hhs.gov/s3/documents/ostp-nucleic-acid-synthesis-screening-framework-sep2024.pdf (accessed on 12 October 2025).
  29. IBBIS International Screening Standards (Common Mechanism Overview). 2024. Available online: https://ibbis.bio/our-work/international-screening-standards/ (accessed on 12 October 2025).
  30. NSABB Proposed Biosecurity Oversight Framework for the Future of Science. 2023. Available online: https://osp.od.nih.gov/wp-content/uploads/2023/03/NSABB-Final-Report-Proposed-Biosecurity-Oversight-Framework-for-the-Future-of-Science.pdf (accessed on 12 October 2025).
  31. NIH OSP US Government Releases Policy for Oversight of DURC and Pathogens with Enhanced Pandemic Potential. 2024. Available online: https://osp.od.nih.gov/us-government-releases-policy-for-oversight-of-dual-use-research-of-concern-and-pathogens-with-enhanced-pandemic-potential/ (accessed on 12 October 2025).
  32. Lin, X.; Liu, Y.; Chemparathy, A.; Pande, T.; La Russa, M.; Qi, L.S. A comprehensive analysis and resource to use CRISPR-Cas13 for broad-spectrum targeting of RNA viruses. Cell Rep. Med. 2021, 2, 100245. [Google Scholar] [CrossRef] [PubMed]
  33. Guo, X.; Rahman, J.; Wessels, H.-H.; Méndez-Mancilla, A.; Haro, D.; Chen, X.; Sanjana, N.E. Transcriptome-wide Cas13 guide RNA design for model organisms and viral RNA pathogens. Cell Genom. 2021, 1, 100001. [Google Scholar] [CrossRef] [PubMed]
  34. Labun, K.; Montague, T.G.; Krause, M.; Torres Cleuren, Y.N.; Tjeldnes, H.; Valen, E. CHOPCHOP v3: Expanding the CRISPR web toolbox beyond genome editing. Nucleic Acids Res. 2019, 47, W171–W174. [Google Scholar] [CrossRef]
  35. Peng, D.; Tarleton, R. EuPaGDT: A web tool tailored to design CRISPR guide RNAs for eukaryotic pathogens. Microb. Genom. 2015, 1, e000033. [Google Scholar] [CrossRef]
  36. Wessels, H.-H.; Stirn, A.; Méndez-Mancilla, A.; Kim, E.J.; Hart, S.K.; Knowles, D.A.; Sanjana, N.E. Prediction of on-target and off-target activity of CRISPR–Cas13d guide RNAs using deep learning. Nat. Biotechnol. 2023, 42, 628–637. [Google Scholar] [CrossRef] [PubMed]
  37. Wei, J.; Lotfy, P.; Faizi, K.; Baungaard, S.; Gibson, E.; Wang, E.; Slabodkin, H.; Kinnaman, E.; Chandrasekaran, S.; Kitano, H.; et al. Deep learning and CRISPR-Cas13d ortholog discovery for optimized RNA targeting. Cell Syst. 2023, 14, 1087–1102.e13. [Google Scholar] [CrossRef]
  38. National Academies of Sciences, Engineering, and Medicine. Biodefense in the Age of Synthetic Biology; National Academies of Sciences, Engineering, and Medicine: Washington, DC, USA, 2018. [Google Scholar]
  39. Pannu, J.; Doni, S.; Tillmann, U.; Marcus, J.; Jacobs, E. Dualuse capabilities of concern of biological AI models. PLoS Comput. Biol. 2025, 21, e1012975. [Google Scholar]
  40. Epstein, G.L. The evolution of United States governance policies for research using pathogens with enhanced pandemic potential. Appl. Biosaf. 2025, 30, 79–96. [Google Scholar] [CrossRef]
  41. Organisation for Economic Co-operation and Development. Recommendation of the Council on Enhancing Access to and Sharing of Data; Organisation for Economic Co-operation and Development: Paris, France, 2021. [Google Scholar]
  42. United States Government. United States Government Policy for Oversight of Life Sciences Dual Use Research of Concern; United States Government: Washington, DC, USA, 2012.
  43. United States Government. United States Government Policy for Oversight of Dual Use Research of Concern and Pathogens with Enhanced Pandemic Potential (DURC/PEPP); United States Government: Washington, DC, USA, 2024.
  44. U.S. Department of Health and Human Services. Framework for Guiding Funding Decisions About Proposed Research Involving Enhanced Potential Pandemic Pathogens (P3CO); U.S. Department of Health and Human Services: Washington, DC, USA, 2025.
  45. Executive Office of the President, Office of Science and Technology Policy. Framework for Nucleic Acid Synthesis Screening; Executive Office of the President, Office of Science and Technology Policy: Washington, DC, USA, 2024.
  46. International Gene Synthesis Consortium. Harmonized Screening Protocol v3.0; International Gene Synthesis Consortium: Trieste, Italy, 2024. [Google Scholar]
  47. Executive Office of the President. Executive Order 14110: Safe, Secure, and Trustworthy Development and Use of Artificial Intelligence; Federal Register; Executive Office of the President: Washington, DC, USA, 2023.
  48. National Institutes of Health. Genomic Data Sharing (GDS) Policy—Overview; National Institutes of Health: Bethesda, MD, USA, 2025.
  49. European Union. General Data Protection Regulation (EU) 2016/679; European Union: Brussels, Belgium, 2016. [Google Scholar]
  50. Council on Strategic Risks. Supporting Follow-Up Screening for Flagged Nucleic Acid Synthesis Orders; Council on Strategic Risks: Washington, DC, USA, 2024. [Google Scholar]
  51. Taubenberger, J.K.; Baltimore, D.; Doherty, P.C.; Markel, H.; Morens, D.M.; Webster, R.G.; Wilson, I.A. Reconstruction of the 1918 influenza virus: Unexpected rewards from the past. mBio 2012, 3, e00201-12. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  52. Herfst, S.; Schrauwen, E.J.; Linster, M.; Chutinimitkul, S.; de Wit, E.; Munster, V.J.; Sorrell, E.M.; Bestebroer, T.M.; Burke, D.F.; Smith, D.J.; et al. Airborne transmission of influenza A/H5N1 virus between ferrets. Science 2012, 336, 1534–1541. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  53. Jackson, R.J.; Ramsay, A.J.; Christensen, C.D.; Beaton, S.; Hall, D.F.; Ramshaw, I.A. Expression of mouse interleukin-4 by a recombinant ectromelia virus suppresses cytolytic lymphocyte responses and overcomes genetic resistance to mousepox. J. Virol. 2001, 75, 1205–1210. [Google Scholar] [CrossRef]
  54. Esvelt, K.M.; Smidler, A.L.; Catteruccia, F.; Church, G.M. Concerning RNA-guided gene drives for the alteration of wild populations. Elife 2014, 3, e03401. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  55. Urbina, F.; Lentzos, F.; Invernizzi, C.; Ekins, S. Dual use of artificial-intelligence-powered drug discovery. Nat. Mach. Intell. 2022, 4, 189–191. [Google Scholar] [CrossRef]
  56. Adam, L.; McArthur GH 4th. Substitution Attacks: A Catalyst to Reframe the DNA Manufacturing Cyberbiosecurity Landscape in the Age of Benchtop Synthesizers. Appl. Biosaf. 2024, 29, 172–180. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  57. Centers for Disease Control and Prevention. Influenza-associated hospitalizations during a high-severity season—United States, 2024–2025. MMWR Morb. Mortal. Wkly. Rep. 2025, 74, 529–537. [Google Scholar]
  58. Centers for Disease Control and Prevention. Preliminary Estimated Flu Disease Burden: 2024–2025 Season; Centers for Disease Control and Prevention: Atlanta, GA, USA, 2025.
  59. World Health Organization. Influenza (Seasonal)—Key Facts; World Health Organization: Geneva, Switzerland, 2025. [Google Scholar]
  60. Frutos, A.M.; Cleary, S.; Reeves, E.L.; Ahmad, H.M.; Price, A.M.; Self, W.H.; Zhu, Y.; Safdar, B.; Peltan, I.D.; Gibbs, K.W.; et al. Interim estimates of 2024–2025 seasonal influenza vaccine effectiveness—United States. MMWR Morb. Mortal. Wkly. Rep. 2025, 74, 83–90. [Google Scholar] [CrossRef] [PubMed]
  61. FAO; WHO; WOAH. Updated Joint Public Health Assessment of Recent Influenza A(H5) Virus Events in Animals and People (March–July 2025 Update). Available online: https://www.who.int/publications/m/item/updated-joint-fao-who-woah-public-health-assessment-of-recent-influenza-a(h5)-virus-events-in-animals-and-people-july2025 (accessed on 28 July 2025).
  62. Capelastegui, F.; Goldhill, D.H. H5N1 2.3.4.4b: A review of mammalian adaptations and risk of pandemic emergence. J. Gen. Virol. 2025, 106, 002109. [Google Scholar] [CrossRef] [PubMed]
  63. Lokugamage, M.P.; Vanover, D.; Beyersdorf, J.; Hatit, M.Z.C.; Rotolo, L.; Echeverri, E.S.; Peck, H.E.; Ni, H.; Yoon, J.K.; Kim, Y.; et al. Optimization of lipid nanoparticles for the delivery of nebulized therapeutic mRNA to the lungs. Nat. Biomed. Eng. 2021, 5, 1059–1068. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  64. Jiang, A.Y.; Witten, J.; Raji, I.O.; Eweje, F.; MacIsaac, C.; Meng, S.; Oladimeji, F.A.; Hu, Y.; Manan, R.S.; Langer, R.; et al. Combinatorial development of nebulized mRNA delivery formulations for the lungs. Nat. Nanotechnol. 2024, 19, 364–375. [Google Scholar] [CrossRef]
  65. Smith, T.G.; Gigante, C.M.; Wynn, N.T.; Matheny, A.; Davidson, W.; Yang, Y.; Condori, R.E.; O’cOnnell, K.; Kovar, L.; Williams, T.L.; et al. Tecovirimat Resistance in Mpox Patients, United States, 2022–2023. Emerg. Infect. Dis. 2023, 29, 2426–2432. [Google Scholar] [CrossRef]
  66. Garrigues, J.M.; Hemarajata, P.; Espinosa, A.; Hacker, J.K.; Wynn, N.T.; Smith, T.G.; Gigante, C.M.; Davidson, W.; Vega, J.; Edmondson, H.; et al. Community spread of a human monkeypox virus variant with a tecovirimat resistance-associated mutation. Antimicrob. Agents Chemother. 2023, 67, e0097223. [Google Scholar] [CrossRef]
  67. Siegrist, C.M.; Kinahan, S.M.; Settecerri, T.; Greene, A.C.; Santarpia, J.L. CRISPR/Cas9 as an antiviral against Orthopoxviruses using an AAV vector. Sci. Rep. 2020, 10, 19307. [Google Scholar] [CrossRef]
  68. Kazemian, P.; Yu, S.Y.; Thomson, S.B.; Birkenshaw, A.; Leavitt, B.R.; Ross, C.J.D. Lipid-Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing Components. Mol. Pharm. 2022, 19, 1669–1686. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  69. Garrigues, J.M.; Hemarajata, P.; Karan, A.; Shah, N.K.; Alarcón, J.; Marutani, A.N.; Finn, L.; Smith, T.G.; Gigante, C.M.; Davidson, W.; et al. Identification of Tecovirimat Resistance-Associated Mutations in Human Monkeypox Virus-Los Angeles County. Antimicrob. Agents Chemother. 2023, 67, e00568-23. [Google Scholar] [CrossRef]
  70. Geng, G.; Xu, Y.; Hu, Z.; Wang, H.; Chen, X.; Yuan, W.; Shu, Y. Viral and non-viral vectors in gene therapy: Current state and clinical perspectives. EBioMedicine 2025, 118, 105834. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  71. Ewaisha, R.; Anderson, K.S. Immunogenicity of CRISPR therapeutics-Critical considerations for clinical translation. Front. Bioeng. Biotechnol. 2023, 11, 1138596. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  72. Shen, X.; Lin, Q.; Liang, Z.; Wang, J.; Yang, X.; Liang, Y.; Liang, H.; Pan, H.; Yang, J.; Zhu, Y.; et al. Reduction of Pre-Existing Adaptive Immune Responses Against SaCas9 in Humans Using Epitope Mapping and Identification. Cris. J. 2022, 5, 445–456. [Google Scholar] [CrossRef] [PubMed]
  73. Rasul, M.F.; Hussen, B.M.; Salihi, A.; Ismael, B.S.; Jalal, P.J.; Zanichelli, A.; Jamali, E.; Baniahmad, A.; Ghafouri-Fard, S.; Basiri, A.; et al. Strategies to overcome the main challenges of the use of CRISPR/Cas9 as a replacement for cancer therapy. Mol. Cancer 2022, 21, 64. [Google Scholar] [CrossRef]
Table 1. Comparison of antiviral options to emerging viral threats.
Table 1. Comparison of antiviral options to emerging viral threats.
Key FeatureCRISPRMonoclonal AntibodiesVaccinesSmall Molecule Antivirals
Mechanism of ActionGenome cleavage to directly degrade viral genomesBind to viral proteins or host receptors to block
entry or replication
Induce adaptive immune response against viral
antigens
Inhibit viral replication enzymes or host factors
Adaptable for Unknown VirusesProgrammable across diverse pathogens with rapid gRNA reconfigurationPredefined antigens with diverse protein modificationsPredefined viral genomes with high gene sequence variationsTarget-based discovery in different biological contexts
Response Time to OutbreakRapid: programmable
design
Moderate: requires
antibody optimization
Slow: requires antigen identification and
validation
Moderate: can
repurpose existing drugs or screens
Sample-to-FormulationPotentially hours to days6–12 months6–18 months1–3 years
Scale-up PotentialPotentially days to weeksMonthsMonthsMonths
Storage RequirementFormulation dependent 1Cold chainCold chainNone
Manufacturing ComplexityModerate: molecular
assembly scalable
High: cell culture-based productionHigh: depends on
platform (mRNA, vector, protein subunit)
Variable: chemical synthesis, often scalable
Breadth of EfficacyBroadly active against multiple viral strainsNarrow, virus-specificVariable, often strain-specificNarrow to moderate
Safety ProfileUnder development;
off-target risk possible
Generally safe; infusion-related reactions possibleEstablished safety; varies by platformKnown pharmacology, but side effects possible
Regulatory MaturityExperimentalWell establishedWell establishedWell established
Resistance PotentialLow (multi-target gRNA
design possible)
Moderate (viral mutations may escape binding)High (antigenic drift/shift)High (point mutations in target enzymes)
Field DeployabilityPotential for on-site
synthesis and/or delivery
Requires cold chain and skilled personnelRequires global manufacturing/distributionGood for oral
formulations
1 Storage requirements vary by formulation (e.g., lyophilized DNA-based CRISPR does not require cold chain; mRNA-based CRISPR may require −80 °C storage).
Table 2. Selected historical synthetically made viruses.
Table 2. Selected historical synthetically made viruses.
VirusGenomeYearMethodSignificanceReference
Poliovirus(+) ssRNA2002Synthesis of full-length cDNA from oligosFirst de novo synthesis of an infectious virus[3]
ΦX174 BacteriophagessDNA2003Synthesis of full-length cDNA from oligosFirst de novo synthesis of bacteriophage[4]
Influenza A (1918 strain)(−) ssRNA2005Reverse genetics from historical samplesHistorical re-creation; raised DURC/bioethics concerns[6]
Horsepox virusdsDNA2018Commercial DNA fragments assembledSparked global dual-use biosecurity debate; possible roadmap to synthetic smallpox[5]
Bluetongue VirusdsRNA2008In vitro transcribed RNA from cloned cDNAsFirst dsRNA virus reconstructed via reverse genetics[9]
Herpes Simplex Virus dsDNA2017Synthetic genomics assemblyGenome-wide engineering of large DNA viruses[10]
African swine fever virusdsDNA2025BAC generation and helper virus rescueGenome-wide engineering of large DNA virus[11]
Table 3. Summary of CRISPR-based antiviral studies by viral genome.
Table 3. Summary of CRISPR-based antiviral studies by viral genome.
VirusCRISPR SystemViral GenomeDelivery ModalityReferences
SARS-CoV-2Cas13(+) sense ssRNAmRNA, LNP
(in vivo)
[13,21]
Influenza ACas13d(−) sense ssRNAmRNA, LNP
(in vivo)
[13,15]
Dengue virusCas13b(+) sense ssRNATracking translation
(in vivo)
[18]
Zika virusCas13b(+) sense ssRNATracking
translation
(in vivo)
[22]
HIV-1Cas9(+) sense ssRNA; proviral DNA (latent)AAV for provirus excision[23]
ASFVCas9dsDNALipid nanoparticles (LNP)[19]
HSV-1Cas9dsDNAAAV[24,25]
HCMVCas9dsDNAAAV[26]
HBVCas9dsDNAsiP[27]
Table 4. Synthetic virology and viral landmark events in DURC.
Table 4. Synthetic virology and viral landmark events in DURC.
CaseOrganism/
Vector
Impact ConcernReal-World OutcomeReference
Synthetic
Polio Virus
PoliovirusCreation of synthetic virus from genetic sequence dataDemonstrated feasibility of synthesizing viruses[3]
Mousepox with IL-4 GeneMousepox VirusIncreased virulence; overcame vaccine protectionRaised fears about modifying poxviruses[53]
CRISPR Gene DrivesMosquitoes (e.g., malaria vectors)Potential ecosystem disruption; irreversible genetic changesProposals for self-regulation and moratoriums[54]
AI-designed Novel PathogensHypothetical or simulated
“pathogens”
AI used to propose de novo viral blueprintsPolicy discussions on dual use in AI and Biosecurity[55]
1918 Influenza ReconstructionInfluenza A (H1N1)Resurrecting high-virulence pandemic virusReconstruction raised global biosecurity and biosafety debate[6]
H5N1 Gain of FunctionAvian influenza (H5N1)Airborne transmission and immune escapeTriggered policy moratorium and global review of GoF research[52]
AI-designed PhagesBacteriophageAI-enabled synthesis of novel viral genomesDemonstrated AI potential in synthetic biology; raised misuse concerns[2]
Benchtop DNA SynthesisVariousDecentralized access to build novel pathogensHighlighted urgent need for synthetic screening policies[56]
Horsepox Virus SynthesisHorsepox virus (Orthopoxvirus)Mail-ordered DNA;
constructed extinct virus similar to smallpox
Ignited debate over dual-use research and synthetic-biology governance[5]
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

Gladue, D.P.; O’Mahony, A. CRISPR Treatments for AI-Designed Synthetic Viruses: Rapid Programmable Countermeasures for Emerging and Engineered Viruses. Viruses 2025, 17, 1588. https://doi.org/10.3390/v17121588

AMA Style

Gladue DP, O’Mahony A. CRISPR Treatments for AI-Designed Synthetic Viruses: Rapid Programmable Countermeasures for Emerging and Engineered Viruses. Viruses. 2025; 17(12):1588. https://doi.org/10.3390/v17121588

Chicago/Turabian Style

Gladue, Douglas P., and Alison O’Mahony. 2025. "CRISPR Treatments for AI-Designed Synthetic Viruses: Rapid Programmable Countermeasures for Emerging and Engineered Viruses" Viruses 17, no. 12: 1588. https://doi.org/10.3390/v17121588

APA Style

Gladue, D. P., & O’Mahony, A. (2025). CRISPR Treatments for AI-Designed Synthetic Viruses: Rapid Programmable Countermeasures for Emerging and Engineered Viruses. Viruses, 17(12), 1588. https://doi.org/10.3390/v17121588

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop