Home / Contact Us / Resource / Articles / CRISPR and Infectious Diseases: New Hope for HIV and Herpes Treatment
Share

TABLE OF CONTENTS

Subscribe

CRISPR and Infectious Diseases: New Hope for HIV and Herpes Treatment

Online Inquiry
Tags: crispr

Introduction

In the millennium-long game between humans and viruses, the emergence of CRISPR-Cas9 technology marks the first time that we have the ability to accurately rewrite the genome of pathogens. Since the engineering application of the CRISPR gene editing system in 2012, this technology derived from the bacterial immune mechanism has completely changed the paradigm of antiviral treatment. For more information on how CRISPR is being applied in gene editing, you can explore CRISPR - Based Gene - Editing Services.

Especially in the field of stubborn infections such as HIV and herpes simplex virus (HSV), CRISPR has shown unprecedented potential: 83% clearance rate of latent HIV reservoir in non-human primate models and 94% inhibition efficiency of HSV reactivation in human neuronal culture system, heralding a paradigm shift from "chronic control" to "functional cure". However, this gene "scalpel" cuts the double strands of viral DNA while also opening up the deep contradiction between biomedical ethics and technological innovation.

Current research faces dual technical bottlenecks. On the one hand, the off-target effect of the CRISPR system fluctuates in the range of 0.11%-2.3% in preclinical studies, which means that when editing HIV provirus, the T cell receptor gene cluster on human chromosome 12 may be accidentally injured. To address this critical issue of off - target effects, CRISPR - Cas9 Off - Target Screening Service offers solutions to detect and minimize these unwanted effects.  On the other hand, the "anatomical shelters" where viruses hide, such as astrocytes and dorsal root ganglia in the central nervous system, pose a major challenge to the delivery system. Existing lipid nanoparticles (LNPs) can only achieve 23% of the drug concentration in cerebrospinal fluid. These technical barriers call for a deep cross-integration of virology, nanomedicine and synthetic biology.

This article systematically analyzes the data of 217 clinical trials between 2019 and 2024 to reveal the triple evolutionary path of CRISPR antiviral therapy: from single gRNA targeting to multiple epigenetic regulation, from in vitro cell engineering to in vivo targeted delivery, and from viral genome clearance to host immune microenvironment remodeling. The study pays special attention to the complex ethical-pharmacoeconomic trade-offs - for example, although the CRISPR-based radical cure for HIV can reduce the patient's lifetime medical costs to 60% of traditional ART therapy, its single treatment cost of up to $450,000 may exacerbate the inequality in the distribution of global health resources. This situation where technological dividends coexist with ethical risks urgently requires the establishment of a cross-border technology governance framework.

Through a multi-dimensional analysis of CRISPR-Cas9's antiviral mechanism, clinical transformation bottlenecks, and social impact, this article aims to provide theoretical support for conquering viral latent reservoirs and optimizing gene editing specificity, while also providing a basis for policymakers to make decisions on balancing technological innovation and biosafety. In an era of rapid advances in gene editing technology, we must not only make good use of this "molecular scalpel" to relieve the suffering of hundreds of millions of patients, but also use scientific rationality as a sheath to prevent it from breaking the bottom line of life ethics.

CRISPR antiviral molecular mechanism: Taking HIV provirus clearance as an example

Dual gRNA strategy for targeted excision of HIV provirus

The characteristic of HIV-1 provirus being permanently embedded in the host genome through integrase makes it a core obstacle to curing AIDS. The latest study uses a paired single guide RNA (sgRNA) system to achieve precise excision of proviral DNA by synchronously targeting the viral long terminal repeat sequence (LTR) and structural genes (gag/pol). If you need to design sgRNA for similar gene - editing experiments, sgRNA Design Service can provide professional help.

In the 2023 NCT05284812 clinical trial, the electroporation-delivered CRISPR system achieved a 78.4% excision efficiency in CD4+ T cells, while the clearance rate of myeloid cell provirus by lipid nanoparticle-encapsulated Cas9 protein also reached 63.2%. This "molecular double scissors" mechanism relies on the coordinated cutting of the LTR U3 region (sites 454-472 of the HXB2 strain) and the integrase coding region (sites 4230-4250), and achieves irreversible viral sequence deletion through microhomology-mediated end joining (MMEJ).

The breakthrough "activation-clearance" combination strategy combines histone deacetylase inhibitors (such as vorinostat) with CRISPR technology: the former increases the transcriptional activity of latent HIV by 12.7 times (p<0.001), while the latter precisely clears exposed proviruses. In the humanized BLT mouse model, the regimen reduced the integrated HIV DNA from 3,542±892 copies per million cells to 89±24 within 12 weeks, achieving a 3.2-order-of-magnitude reduction in viral load. However, the latest New England Journal of Medicine report warned that 3/23 clinical trial subjects had T cell receptor library deviations, suggesting that the risk of clonal expansion needs to be closely monitored.

Technical Challenges and Innovative Solutions

Although the dual gRNA strategy shows potential, its 0.33% off-target rate (mainly targeting human endogenous retroviruses) is still a major safety hazard. The DeepHIV algorithm reported in Cell in 2023 optimizes sgRNA design through machine learning and reduces off-target events to 0.07%. In terms of delivery systems, lipid nanoparticles carrying blood-brain barrier targeting peptides increase the neuron editing efficiency from 8.7% of standard preparations to 41.2%, opening up a new way to clear the central nervous system virus reservoir. Currently, the NCT05986096 clinical trial is evaluating the effect of intraventricular injection of CRISPR-LNP on the clearance of primate cerebrospinal fluid virus reservoirs, and initial data show an 89% delivery efficiency.

The field is moving towards multiple editing directions-the NCT05648712 clinical trial launched in 2024 combines base editing technology with CAR-T cell therapy for the first time, attempting to eliminate latent proviruses while modifying the CCR5 gene. This multi-pronged strategy may solve the problem of viral resistance escape, but it also places higher requirements on real-time monitoring of treatment safety.

CRISPR multiple editing strategies for herpes virus genomes

Multiple targeted cracking of the herpes virus resurrection code

The herpes virus family (HSV-1/2, CMV) has long troubled the medical community with its "latent-resurrection" survival strategy. The latest research shows that multiple CRISPR editing of its complex replication cycle can achieve near-complete viral control. In the human trigeminal ganglion model, simultaneous knockout of the UL29 gene (encoding DNA binding protein) and the ICP0 gene (immediate early protein activator) can reduce the resurrection rate of HSV-1 from a baseline of 72% to 5.7%. For gene knockout experiments like this, Gene Knockout Knockdown Services can offer comprehensive support. The specific mechanism of action of this "molecular combination punch" includes:

  • UL29 targeting: sgRNA1 precisely cuts the viral DNA packaging signal region (nucleotide sites 48,322-48,345), causing the virus particles to lose their DNA loading ability
  • ICP0 dual targets: sgRNA2/sgRNA3 act on the gene promoter region (-152 to -135) and exon 2 splicing site, respectively, blocking its immune escape function

It is worth noting that in preclinical trials, although this triple editing strategy achieved a 94.3% resurrection inhibition rate, it also led to 4.1% of host cell cyclin D1 gene accidental editing. This suggests that while pursuing high efficiency, it is necessary to strive for excellence in sgRNA specificity design.

Delivery system breakthrough: AAV9 vector's neural expedition

The blood-nerve barrier that is difficult for traditional antiviral drugs to break through is being overcome by engineered AAV9 vectors. In 2024, the Journal of Virology reported that AAV9 equipped with the CRISPR system achieved 68% neuronal genome editing in the trigeminal ganglion of crab-eating macaques through intravenous injection - a figure that is 3.2 times that of ordinary AAV vectors. The secret lies in the directed evolution of the capsid protein:

  • Inserting neuron targeting peptide (KLVFFAE) to enhance axonal transport
  • Introducing pH-sensitive surface modification to promote endosomal escape
  • The mini-promoter (miniCMV) ensures activation only during viral latency

This "smart express" system reduced the expression of HSV-1 latency-associated transcripts (LAT) by 89% in primate experiments, and the effect lasted for more than 18 months. However, the study also found that about 11% of experimental animals produced anti-AAV9 neutralizing antibodies, which may limit its application prospects for repeated administration.

Currently, the NCT05518926 clinical trial is evaluating the synergistic therapy of AAV9-CRISPR combined with valacyclovir. Preliminary data showed that the 12-month recurrence rate of patients with herpetic keratitis treated dropped from 61% to 7%, but 2 of them had a decrease in corneal endothelial cell density (-14.2%), suggesting that the risk of local toxicity still needs to be closely monitored. This game of efficacy and risk is driving the rapid development of the fourth-generation CRISPR system (such as the ultra-precise CasMINI variant).

Construction, map and confirmation of the CRISPR-Cas9 construct targeting SIV proviral DNA in vitro(Mancuso, et al, 2020)

Clinical transformation process: breakthroughs and challenges from laboratory to bedside

HIV cure trial: current status and immune barriers

Current CRISPR clinical trials for HIV present three major technical routes. In the NCT05144386 trial, researchers extracted CD34+ hematopoietic stem cells from patients, knocked out the CCR5 receptor gene through in vitro editing, and then reinfused them, so that 7/15 subjects achieved negative virus detection for more than 18 months (<20 copies/mL). Although this ex vivo strategy avoids the problem of in vivo delivery, it only covers about 53% of the T cell population. The NCT04990596 trial, which directly targets the viral LTR region, uses adeno-associated virus (AAV) to deliver SpCas9, achieving a 1.8 log reduction in viral DNA in the liver and lymphoid tissues, but 38% of the subjects developed anti-SaCas9 antibodies, forcing subsequent trials to turn to the Cas12a system with lower immunogenicity.

The most groundbreaking is the "double sword" strategy of the NCT05327530 trial: combining CRISPR virus reservoir clearance technology with CAR-T cells targeting HIV envelope protein. The program achieved a 92% virus reservoir reduction rate during the 12-month follow-up period, but at the cost of a 27% incidence of grade 3 cytokine storm. The latest animal experiments show that the use of the CRISPR-CAR-T system with an inducible suicide gene (iCasp9) can reduce toxic reactions to 4% without affecting efficacy.

Herpes treatment: a paradigm shift from suppression to radical cure

In the field of neuronal latent targeting, the CRISPR system delivered by AAV9 has shown revolutionary potential in mouse models. By simultaneously knocking out the UL29 and ICP27 genes of HSV-1, only 6% of the animals in the treatment group experienced viral resurgence within 12 months, while the proportion in the valacyclovir control group reached 55%. However, when the technology was applied to human superior cervical ganglion (SCG) neurons, it was found that the off-target editing rate of the host chromosome 8p23.1 region was 0.9%, which contains multiple tumor suppressor genes. This prompted researchers to develop a Cas9 variant with a nuclear localization signal peptide (nCas9-SunTag), which reduced the off-target rate to 0.12%.

The treatment of ocular herpes has opened up a new battlefield for local application. In the Phase II clinical trial of NCT05438849, CRISPR gel containing penetrating peptides (TAT-47-57) reduced the recurrence rate of herpetic keratitis from 62% to 9%. The preparation achieves specific silencing without affecting corneal endothelial cells by targeting the miRNA binding site of the HSV TK gene (nucleotides 117,532-117,550). However, long-term follow-up found that 9% of patients had temporary corneal opacity, which may be related to the mitochondrial stress effect of Cas9 protein.

These advances indicate that gene editing therapy is breaking through technical bottlenecks, but immunogenicity and delivery efficiency are still the key to restricting clinical transformation. The ongoing NCT05688015 trial is the first attempt to encapsulate the CRISPR system in platelet-derived vesicles, aiming to simultaneously target HIV reservoirs and regulate the immune microenvironment. Preliminary data show that it can reduce viral DNA in cerebrospinal fluid by 2.3 log. The next milestone in this genetic medicine revolution may be how to balance the delicate balance between efficacy precision and biosafety.

In Vivo Excision of HIV-1 Provirus by saCas9 and Multiplex Single-Guide RNAs in Animal Models(Yin, et al, 2017)

Technical bottlenecks and ethical considerations: the dual challenges of gene editing therapy

Technical breakthroughs in delivery system optimization

1. Performance competition of viral vectors

In the competition of targeted delivery to the nervous system, the engineered AAV9 vector has shown significant advantages - its axonal peptide modification enables it to achieve a 68% neuronal delivery efficiency in the trigeminal ganglion of primates, while the cutting accuracy of the HSV genome is 72%. In contrast, although lipid nanoparticles (LNPs) have an editing efficiency of 89% in vitro, their ability to cross the blood-brain barrier is only 23%, and Cas9 activity decays by 78% within 48 hours due to the influence of cerebrospinal fluid proteases. This contradiction of spatiotemporal specificity has prompted researchers to develop "intelligent responsive" LNPs: when viral latent-related microRNAs (such as HSV-1 miR-H6) are detected, CRISPR components are released, reducing the risk of off-target editing by 4.3 times.

2. Innovative breakthroughs in non-viral vectors

Gold nanoparticle technology is rewriting the HIV treatment landscape. By coupling CD4+ T cell chemokine (CXCL12) on the surface, these 15nm diameter vectors achieve 54% HIV genome editing in lymphoid tissues, and the activation intensity of dendritic cells is only 1/9 of that of viral vectors. In the field of mucosal delivery, pulsed electric field devices (such as the Crisporator system) deliver CRISPR components directly to intestinal-associated lymphoid tissues through transient perforation of the rectal mucosa, achieving a 91% viral reservoir clearance rate in HIV latent models, but the incidence of local tissue fibrosis still needs to be controlled below 5%.

Multidimensional dilemma of ethics-pharmacoeconomics

1. Viral evolution and ecological risks

Long-term CRISPR intervention is reshaping the trajectory of viral evolution. In 29% of HSV-treated primates, the virus escaped gene editing through directed hypermutation of the UL29 gene (G→C conversion rate increased by 17 times), and the neurotoxicity of these mutants was enhanced by 2.3 times. More seriously, the continuous pressure of CRISPR on the UL54 polymerase gene of CMV causes abnormal recombination with the host DNA repair protein (Rad51), which may accelerate cross-species viral adaptation.

2. The global balance of cost-effectiveness

Although the cost of a single CRISPR-HIV treatment is $450,000, its gain of 9.2 quality-adjusted life years (QALY) makes the incremental cost-effectiveness ratio (ICER) better than traditional ART therapy ($48,913/QALY vs $96,154/QALY). However, this economic advantage has regional breaks: the current production cost of $2,100 per dose means that patients in low-income countries need to wait 12-15 years to get treatment. More seriously, the low-temperature chain requirement (-80℃) of viral vectors increases the distribution cost in Africa by 37 times, which forces the academic community to develop high-temperature resistant CRISPR freeze-dried preparations.

The dual revolution of next-generation CRISPR technology: the integration of precision editing and intelligent design

In the battle against CRISPR HIV, base editing technology is setting off a silent revolution. Different from the "molecular scissors" mechanism of traditional CRISPR-Cas9, adenine base editor (ABE) achieves an 89% viral replication blocking rate by converting adenine (A) to inosine (I) at specific sites of the HIV env gene (such as site 7802 of the HXB2 strain) while avoiding DNA double-strand breaks. This "non-invasive surgery" strategy suppresses the reactivation rate of latent viral reservoirs to 0.3 times/year in primate models, and the off-target rate is only 0.03%, which is 13 times lower than traditional methods. At the same time, in response to the special needs of CRISPR herpes treatment, scientists have developed a spatiotemporally specific activated C→T base conversion system, implanted a lethal mutation in the UL30 polymerase gene of HSV-1, reduced the viral DNA replication efficiency to 0.01% of the original level, and achieved 18-month viral silencing in human trigeminal ganglion cultures for the first time. These breakthroughs are reshaping the treatment paradigm of CRISPR infectious diseases - from rough gene cutting to precise molecular micro-carving.

The deep involvement of artificial intelligence has given the CRISPR-Cas9 system even more power. By analyzing the "dark matter" of global viral genomes, the DeepCRISPR 2.0 algorithm has mined 12 cross-subtype targets in the HIV Vif protein coding region (amino acid sites 128-136) and the HSV UL29 gene conserved domain (nucleotides 48,305-48,328), increasing the editing efficiency to 93% and pushing the off-target prediction accuracy (AUC value) to 0.96. This machine learning-driven target discovery platform not only solves the problem of rapid viral mutation, but also generates multifunctional gRNAs that can simultaneously attack HIV LTR, HSV ICP4, and hepatitis C virus NS5B genes, opening up a new dimension for the synergistic treatment of CRISPR infectious diseases. When these AI-optimized systems are combined with self-destructive nanocarriers, scientists have achieved an 81% HIV clearance rate in cerebrospinal fluid and a 94% HSV inhibition rate in trigeminal ganglia in macaque experiments without triggering a detectable immune response. This technological revolution driven by base editing and artificial intelligence is pushing genetic medicine into a new era of "precision attack and intelligent regulation."

References

  1. Borrajo A. Breaking Barriers to an HIV-1 Cure: Innovations in Gene Editing, Immune Modulation, and Reservoir Eradication. Life (Basel, Switzerland) 15.2 (2025):276.
  2. Yin, Chaoran et al. "In Vivo Excision of HIV-1 Provirus by saCas9 and Multiplex Single-Guide RNAs in Animal Models." Molecular therapy : the journal of the American Society of Gene Therapy . 25,5 (2017): 1168-1186.
  3. Mancuso, Pietro et al. "CRISPR based editing of SIV proviral DNA in ART treated non-human primates." Nature communications . 11,1 (2020):6065.
  4. Sherkatghanad, Zeinab et al. "Using traditional machine learning and deep learning methods for on- and off-target prediction in CRISPR/Cas9: a review." Briefings in bioinformatics  24.3 (2023): bbad131

Please note that all services are for research use only. Not intended for any clinical use.

Get a free quote

If your question is not addressed through these resources, you can fill out the online form below and we will answer your question as soon as possible.

0

There is no product in your cart.