Creative Biolabs' E2F Promoter-driven Oncolytic Adenovirus Service revolutionizes cancer therapy with specific, potent anti-tumor solutions. Engineered for selective replication in pRB-deregulated cancer cells, our adenoviruses lyse tumors while sparing healthy tissues.
[Discover How We Can Help - Request a Consultation Today]
Fig.1 Schematic representation of the possible involvement of E2F in cell cycle regulation.1,3
E2F transcription factors regulate cell cycle progression, DNA synthesis, and apoptosis. In normal cells, pRB binds E2F to repress cell division genes. However, most human cancers feature deregulated pRB pathways, leading to uncontrolled E2F activation that drives malignant proliferation. Early research into adenovirus E1A-mediated gene control revealed how viral proteins exploit E2F for replication.
The core principle behind E2F Promoter-driven Oncolytic Adenoviruses lies in their ability to exploit the specific molecular vulnerabilities of cancer cells. These viruses are engineered such that the expression of their essential early genes, particularly E1A and E4, is placed under the transcriptional control of the human E2F1 promoter.
In normal cells with an intact pRB pathway:
In cancer cells with a disrupted pRB pathway and deregulated E2F activity:
Furthermore, some advanced oncolytic adenoviruses might also incorporate strategies to modulate the tumor microenvironment (e.g., by expressing therapeutic cytokines) to enhance anti-tumor immune responses, providing a dual mechanism of action.
| Required Starting Materials | Initial Consultation & Project Design |
|---|---|
|
Scientists engineer oncolytic adenovirus vectors by integrating E2F1 promoter for pRB dependency. Post-engineering, large-scale production, purification, and QC ensure high-titer, pure vectors with genomic integrity for immediate use. |
| Vector Production | In Vitro Characterization |
| Following vector engineering with E2F1 promoter integration and E1A-CR2 deletion, we conduct large-scale viral production, purification, and rigorous QC to ensure high-titer, pure vectors with intact genomes. | Through conduct in vitro assays on tumor/normal cell lines to evaluate viral replication, tumor-selective cytotoxicity, gene expression (E1A/E4), and host cell cycle. Results provide robust data on the virus' oncolytic selectivity and potency. |
| In Vivo Efficacy & Safety Evaluation | Data Analysis & Strategic Recommendations |
| We administer oncolytic adenoviruses systemically, monitoring tumor growth inhibition, survival, and intratumoral replication. Safety assessments include body weight, liver enzymes, and organ histology to confirm low systemic toxicity, providing in vivo proof-of-concept. | Our bioinformatics and oncology specialists meticulously analyze all data, providing comprehensive reports with raw data, results, statistics, and figures. Leveraging decades of expertise, we also offer strategic recommendations for preclinical optimization or clinical development. |
| Final Deliverables | Estimated Timeframe |
|
The typical timeframe for this service ranges from 8 to 14 weeks, depending on the complexity of the viral construct, the inclusion of optional preclinical evaluation, and the specific requirements of your project. More intricate designs or extensive in vivo studies may extend the duration. |
[Contact us to get more details]
our service is designed to provide comprehensive, custom-tailored solutions for your cancer research and therapeutic development. Our expertise ensures you receive a product and a partnership that drives success:
[Experience the Creative Biolabs' Advantage - Get a Quote Today]
The use of E2F promoter-driven oncolytic adenoviruses in preclinical models and pRB-deregulated cancer cell lines enhances tumor-selective cytotoxicity. Studies validate its broad potential for solid tumors, emphasizing the promoter's reliance on the universal pRB pathway defect in cancer.
| Western Blot | Cell Viability |
|---|---|
|
|
| Fig.2 E1A expression in tumor cells infected with an oncolytic adenovirus driven by the E2F promoter.2,3 | Fig.3 MTT assay is used to detect the effect of oncolytic adenovirus on the activity of tumor cells.2,3 |
| Cytotoxicity | Tumor Volume |
|
|
| Fig.4 Cytotoxicity is assessed by crystal violet staining.2,3 | Fig.5 An E2F promoter-driven oncolytic adenovirus retards tumor growth in mice.2,3 |
| Cell Cycle | |
|
|
| Fig.6 The effect of E2F promoter-driven oncolytic adenovirus on tumor cell cycle was determined using flow cytometry.2,3 | |
[Contact Our Team to Discuss Your Project]
A: Our services are designed to target virtually any human cancer with a disrupted retinoblastoma (pRB) pathway, which includes the vast majority of solid tumors. This broad applicability is a significant advantage over many therapies.
A: Our oncolytic adenoviruses work as potent monotherapies via direct cancer cell lysis. Their tumor-selective replication also makes them ideal for combinations-synergizing with chemo, radiation, or immune checkpoint inhibitors to boost anti-tumor effects.
A: The key advantage of E2F promoter control is leveraging the universal pRB pathway deregulation in cancer. Unlike tissue-specific promoters, E2F-driven selectivity enables broad tumor applicability and robust replication, ensuring a superior therapeutic index.
A: Though designed for low systemic toxicity, biological therapy considerations include pre-existing anti-adenoviral immunity and optimal delivery for tumor transduction. Our preclinical testing and viral immunology expertise mitigate these challenges.
A: E2F promoter-driven oncolytic adenoviruses excel over non-replicating vectors/gene therapies via self-amplification in tumors. Unlike single-payload delivery, they continuously produce virions to spread and amplify anti-tumor effects. Targeting core oncogenic pathways, this approach offers superior precision and safety versus non-specific treatments.
Creative Biolabs leads in advanced oncolytic virotherapies. Our E2F Promoter-driven Oncolytic Adenovirus Service demonstrates scientific innovation, offering precise, safe, and effective cancer targeting. By leveraging cancer cell biological defects, we enhance tumor selectivity, reduce systemic toxicity, and unlock new therapeutic possibilities.
[Connect for an in-depth discussion]
References