1. Home
  2. Community
  3. Promotions
  4. AAV Innovation 100 Plan

AAV is a widely used delivery vector in gene therapy, and the modification of AAV capsid proteins is beneficial for ensuring the safe and effective delivery of target genes to the human body. Cyagen's AI-AAV platform revolutionizes gene therapy by addressing industry hurdles through efficient screening for multi-target AAV mutants used in gene therapy drugs. Cyagen's established artificial intelligence (AI) model, powered by deep learning, enables targeted predictions for the brain via intravenous injection or intrathecal injection, whole-eye expression and retinal penetration via intravitreal injection. Compared to traditional directed evolution, AI-assisted screening of AAV has achieved multidimensional breakthroughs by providing high yield, high tissue targeting, and superior sequence quantity/quality in less time—overcoming current bottlenecks in gene therapy.

Leveraging the advantages of the AI-AAV platform, empowered by our AI-screening for potential AAV mutants, Cyagen has launched the "AAV Innovation 100 Plan." This initiative aims to develop over 100 new AAV capsids for gene therapy in neurology and ophthalmology, seeking to help preclinical research industry partners identify novel vectors that work well in non-human primates (NHPs)—and could provide greater efficacy in treating human conditions.

We hope to collaborate with global research institutions and companies to combine the technological advantages of AI-screened innovative AAV mutants with partner validations in NHP models. Together, we aim to develop safe, efficient, and clinically suitable gene therapy vectors.

AAV Innovation 100 Plan
Researchers only need to pay a nominal fee (around $100K for ophthalmological vectors [AAV2 Variants] and $200K for neurological vectors [AAV9 Variants]) to acquire 50% of the IP rights for the new AAV capsid sequences and will receive a sufficient quantity of the virus (1-5E12vg for ophthalmological vectors and 1E13-3E14vg neurological vectors) for NHP experimental validation free of charge.
Should the in vivo NHP validation prove successful, researchers may opt to purchase the remaining 50% of the IP rights.
If the NHP in vivo validation fails, we will offer a certain amount of virus packaging services free of charge.
Join Cyagen's AAV Innovation 100 Plan
We promise

1. Each client's unique novel AAV capsid sequence (new variants based on AAV2 or AAV9 backbone), with proprietary intellectual property (IP) rights.

2. The viral yield of AAV variants can meet the needs of animal experiments and industrial production.

3. Our AAV capsid variants can achieve similar or even better infection effects in mice compared to top AAV capsid variant sequences globally, such as AAV2.7MB, AAV-PHP.eB, or other control viruses specified by the client.

4. The capsid sequences and validation data can be inspected by third-party institutions.

AAV9 Capsid Variants
AAV2 Capsid Variants
AAV9 capsid variants, PM167 and PM170, selected through AI screening, were administered via tail vein injection at a low dosage in C57BL/6J mouse models. Data indicate their strong infectivity in various neural tissues, efficient transduction of the central nervous system, and liver de-targeting of the liver. These variants are suitable for both basic and applied research in various neurodegenerative diseases.
Group Virus Expression Capacity in Different Brain Regions Order
Hippocampus Cortex Corpus Callosum Midbrain Spinal Cord Liver
Wild-type Control AAV9 1 1 1 1 1 1
Positive Control AAV9.phpeB 13.2 17.3 12.8 20.2 18.7 0.16
Novel AAV9 Capsid Variant 1 PM167 18.5 19.2 16.4 23.2 24.4 0.25
Novel AAV9 Capsid Variant 2 PM170 8.6 13.2 5.9 25.3 19.7 0.14
AAV2 capsid variants selected through AI screening were injected into the vitreous cavity of C57BL/6J mice. Data indicate that these viral variants exhibit strong infectivity in the eyes, making them suitable for basic and applied research in various retinal diseases.
Group Virus Retinal Back Layer
Penetration Capability
Whole-Eye
Expression Capability
Order
Wild-type Control AAV2-WT 0 1
Positive Control AAV2.7M8 1 2
Novel AAV2 Capsid
Variant 1
PM077 3 10
Novel AAV2 Capsid
Variant 2
PM021 1.5 12.5
Novel AAV2 Capsid
Variant 3
PM054 10 15
In addition to the trial samples mentioned above, Cyagen can provide a wide range of services, including AAV vector design and construction, virus packaging, purification, expression analysis, and functional validation, aimed at significantly reducing the experimental timeline for our customers. Various titer options are available, and we employ rigorous purification processes to effectively achieve the expression of exogenous human/mouse ORFs, shRNA, lncRNA, and CRISPR/gRNA.
Service Specification Titer Timeline Order
Adeno-Associated Virus
(AAV) Packaging
1×10¹² GC ≥5×10¹² GC/ml As Fast As 3 Weeks
2×10¹² GC ≥5×10¹² GC/ml
5×10¹² GC ≥1×10¹³ GC/ml
1×10¹³ GC ≥1×10¹³ GC/ml
2×10¹³ GC ≥1×10¹³ GC/ml
Others Others
*Additionally, we can offer AAV viruses in other specifications, as well as packaging services for other virus types such as lentivirus and adenovirus. Please feel free to reach out to us at 800-921-8930 or email us at animal-service@cyagen.com for further inquiries.
In addition to the trial samples mentioned above, Cyagen can provide a wide range of services, including AAV vector design and construction, virus packaging, purification, expression analysis, and functional validation, aimed at significantly reducing the experimental timeline for our customers. Various titer options are available, and we employ rigorous purification processes to effectively achieve the expression of exogenous human/mouse ORFs, shRNA, lncRNA, and CRISPR/gRNA.
Service Specification Titer Timeline Order
Adeno-Associated Virus (AAV)
Packaging
1×10¹² GC ≥5×10¹² GC/ml As Fast As 3 Weeks
2×10¹² GC ≥5×10¹² GC/ml
5×10¹² GC ≥1×10¹³ GC/ml
1×10¹³ GC ≥1×10¹³ GC/ml
2×10¹³ GC ≥1×10¹³ GC/ml
Others Others

*Additionally, we can offer AAV viruses in other specifications, as well as packaging services for other virus types such as lentivirus and adenovirus. Please feel free to reach out to us at 800-921-8930 or email us at animal-service@cyagen.com for further inquiries.

  Validation Data
AAV9 Capsid Variants   
AAV2 Capsid Variants   
Liver De-Targeting

Cyagen has developed an AI platform that integrates big data, cloud computing, machine learning, and other technologies to optimize the AAV9 capsid protein. This has generated a large number of candidate variants. The results show a high level of confidence in the predicted liver de-targeting data (Figure 1), with a PearsonR correlation coefficient as high as 0.884.

Figure 1. Predicted Liver Infection Efficiency

Top sequences selected from AI-predicted sequences were individually validated by tail vein injection in mice (5E11 vg/each) and examined after 21 days. In vivo imaging results (Figure 2) demonstrate that PM167 exhibits significantly better liver de-targeting than PHP.eB, while PM170 shows significantly better liver de-targeting than AAV9 wild-type (WT) and slightly higher than PHP.eB.

Figure 2. In Vivo Validation of Liver Infection Efficiency (In Vivo Imaging - Luc)

After tail vein injection in mice (5E11 vg/each) and a 21-day incubation period, frozen section results (Figure 3) reveal that PM167 exhibits significantly lower green fluorescent protein signals in the liver compared to PHP.eB. PM170, on the other hand, shows slightly higher green fluorescent protein signals in the liver than PHP.eB but still significantly lower than AAV9 wild-type (WT).

Figure 3. In Vivo Validation of Liver Infection Efficiency (Frozen Section EGFP)
High Targeting to the Central Nervous System

Cyagen has developed an AI platform that integrates big data, cloud computing, machine learning, and other technologies to optimize the AAV9 capsid protein. This has generated a large number of candidate variants. The results show a high level of confidence in the predicted central nervous system targeting data (Figure 4), with a PearsonR correlation coefficient as high as 0.843.

Figure 4. Predicted Central Nervous System Infection Efficiency

Top sequences selected from AI-predicted sequences were individually validated by tail vein injection in mice (5E11 vg/each) and examined after 21 days. In vivo imaging results (Figure 5) show a high accumulation of Top sequences in the brain. The Luc signal intensity in the brain expressed by PM167 is approximately two times that of PHP.eB, while the Luc signal intensity in the brain expressed by PM170 is approximately 1.5 times that of PHP.eB.

Figure 5. In Vivo Validation of Brain Infection Efficiency (In Vivo Imaging, Luc)

To further investigate the distribution of Top sequences in different regions of the central nervous system, we conducted another examination in mice after tail vein injection (5E11 vg/each) and a 21-day incubation period. Frozen section results (Figure 6) demonstrate that PM167 exhibits significantly higher green fluorescent protein signals than PHP.eB in various brain regions (cortex, corpus callosum, hippocampus, midbrain) and the spinal cord. PM170 also shows higher green fluorescent protein signals than PHP.eB in various brain regions and the spinal cord, except for the corpus callosum.

Cortex
Corpus callosum
Hippocampus
Midbrain
Spinal cord
Figure 6. In Vivo Validation of Brain and Spinal Cord Infection Efficiency (Frozen Section EGFP)

Top sequences selected from AI-predicted sequences were pooled in equal ratios into a single test article and used to deliver barcoded transgene reporters respectively. We injected the mixed test article (5E12 vg total) into the cisterna magna of a non-human primate (♀, 3.6 kg). Following 16 days in-life, animal was sacrificed and tissues were processed for next generation sequencing (NGS) and and histology.

Figure 7. In Vivo Validation of Central Nervous
System Infection Efficiency (NGS)
Figure 8. In Vivo Validation of Central Nervous
System Infection Efficiency (Frozen Slice EGFP)

 

Using the Cyagen AI-AAV platform, we constructed a high-capacity mutant plasmid library, packaged a virus library, and performed NGS sequencing. We built a DualConvLSTM network to establish an AAV2 production prediction model. The model's credibility was validated on the test set, achieving a high correlation with Pearson=0.929 and Spearman=0.859 (Figure 9). Additionally, the AI-generated retinal targeting model showed a correlation of Pearson=0.874 and Spearman=0.871 on the test set (Figure 10).

Figure 9. Correlation of the Production Prediction Model on the Test Set
Figure 10. Correlation of the Retinal Targeting Model on the Test Set

We used the production model and the retinal targeting AI model to predict variants with high production and expression capabilities. We selected the top sequences and constructed RC mutant plasmids. These plasmids were separately packaged with wild-type AAV2 plasmids and 7M8 plasmids to produce Luciferase viruses. After virus packaging, purification, and QPCR titer testing, all three variants showed higher yields compared to AAV2 and AAV2.7M8. Specifically, PM054 had the highest yield, which was 3.48 times that of AAV2. PM021 and PM077 had yields 1.5 times and 2.01 times that of AAV2, respectively.

Figure 11. AI Predicted AAV2 Variant Virus Yields

The packaged Luciferase viruses were injected into the vitreous cavity of mice at a dosage of 3E+9 vg (viral genomes) per eye. After 3 weeks, Luciferase expression was detected using both in vivo imaging (Figure 12) and chemiluminescence assays. The in vivo imaging results showed that the signal intensity of Luciferase for all three variants was higher than that of AAV2 and AAV2.7M8.

For a more precise quantification, mice were euthanized, and their eyeballs were collected and homogenized for chemiluminescence detection. The PM054 variant exhibited the highest Luciferase expression level, which was 15 times that of AAV2. PM021 and PM077 had Luciferase expression levels of 12.5 times and 10 times that of AAV2, respectively.

Figure 12. In vivo Validation of Whole Eye Expression Capability (In Vivo Imaging)
Figure 13. In vivo Validation of Whole Eye Expression Capability (Chemiluminescence)

To further validate the in vivo infection efficiency of the variants and explore the cell types infected, EGFP viruses were packaged and injected into the vitreous cavity of mice (3E+9 vg per eye). After 3 weeks, the overall EGFP expression was detected through fundus fluorescence photography, and eyeball samples were collected for pathological examination. The fundus photography results (Figure 14) showed that the GFP signal of PM054 was the strongest, and the fluorescence signals of all three variants were significantly higher than those of AAV2 and AAV2.7M8.

Figure 14. In vivo Validation of Whole Eye Infection Capability (Fundus Photography)

DAPI staining was performed on frozen sections of eyeballs, and the results showed that the infection range of all three variants was greater than that of AAV2-WT and AAV2.7M8. In particular, PM021 and PM054 could infect almost the entire retinal area. In terms of the infection depth, AAV2-WT only infected the RGC layer cells, while AAV2.7M8 had some penetration capability, infecting a small number of optic nerve cells in the posterior retina. All three variants exhibited greater penetration capability than AAV2-WT and AAV2.7M8, infecting cells in various layers of the retina from RGC to PRC. Among them, PM054 had the best infection rate and expression in optic nerve cells, with an infectivity in the posterior retina approximately 10 times that of AAV2.7M8.

Figure 15. In vivo Validation of Whole Eye Expression Capability and Retinal Posterior Layer Penetration Ability
  Platform Advantages
AI-Powered Capabilities
Leveraging in-house AI technology and wet lab methods for rapid design of highly targeted, highly expressed, and high-yield AAV capsids, reducing trial-and-error costs.
Extensive Virus Library
Currently holding libraries of 5 neuro-related and 6 ophthalmology-related variants, along with a variety of tissue-specific promoter ready-to-use vectors for quick virus delivery in as little as 2 weeks.
Stable Validation Data
Viruses are rigorously validated for specificity and transduction efficiency through in vitro cell cultures and in vivo mouse models by our in-house technical team.
Comprehensive Services
From innovative AAV virus packaging aided by AI screening to downstream efficacy evaluations, our gene therapy CRO service platform offers full support, ensuring the coherence of project research data within the same system.