Virionics Logo

Pushing the boundaries of Oncology with Chimeric and Synthetic Virotherapy

Powered by ONCORITHMS

Intelligent framework for chimeric and synthetic RNA-based oncolytic virotherapy, capable of automatically generating complete and personalized therapies

Cutting-Edge Technology

Artificial Intelligence

State-of-the-art AI integrated with advanced viral engineering to design modified viruses

Tumor Selectivity

Viruses that selectively infect tumor cells with multiple layers of safety

Integrated Database

Vast database composed of the union of various public databases and scientific portals

Oncolytic Virotherapy

What is Oncolytic Virotherapy?

Oncolytic virotherapy is an innovative therapeutic approach that uses modified viruses to selectively infect and destroy cancer cells while preserving healthy cells.

Our oncolytic viruses infect tumor cells and replicate intracellularly without causing cell lysis, avoiding any risk of contaminating the body, and instead induce tumor cell death from within.

Safety for healthy cells
Specific tumor targeting
Immune system activation

Mechanism of Action

Oncolytic viruses infect tumor cells, replicate, and cause cell lysis, releasing tumor antigens that stimulate immune response

Personalized Oncolytic Virotherapy

Personalized AI
Genetic Profile
Viral Chassis
Unique Therapy

Complete Personalization

Our platform automatically generates complete and personalized therapies based on the specific type of cancer informed, using advanced machine learning algorithms.

Specific Therapeutic Payloads

Immunomodulators, immune checkpoint inhibitors, CD8+ cell ligands, NK and pro-apoptotic proteins selected based on tumor cell biochemical metabolism

Customized Viral Chassis

Replication and immune escape profiles optimized for different tumor microenvironments

Temporal Modulation

Control of action timing (lysis versus programmed apoptosis), adjusting infection dynamics, replication and immune activation

Revolutionary Innovation

Sequential Dual Virotherapy

With the desire to further increase the efficiency of our Virotherapy, we developed an innovative technique using two oncolytic viruses in sequence.

This approach was inspired by the revolutionary work of a virologist who implemented this technique in self-experimentation to treat her own cancer in 2024, obtaining exceptional results.

Dual Therapy Advantages:

  • Efficiency superior to 80%
  • Synergistic action between viruses
  • Reduction of tumor resistance
V1
V2

Optimized Sequence

Two oncolytic viruses working in sequence to maximize therapeutic efficacy and overcome individual limitations

Advanced Molecular Safety

RNA Inhibitor

RNA reading inhibitor in healthy cells, ensuring absolute tumor selectivity

Genetic Logic

Conditional genetic logic (AND/OR gates) for precise control of viral activation

Safety Mechanisms

Multiple layers of molecular safety to ensure safe and effective treatment

Our Comprehensive Framework

Design and Viral Engineering Focus

Chimeric Viral Design Module

  • • Intuitive interface for combining genetic sequences from different viruses
  • • Library of characterized viral genetic "building blocks"
  • • In silico prediction tools for genetic stability and replication capacity
  • • Integration of synthetic elements like genetic switches
  • • Multi-virus sequence incorporation for enhanced potency

Tropism and Selectivity Optimization

  • • Design module for receptor-specific targeting
  • • Options to "deactivate" tropism for normal cells
  • • Modeling tools for virus-cell interaction prediction
  • • Superexpressed receptor targeting in tumor cells

Synthetic Therapeutic Payload Integration

  • • Gene insertion for therapeutic protein expression (immunomodulators, cytokines, monoclonal antibodies)
  • • Design module for gene editing system delivery (CRISPR-Cas9)
  • • Integration with synthetic peptide libraries with antitumor activity

Evaluation and Prediction Focus

In Silico Virus-Tumor Interaction Simulation

  • • Modeling of infection and viral replication dynamics
  • • Prediction of immune response induced by chimeric virus
  • • Virtual efficacy assessment in different cancer types

Safety Prediction Module

  • • Viral sequence analysis for recombination potential
  • • Algorithms for off-target effect probability prediction
  • • Integration with viral toxicity databases

Production and Scalability Optimization

Suggestions for virus design that are more efficient for large-scale production in cell culture systems.

Personalization and Clinical Application Focus

Patient Data Integration Interface

  • • Upload of genomic, transcriptomic and immunological patient data
  • • "Tailored" chimeric virus recommendations based on patient's molecular profile

Combinatorial Therapy Selection Module

  • • Data-based suggestions for synergistic therapies
  • • Immunotherapies and targeted therapies compatibility analysis

Monitoring and Results Analysis Tools

  • • Interface for tracking patient response to treatment
  • • Statistical analysis of results for model refinement
  • • Imaging data and viral/tumor biomarkers integration

Additional Features

Updated Knowledge Base

Integration with scientific and clinical databases on virotherapy

Collaborative Interface

Enabling researchers and clinicians to work together

Regulatory Compliance

Incorporating relevant regulatory guidelines and information

Interactive Visualization

Clear presentation of viral design data and simulations

Our Viral Options Portfolio

List of our viral options that have great affinity with various types of cancer:

Adenovirus (AdVs)

Herpes Simplex Virus (HSV)

Vaccinia Virus (VV)

Reovirus (ReoV)

Newcastle Disease Virus (NDV)

Measles Virus (MeV)

Vesicular Stomatitis Virus (VSV)

Viral Armament Optimization

In addition to virus-specific modifications, our framework also features various strategies that are aligned with the immunological barriers present in the specific tumor, making the virotherapy response as specialized and efficient as possible:

Abolition of Immunosuppression

Tumors frequently escape immune responses via checkpoint ligand expression (PD-L1), recruitment of Tregs and MDSCs, and secretion of immunosuppressive cytokines (TGF-β, IL-10). Engineering tactics include:

Viral Evasion Gene Deletion
  • • ICP47 (HSV)
  • • B18R (VV)
Local Checkpoint Inhibitor Expression
  • • anti-PD-1/PD-L1, anti-CTLA-4
  • • Costimulatory Ligands (CD40L, 4-1BBL)
Cytokine Diversification
  • • DC Maturation
  • • Th1 Polarization
  • • APC Enhancement IFN-α/β
  • • Chemokines (CCL5, CXCL10)
APC Function Enhancement
  • • FLT3L
  • • CD40L TNF Family Transmembrane Protein
  • • GM-CSF
Effector T Cell Response Enhancement
  • • Costimulatory Molecules
  • • Local Checkpoint Blockers
  • • Targeted T Cell Recruitment (BiTEs)

Delivery Improvements

Capsid Engineering and Receptor Redirection

Fiber modifications (AdV): Inserting RGD motifs or scFvs targeting tumor antigens (e.g., HER2) in the fiber knob allows infection of cells with low CAR expression, using alternative receptors (integrins or specific antigens).

Directed Evolution of OVs

Through multiple rounds of controlled error replication, recombination and selection in tumor cell cultures, viral variants with optimized tumor tropism, enhanced replication kinetics and immune evasion profiles are generated.

Protease-Activatable Vectors (PAVs)

Incorporating protease-dependent "locks" (e.g., tetra-aspartic acid motifs flanked by MMP-2/9 sensitive sites) in AAV capsids blocks infectivity until highly active TME proteases release the virus locally.

Personalized Neoantigen Vaccination Integration

Loading OVs with patient-specific neoantigens (identified by tumor exome sequencing) enables personalized intratumoral vaccination, promoting T responses directed to the tumor's unique mutational profile.

Nanocarrier-Mediated Systemic Delivery

Encapsulation of OVs in liposomes, polymeric nanoparticles or extracellular vesicles coated with tumor-homing peptides hides the virus from neutralizing antibodies, increases circulation half-life and favors accumulation in metastases via active targeting (e.g., RGD-modified liposomes).

Challenges and Considerations

Despite advances, several obstacles remain in the clinical translation of enhanced OVs:

Antiviral Neutralization

Problem:

Pre-existing neutralizing antibodies (NAbs) against viral capsids can eliminate systemically administered OVs.

Our Solution:

Pseudotyping with rare serotypes, detargeting via MREs or nanoparticle encapsulation. We possess a vast database on pseudotyping correlated to each cancer we work with, currently 15.

Tumor Heterogeneity and Receptor Expression

Problem:

Variations in receptor expression (CAR, CD46, integrins) between patient tumors reduce OV infectivity.

Our Solution:

Development of multi-ligand or bispecific capsids aims to cover heterogeneous receptor profiles in tumors.