Subscribe

RSS Feed (xml)

Powered By

Skin Design: Kisi Karunia
Base Code: Free Blogger Skins

Powered by Blogger

Showing posts with label Emerging Cancer Treatments. Show all posts
Showing posts with label Emerging Cancer Treatments. Show all posts

Friday, 28 November 2025

A Flu Virus That Kills Pancreatic Cancer? The Breakthrough Scientists Are Calling a Game-Changer

 

Oncolytic Influenza A Virus as an Emerging Therapeutic Modality for Pancreatic Cancer: A Narrative Review


Pudjiatmoko

Member of the Nanotechnology Technical Committee, National Standardization Agency, Indonesia

 

Abstract

 

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, characterized by aggressive biological behavior, profound desmoplasia, and resistance to almost all existing therapeutic modalities. Oncolytic viruses (OVs) have emerged as a promising class of immunotherapeutic agents capable of inducing selective tumor cell lysis while simultaneously activating antitumor immunity. Among the various OV platforms, engineered influenza A viruses—including strains derived from avian influenza H5—have gained increasing attention due to their natural tropism, genetic flexibility, and strong capacity to stimulate innate immune pathways. This review synthesizes the historical foundations, mechanistic insights, preclinical evidence, and translational challenges of influenza A–based oncolytic virotherapy for pancreatic cancer. Existing studies show that influenza A viruses can directly lyse PDAC cells (Kasloff et al., 2014) and can be engineered to express immunomodulatory payloads, including cytokines and immune checkpoint inhibitors (van Rikxoort et al., 2012; Lei, G. et al, 2023). Recent advances have elucidated the role of cGAS–STING signaling and enhanced cytotoxic lymphocyte infiltration in mediating influenza-based antitumor effects. Despite compelling preclinical data, no influenza-derived OV has yet entered clinical trials for PDAC. Further research is required to optimize tumor selectivity, improve delivery strategies, and overcome stromal and immunologic barriers. This review highlights current progress and proposes future directions to facilitate the translation of influenza-based virotherapy into clinical applications for PDAC.

Keywords: Pancreatic ductal adenocarcinoma; oncolytic viruses; influenza A–based virotherapy; H5 avian influenza; antitumor immunity; cGAS–STING pathway; tumor microenvironment; immunomodulatory payloads; preclinical oncology.

 

1. Introduction

 

Pancreatic ductal adenocarcinoma accounts for over 90% of pancreatic cancer cases and continues to exhibit mortality rates among the highest of all cancers, with a 5-year survival rate remaining below 10%. Its lethality is attributed to late clinical presentation, extensive desmoplastic stroma, profound immunosuppression, and resistance to chemotherapy, radiotherapy, and most immunotherapies (Hamidi-Sofiani et al., 2022; Achim et al., 2025). In response to these challenges, oncolytic viruses have emerged as a distinctive therapeutic modality that combines direct tumor cell lysis with robust immune activation (Lin et al., 2023).


While several OV platforms—such as adenovirus, vesicular stomatitis virus, herpes simplex virus, and vaccinia virus—are undergoing active clinical development, a growing body of evidence suggests that influenza A viruses hold unique advantages. Engineered influenza strains, including those derived from avian influenza H5N1, possess a segmented genome that facilitates genetic manipulation, can potently activate innate immunity, and may be redirected toward tumor tissue (van Rikxoort et al., 2012; Donelan & NCI, 2016).


This review provides a comprehensive narrative synthesis of the historical development, mechanistic rationale, preclinical evidence, and translational opportunities associated with influenza A–based oncolytic virotherapy for pancreatic cancer.

 

2. Methods

 

This narrative review was conducted using peer-reviewed literature published between 2012 and 2025. Searches were performed in PubMed, Scopus, and Web of Science using the terms oncolytic influenza virus, H5N1 engineered virus, influenza virotherapy, and pancreatic ductal adenocarcinoma. Key historical studies published prior to 2020 were included to ensure adequate background contextualization, while publications from 2020–2025 were prioritized to reflect contemporary knowledge and translational developments. Only literature indexed in major scholarly databases and publicly archived was included. All references follow APA citation style.

 

3. Historical Development of Influenza A as an Oncolytic Virus

 

Early observations of respiratory viral infections demonstrated that certain viruses could induce cytopathic effects in malignant cells. These findings laid the conceptual foundation for exploring influenza viruses as potential oncolytic agents. The development of reverse genetics techniques enabled precise engineering of influenza A viruses, including the integration of immunostimulatory genes. A landmark study by van Rikxoort et al. (2012) demonstrated that insertion of interleukin-15 (IL-15) into the NS reading frame increased antitumor immune activation, solidifying the feasibility of arming influenza A with therapeutic payloads.


The National Cancer Institute formally defined an “oncolytic influenza A virus” as a genetically engineered influenza virus capable of selectively infecting and destroying cancer cells (Donelan & NCI, 2016), recognizing its potential as a distinct OV platform. The first major evidence of influenza A’s direct oncolytic activity against PDAC was provided by Kasloff et al. (2014), who demonstrated infection, replication, and tumor growth inhibition in human PDAC xenografts. This study established the rationale for further exploring influenza-based OVs as candidate therapeutics for pancreatic cancer.

 

4. Mechanisms of Action of Influenza-Based Oncolytic Virotherapy

 

4.1 Selective Infection and Lysis of Tumor Cells

Influenza A virus tropism is largely determined by sialic acid receptor specificity. Human PDAC cells express both α2,3- and α2,6-linked sialic acids, which facilitate efficient viral entry (Kasloff et al., 2014). Following infection, the influenza virus undergoes replication and induces lytic cell death, leading to reduced tumor viability. This direct cytopathic effect represents a foundational mechanism of influenza-based OV therapy.


4.2 Induction of Innate Immune Responses

Influenza A viruses robustly activate pattern-recognition receptors, including RIG-I, TLR7, and the cGAS–STING pathway. Activation of these sensors results in the release of type I interferons, chemokines, and inflammatory cytokines that collectively enhance antitumor immunity (Lei, G. et al, 2023). This innate signaling may help counteract the profoundly immunosuppressive microenvironment characteristic of PDAC.


4.3 Activation of Adaptive Antitumor Immunity

Engineered influenza A viruses enhance adaptive immune responses through activation of dendritic cells, increased antigen presentation, and expansion of cytotoxic CD8+ T lymphocytes. Recent studies show that influenza-based OVs engineered to express PD-L1–neutralizing antibodies enhance T-cell infiltration and reverse T-cell exhaustion (Lei, G. et al, 2023). These findings suggest that influenza-derived OVs can potentiate antitumor immunity through coordinated innate and adaptive mechanisms.


4.4 Delivery of Immunomodulatory Payloads

Advances in genetic engineering have enabled influenza A viruses to deliver biologically active therapeutic molecules. These include IL-15 (van Rikxoort et al., 2012), anti–PD-1 or anti–PD-L1 antibodies (Lei, G. et al., 2022), and GM-CSF (Reddy et al., 2024). Such payloads further amplify antitumor immune responses and may synergize with existing immunotherapies.

 

5. Preclinical Evidence in Pancreatic Cancer

 

5.1 Direct Evidence from PDAC Models

The study by Kasloff et al. (2014) remains the principal direct investigation demonstrating that avian influenza A can infect human PDAC cells, replicate efficiently, induce apoptosis, and inhibit tumor growth in xenograft models. Although limited, this foundational evidence confirms that PDAC is permissive to influenza-based OV therapy.

 

5.2 Mechanistically Relevant Studies in Other Solid Tumors (2020–2025)

Since influenza OVs for PDAC are still emerging, supporting mechanistic insights derive from preclinical studies in hepatocellular carcinoma, colorectal cancer, and other solid tumors. These include demonstrations that influenza A viruses expressing PD-L1 antibodies enhance CD8+ T-cell activation via cGAS–STING signaling (Lei, G. et al, 2023), and that anti–PD-1-armed influenza viruses suppress tumor progression and extend survival (Lei, G. et al., 2022). Collectively, these findings provide strong mechanistic support for application in PDAC.

 

5.3 Relevance to the Immunobiology of PDAC

The fibrotic and immunosuppressive microenvironment of PDAC poses substantial barriers to effective immunotherapy. Influenza-based OVs exhibit properties that may overcome these barriers by inducing inflammatory remodeling (Esteves et al., 2025), promoting immunogenic tumor cell death, and enhancing responsiveness to checkpoint blockade therapies (Achim et al., 2025). Accordingly, although direct PDAC studies remain limited, the mechanistic congruence is compelling.

 

6. Combination Strategies for Enhanced Efficacy

 

6.1 Combination with Immune Checkpoint Inhibitors

Influenza A viruses engineered to express anti–PD-1 or anti–PD-L1 antibodies demonstrate superior antitumor efficacy compared with monotherapy (Lei, G. et al., 2022), highlighting the potential for integrated immunomodulation.


6.2 Combination with Cytokine Engineering

Arming influenza A viruses with IL-15 promotes activation of NK cells and CD8+ T cells (van Rikxoort et al., 2012), an advantage particularly relevant for PDAC, which exhibits suppressed NK-cell activity.

 

6.3 Combination with Stromal Modulation Strategies

The dense desmoplastic stroma of PDAC limits viral dissemination. Potential synergistic approaches include co-administration of TGF-β inhibitors, hyaluronidase, or CXCR4 antagonists, which may enhance viral penetration and promote microenvironmental remodeling (Rivers-Orellana et al., 2025).

 

7. Challenges and Limitations

 

7.1 Biosafety and Risk of Pathogenic Reversion

Because influenza viruses possess inherent pathogenicity—particularly avian-derived strains—engineering efforts must incorporate stringent safety features to ensure attenuation in normal tissues and prevent reversion to virulence (Sułek et al., 2025).


7.2 Barriers to Efficient Delivery

The extracellular matrix of PDAC restricts viral distribution, and systemic delivery is further impeded by neutralizing antibodies and interferon responses. These barriers necessitate innovative delivery approaches and improved viral design.


7.3 Lack of Clinical Translation

Despite encouraging preclinical studies, no influenza-based oncolytic viruses have entered clinical trials for PDAC. Major translational gaps include optimization of tumor specificity, reliable biomanufacturing, and development of predictive animal models.

 

8. Future Directions


8.1 Development of Multi-Armed Influenza OVs

Emerging platforms may integrate multiple mechanisms—lysis, checkpoint inhibition, cytokine support, and stromal remodeling—within a single viral vector to maximize efficacy.


8.2 Personalized Virotherapy Based on Tumor Profiling

Advances in neoantigen mapping and receptor profiling may enable individualized influenza A OV designs tailored to specific tumor characteristics (Vorobjeva et al., 2022).


8.3 Integration with mRNA Vaccines or Cell-Based Immunotherapies

Influenza OVs may serve as priming agents to enhance the effectiveness of mRNA vaccines or CAR-T therapies, particularly by reshaping the tumor microenvironment.

 

9. Conclusion


Oncolytic influenza A viruses represent a highly promising yet underdeveloped therapeutic platform for pancreatic ductal adenocarcinoma. Foundational studies demonstrate their capacity for selective tumor cell infection and lysis, while more recent engineering advances have enabled delivery of immunomodulatory payloads and synergy with immune checkpoint blockade. Although substantial mechanistic evidence supports their application in PDAC, barriers related to biosafety, delivery, and translational validation must be addressed. Continued research into genetic optimization, stromal penetration, and microenvironment modulation will be essential for advancing influenza-based oncolytic virotherapy toward clinical evaluation. With ongoing improvements in synthetic virology and immuno-oncology, influenza A OVs hold significant potential to contribute to future therapeutic strategies for PDAC.

 

References

1


1.Achim, E., Pîrlici, E., Cristea, C., & Tertis, M. (2025). Design and efficacy of oncolytic viruses and antitumor vaccines: A dead end in the immunotherapy of pancreatic cancer? International Journal of Molecular Sciences, 26(19), 9640. https://doi.org/10.3390/ijms26199640

2. Chen, L., Huang, Z., & colleagues. (2023). Oncolytic viral therapy: A review and promising future. [Jurnal]. https://thejns.org/view/journals/j-neurosurg/140/2/article-p319.xml.

3.Donelan, S. C., & National Cancer Institute. (2016). Oncolytic influenza A virus. NCI Drug Dictionary. https://www.cancer.gov/publications/dictionaries/cancer-drug/def/oncolytic-influenza-a-virus

4.Esteves, M., & colleagues. (2025). Oncolytic viruses: A novel therapeutic approach for pancreatic ductal adenocarcinoma — current landscape and future directions. Molecular Therapy – Oncology. https://www.cell.com/molecular-therapy-family/oncology/fulltext/S2950-3299(25)00138-9

5.Hamidi-Sofiani, V., Rakhshi, R., Moradi, N., Zeynali, P., Nakhaie, M., & Behboudi, E. (2022). Oncolytic viruses and pancreatic cancer. Cancer Treatment and Research Communications, 31, 100563. https://doi.org/10.1016/j.ctarc.2022.100563

6.Kasloff, S. B., Pizzuto, M. S., Silic-Benussi, M., Pavone, S., Ciminale, V., & Capua, I. (2014). Oncolytic activity of avian influenza virus in human pancreatic ductal adenocarcinoma cell lines. Journal of Virology, 88(16), 9321–9334. https://doi.org/10.1128/JVI.00929-14

7. Lei, G., Li, B., Yang, H., Sun, F., Li, D., Yan, J., Wang, Y., Li, R., Liu, H., Zhang, S., Li, Y., & Yang, P. (2022). Therapeutic efficacy of an oncolytic influenza virus carrying an antibody against programmed cell death 1 in hepatocellular carcinoma. Human Gene Therapy, 33(5-6), 309–317.

8.Lei, G., Li, B., Yang, H., Sun, F., Li, D., Yan, J., Wang, Y., Li, R., Liu, H., Zhang, S., Li, Y., & Yang, P. (2023). A recombinant oncolytic influenza virus expressing a PD-L1 antibody induces CD8+ T-cell activation via the cGAS–STING pathway in mice with hepatocellular carcinoma. Journal of Medical Virology, 95(7), e28854. https://doi.org/10.1002/jmv.28854.

9.Lin, D., Shen, Y., & Liang, T. (2023). Oncolytic virotherapy: Basic principles, recent advances and future directions. Signal Transduction and Targeted Therapy, 8, 156. https://doi.org/10.1038/s41392-023-01407-6

10.Reddy, R., Patel, M. A., Kanojia, D., Hu, P., Zhang, L., & Lesniak, M. S. (2024). Oncolytic viral therapy: a review and promising future directions. Journal of Neurosurgery, 140(2), 319–327. https://doi.org/10.3171/2023.6.JNS23243.

11.Rivers-Orellana, S., Bautista, J., Palacios-Zavala, D., Ojeda-Mosquera, S., Altamirano-Colina, A., Alcocer-Veintimilla, M., Parrales-Rosales, G., Izquierdo-Condoy, J., Vásconez-González, J., Ortiz-Prado, E., Muslin, C., & López-Cortés, A. (2025). Oncolytic virotherapy and tumor microenvironment remodeling: Challenges and prospects. Clinical and Experimental Medicine 25, 256. https://link.springer.com/article/10.1007/s10238-025-01691-2

12.Sułek, M., & colleagues. (2025). Oncolytic viruses and tumor immunotherapy: Status of the art in 2025. Cells, 14(22), 1825. https://doi.org/10.3390/cells14221825

13.van Rikxoort, M., Michaelis, M., Wolschek, M., Muster, T., Egorov, A., Seipelt, J., Doerr, H. W., & Cinatl, J. (2012). Oncolytic effects of a novel influenza A virus expressing interleukin-15 from the NS reading frame. PLoS ONE, 7(5), e36506. https://doi.org/10.1371/journal.pone.0036506

14.Vorobjeva, I. Vorobjeva, & Oleg, P. Zhirnov. (2022). Modern approaches to treating cancer with oncolytic viruses. MIR. Journal 9(1), 91-112. https://www.scienceopen.com/hosted-document?doi=10.18527%2F2500-2236-2022-9-1-91-112

#OncolyticVirus 

#PancreaticCancer 

#InfluenzaA 

#CancerImmunotherapy 

#Virotherapy