2,115
Views
2
CrossRef citations to date
0
Altmetric
Original Research

Effective intravenous delivery of adenovirus armed with TNFα and IL-2 improves anti-PD-1 checkpoint blockade in non-small cell lung cancer

, , , , , , , , , , , , , , , , , , , & ORCID Icon show all
Article: 2241710 | Received 14 Jun 2023, Accepted 24 Jul 2023, Published online: 02 Aug 2023
 

ABSTRACT

Lung cancer remains among the most difficult-to-treat malignancies and is the leading cause of cancer-related deaths worldwide. The introduction of targeted therapies and checkpoint inhibitors has improved treatment outcomes; however, most patients with advanced-stage non-small cell lung cancer (NSCLC) eventually fail these therapies. Therefore, there is a major unmet clinical need for checkpoint refractory/resistant NSCLC. Here, we tested the combination of aPD-1 and adenovirus armed with TNFα and IL-2 (Ad5-CMV-mTNFα/mIL-2) in an immunocompetent murine NSCLC model. Moreover, although local delivery has been standard for virotherapy, treatment was administered intravenously to facilitate clinical translation and putative routine use. We showed that treatment of tumor-bearing animals with aPD-1 in combination with intravenously injected armed adenovirus significantly decreased cancer growth, even in the presence of neutralizing antibodies. We observed an increased frequency of cytotoxic tumor-infiltrating lymphocytes, including tumor-specific cells. Combination treatment led to a decreased percentage of immunosuppressive tumor-associated macrophages and an improvement in dendritic cell maturation. Moreover, we observed expansion of the tumor-specific memory T cell compartment in secondary lymphoid organs in the group that received aPD-1 with the virus. However, although the non-replicative Ad5-CMV-mTNFα/mIL-2 virus allows high transgene expression in the murine model, it does not fully reflect the clinical outcome in humans. Thus, we complemented our findings using NSCLC ex vivo models fully permissive for the TNFα and IL-2- armed oncolytic adenovirus TILT-123. Overall, our data demonstrate the ability of systemically administered adenovirus armed with TNFα and IL-2 to potentiate the anti-tumor efficacy of aPD-1 and warrant further investigation in clinical trials.

Authors’ contributions

  • TVK, JC, DCAQ, IAKL, RH, CH, VCC, JMS, SS, OH, AK, EWV, and AH designed experiments.

  • TVK, JC, SP, DCAQ, IAKL, SGVK, VA, and EA conducted experiments.

  • JR, II, KB, and ES and collected the patient-derived samples;

  • TVK, JC, SP, and MIM analyzed the results;

  • All the authors contributed to writing and reviewing the manuscript.

Acknowledgments

We thank Minna Oksanen and Susanna Grönberg-Vähä-Koskela for expert experimental and administrative assistance. We also thank Mikko Räsänen, Lotta Ahlfors and Laboratory Animal Center (LAC, University of Helsinki, Helsinki, Finland), Biomedicum Flow Cytometry Unit (University of Helsinki, Helsinki, Finland) and Biomedicum Imaging Unit (University of Helsinki, Helsinki, Finland). Open access funded by the Helsinki University Library (University of Helsinki, Finland).

Disclosure statement

AH is a shareholder in Targovax ASA. AH, JC, JMS, VCC, RH, SS, and DCAQ are employees and shareholders of TILT Biotherapeutics, Ltd. The authors declare no conflicts of interest.

Availability of data and material

Upon reasonable request.

Ethics statement

All animal experiments described in the paper were approved by the Provincial Government of Southern Finland and the Experimental Animal Committee of the University of Helsinki (license number ESAVI/12559/2021).

Cancer samples were collected from patients undergoing surgical resection at Helsinki University Central Hospital (HUS, Helsinki, Finland). Sample collection was approved by the HUS Ethics Committee (47§/17.3.2021; HUS/552/2021), and study permits were obtained (17.05.2021; reference number HUS/259/2021). Written informed consent was obtained from all study participants.

List of abbreviations

NSCLC – non-small cell lung cancer, ICIs – immune checkpoint inhibitors, OVs – oncolytic viruses, aPD-1 – anti-programmed cell death protein 1, IL – interleukin, TNFα–tumor necrosis factor alpha, TME – tumor microenvironment, VP – virus particles, NAb-neutralizing antibodies, IFNγ – interferon gamma, IL, GzmB – Granzyme B, Perf – Perforin, NK cell – natural killer cell, TAM – tumor-associated macrophages, DCs – dendritic cells, SEM – standard error of the mean.

Supplementary material

Supplemental data for this article can be accessed online at https://doi.org/10.1080/2162402X.2023.2241710

Correction Statement

This article has been republished with minor changes. These changes do not impact the academic content of the article.

Additional information

Funding

This study was supported by the Doctoral Program in Clinical Research (University of Helsinki), Jane and Aatos Erkko Foundation, Finnish Cultural Foundation, Ida Montinin Foundation, Orion Foundation, K. Albin Johansson Foundation, HUCH Research Funds (VTR), Finnish Cancer Organizations, Sigrid Juselius Foundation, Novo Nordisk Foundation, Päivikki and Sakari Sohlberg Foundation, Finnish Red Cross Blood Service, and TILT Biotherapeutics Ltd. This study received funding from the European Union’s Horizon 2020 Research and Innovation Programme under Marie Skłodowska-Curie grant agreement No 813453. We thank Albert Ehrnrooth and Karl Fazer for their support. Part of this work was carried out with the support of the HiLIFE Laboratory Animal Center Core Facility, University of Helsinki.