Cytokine-Coding Oncolytic Adenovirus TILT-123 Is Safe, Selective, and Effective as a Single Agent and in Combination with Immune Checkpoint Inhibitor Anti-PD-1

Riikka Havunen, Riikka Kalliokoski, Mikko Siurala, Suvi Sorsa, João M Santos, Victor Cervera-Carrascon, Marjukka Anttila, Akseli Hemminki, Riikka Havunen, Riikka Kalliokoski, Mikko Siurala, Suvi Sorsa, João M Santos, Victor Cervera-Carrascon, Marjukka Anttila, Akseli Hemminki

Abstract

Oncolytic viruses provide a biologically multi-faceted treatment option for patients who cannot be cured with currently available treatment options. We constructed an oncolytic adenovirus, TILT-123, to support T-cell therapies and immune checkpoint inhibitors in solid tumors. Adenoviruses are immunogenic by nature, are easy to produce in large quantities, and can carry relatively large transgenes. They are the most commonly used gene therapy vectors and are well tolerated in patients. TILT-123 expresses two potent cytokines, tumor necrosis factor alpha and interleukin-2, to stimulate especially the T-cell compartment in the tumor microenvironment. Before entering clinical studies, the safety and biodistribution of TILT-123 was studied in Syrian hamsters and in mice. The results show that TILT-123 is safe in animals as monotherapy and in combination with an immune checkpoint inhibitor anti-PD-1. The virus treatment induces acute changes in circulating immune cell compartments, but the levels return to normal by the middle of the treatment period. The virus is rapidly cleared from healthy tissues, and it does not cause damage to vital organs. The results support the initiation of a phase 1 dose-escalation trial, where melanoma patients receiving a tumor-infiltrating lymphocyte therapy are treated with TILT-123 (NCT04217473).

Keywords: adenovirus; biodistribution; immunotherapy; oncolytic virus; safety.

Conflict of interest statement

R.H., R.K., and S.S. are employees of TILT Biotherapeutics Ltd. A.H., J.S., and V.C-C are employees and shareholders of TILT Biotherapeutics Ltd. A.H is a shareholder of Targovax ASA.

Figures

Figure 1
Figure 1
Treatment schedule for TILT-123 safety studies: (A) 15 male and female hamsters received saline or 1 × 1010, 1 × 1011, or 1 × 1012 virus particles (VP)/kg of TILT-123 six times intraperitoneally (i.p.). Five animals/sex/group were sacrificed 3, 31, and 185 days after the first treatment. (B) Ten male and female hamsters received saline, 1 × 1011 VP/kg, or 1 × 1012 VP/kg of TILT-123 once intravenously (i.v.) and five times i.p. Five animals/sex/group were sacrificed 3 days after the first injection and two weeks after the last injections (on day 77). (C) Human fibrosarcoma tumors were implanted subcutaneously to both flanks of nude NMRI mice. Nine days after implantation, the animals received 1 × 105 VP, 1 × 107 VP, or 1 × 109 VP i.v. The following treatments were injected intratumorally (i.t.) Anti-PD-1 (100 µg) treatments were started on day 7 after two virus treatments. Five animals per group were sacrificed on day 20, and the rest (6–7 animals/group) continued in the survival study.
Figure 2
Figure 2
Circulating blood cell levels in hamsters: the hamsters received six TILT-123 injections i.p. at three different doses. Leucocytes (A), lymphocytes (B), large unstained cells (C), red blood cells (D), reticulocytes (E), thrombocytes (F), and myeloblast-derived cells (monocytes, basophils, eosinophils, and neutrophils) (G) were counted from the whole blood at three time points (3, 31, and 185 days after the first treatment). The data show the means plus standard error of mean. Statistical significance between the cell number in control group vs. treatment group on each time point was evaluated with the Mann–Whitney test (* p < 0.05).
Figure 3
Figure 3
Blood chemistry values in hamsters: the hamsters received six i.p. TILT-123 injections at three different doses. Alanine transaminase (A), aspartate aminotransferase (B), alkaline phosphatase (C), lactic acid dehydrogenase (D), gamma-glutamyl transpeptidase (E), total protein level (F), serum albumin (G), and serum globulin (H) were measured from hamster serum samples at three time points (3, 31, and 185 days after the first treatment). In addition, albumin to globulin ratio was calculated to depict the relative amounts of these proteins in serum (I). The data show means plus standard error of mean. Statistical significance between the molecule levels in the control group vs. treatment group on each time point was evaluated with the Mann–Whitney test (* p < 0.05; ** p < 0.01; *** p < 0.001).
Figure 4
Figure 4
TILT-123 transgene expression and neutralizing antibodies in hamster sera: human TNFa and IL-2 were analyzed from hamster sera by ELISA after three i.p. injections on day 31 (A,B) and after six injections at the end of the experiment (day 185) (C,D). In addition, the transgene expression (E,F) and Ad5/3 neutralizing antibodies (G) were measured after one i.v. injection (day 3) and two weeks after the last i.p. injection (day 77). LOD: Limit of Detection; LLOQ: Lower Limit of Quantification.
Figure 5
Figure 5
TILT-123 biodistribution in hamsters: the hamsters received six TILT-123 injections i.p. at the dose of 1 × 1011 VP/kg. The presence of TILT-123 in hamster tissues was studied with qPCR at three time points (3, 31, and 185 days after the first treatment). Only values above the limit of quantification are shown.
Figure 6
Figure 6
Mice treated with TILT-123 and anti-PD-1 show tumor growth reduction. Nude mice bearing subcutaneous human fibrosarcoma tumors received TILT-123 (black arrows) and anti-PD-1 treatments (grey arrows) twice a week. Anti-PD-1 treatments started on day 7 and were given i.p. The first virus injection was given i.v., and the rest was given i.t. Tumor size was measured with a digital caliper (A,B), and animals were weighed (C,D) two to three times a week. Tumor size and animal weight at the beginning of the treatment period was set to 100%. Adenovirus genomes were detected from the tumors and healthy tissue samples (E) by qPCR and normalized against the genomic DNA amount or sample volume. TILT-123 transgenes human TNFa (F) and human IL-2 (G) were analyzed from tumors and healthy tissue samples with ELISA and were normalized to total protein contents in the samples. (H) Whole blood was collected from mice 48 h after last treatment (day 20). The serum was separated after blood coagulation. The data show mean plus standard error of mean. Statistical significance was evaluated with the log-linear mixed models analysis (tumor growth and weight development) or with one-way ANOVA. * p < 0.05; **** p < 0.0001; Ad = TILT-123 injection; aPD1 = anti-PD-1; VP = Virus Particles.
Figure 7
Figure 7
TILT-123 replicates selectively in cancer cells in vitro. A549 lung adenocarcinoma cells, human primary hepatocytes, MRC5 fibroblasts, and human vascular endothelial HUVEC cells were infected with TILT-123, Ad5/3 replicative control virus (similar selection device as TILT-123 but no cytokine payload), wild-type adenovirus type 5 (Ad5 wt), or replication-incompetent Ad5/3-Luc1 virus or left uninfected as a control (mock). (A) Virus genomes and (B) infectious virus particles were analyzed from the infected cells 72 h after infection. TILT-123 transgenes TNFa (C) and IL-2 (D) were analyzed from cell culture supernatants.

References

    1. Garber K. China approves world’s first oncolytic virus therapy for cancer treatment. J. Natl. Cancer Inst. 2006;98:298–300. doi: 10.1093/jnci/djj111.
    1. Ranki T., Pesonen S., Hemminki A., Partanen K., Kairemo K., Alanko T., Lundin J., Linder N., Turkki R., Ristimaki A., et al. Phase I study with ONCOS-102 for the treatment of solid tumors-an evaluation of clinical response and exploratory analyses of immune markers. J. Immunother Cancer. 2016;4:1–18. doi: 10.1186/s40425-016-0121-5.
    1. Lang F.F., Conrad C., Gomez-Manzano C., Yung W.K.A., Sawaya R., Weinberg J.S., Prabhu S.S., Rao G., Fuller G.N., Aldape K.D., et al. Phase I Study of DNX-2401 (Delta-24-RGD) Oncolytic Adenovirus: Replication and Immunotherapeutic Effects in Recurrent Malignant Glioma. J. Clin. Oncol. 2018 doi: 10.1200/JCO.2017.75.8219.
    1. Nemunaitis J., Tong A.W., Nemunaitis M., Senzer N., Phadke A.P., Bedell C., Adams N., Zhang Y.A., Maples P.B., Chen S., et al. A phase I study of telomerase-specific replication competent oncolytic adenovirus (telomelysin) for various solid tumors. Mol. Ther. 2010;18:429–434. doi: 10.1038/mt.2009.262.
    1. Eriksson E., Milenova I., Wenthe J., Stahle M., Leja-Jarblad J., Ullenhag G., Dimberg A., Moreno R., Alemany R., Loskog A. Shaping the Tumor Stroma and Sparking Immune Activation by CD40 and 4-1BB Signaling Induced by an Armed Oncolytic Virus. Clin. Cancer Res. 2017;23:5846–5857. doi: 10.1158/1078-0432.CCR-17-0285.
    1. Kuryk L., Moller A.W., Jaderberg M. Combination of immunogenic oncolytic adenovirus ONCOS-102 with anti-PD-1 pembrolizumab exhibits synergistic antitumor effect in humanized A2058 melanoma huNOG mouse model. Oncoimmunology. 2018;8:e1532763. doi: 10.1080/2162402X.2018.1532763.
    1. Thomas M.A., Spencer J.F., La Regina M.C., Dhar D., Tollefson A.E., Toth K., Wold W.S. Syrian hamster as a permissive immunocompetent animal model for the study of oncolytic adenovirus vectors. Cancer Res. 2006;66:1270–1276. doi: 10.1158/0008-5472.CAN-05-3497.
    1. Kanerva A., Zinn K.R., Chaudhuri T.R., Lam J.T., Suzuki K., Uil T.G., Hakkarainen T., Bauerschmitz G.J., Wang M., Liu B., et al. Enhanced therapeutic efficacy for ovarian cancer with a serotype 3 receptor-targeted oncolytic adenovirus. Mol. Ther. 2003;8:449–458. doi: 10.1016/S1525-0016(03)00200-4.
    1. Wang H., Li Z.Y., Liu Y., Persson J., Beyer I., Moller T., Koyuncu D., Drescher M.R., Strauss R., Zhang X.B., et al. Desmoglein 2 is a receptor for adenovirus serotypes 3, 7, 11 and 14. Nat. Med. 2011;17:96–104. doi: 10.1038/nm.2270.
    1. Havunen R., Siurala M., Sorsa S., Gronberg-Vaha-Koskela S., Behr M., Tahtinen S., Santos J.M., Karell P., Rusanen J., Nettelbeck D.M., et al. Oncolytic Adenoviruses Armed with Tumor Necrosis Factor Alpha and Interleukin-2 Enable Successful Adoptive Cell Therapy. Mol. Ther. Oncolytics. 2016;4:77–86. doi: 10.1016/j.omto.2016.12.004.
    1. Watanabe K., Luo Y., Da T., Guedan S., Ruella M., Scholler J., Keith B., Young R.M., Engels B., Sorsa S., et al. Pancreatic cancer therapy with combined mesothelin-redirected chimeric antigen receptor T cells and cytokine-armed oncolytic adenoviruses. JCI Insight. 2018;3 doi: 10.1172/jci.insight.99573.
    1. Cervera-Carrascon V., Siurala M., Santos J.M., Havunen R., Tahtinen S., Karell P., Sorsa S., Kanerva A., Hemminki A. TNFa and IL-2 armed adenoviruses enable complete responses by anti-PD-1 checkpoint blockade. Oncoimmunology. 2018;7:e1412902. doi: 10.1080/2162402X.2017.1412902.
    1. Cervera-Carrascon V., Quixabeira D.C.A., Havunen R., Santos J.M., Kutvonen E., Clubb J.H.A., Siurala M., Heinio C., Zafar S., Koivula T., et al. Comparison of Clinically Relevant Oncolytic Virus Platforms for Enhancing T Cell Therapy of Solid Tumors. Mol. Ther. Oncolytics. 2020;17:47–60. doi: 10.1016/j.omto.2020.03.003.
    1. Santos J.M., Havunen R., Siurala M., Cervera-Carrascon V., Tahtinen S., Sorsa S., Anttila M., Karell P., Kanerva A., Hemminki A. Adenoviral production of interleukin-2 at the tumor site removes the need for systemic postconditioning in adoptive cell therapy. Int. J. Cancer. 2017 doi: 10.1002/ijc.30839.
    1. Santos J.M., Cervera-Carrascon V., Havunen R., Zafar S., Siurala M., Sorsa S., Anttila M., Kanerva A., Hemminki A. Adenovirus Coding for Interleukin-2 and Tumor Necrosis Factor Alpha Replaces Lymphodepleting Chemotherapy in Adoptive T Cell Therapy. Mol. Ther. 2018;26:2243–2254. doi: 10.1016/j.ymthe.2018.06.001.
    1. Akinleye A., Rasool Z. Immune checkpoint inhibitors of PD-L1 as cancer therapeutics. J. Hematol. Oncol. 2019;12:1–13. doi: 10.1186/s13045-019-0779-5.
    1. Ribas A., Dummer R., Puzanov I., VanderWalde A., Andtbacka R.H.I., Michielin O., Olszanski A.J., Malvehy J., Cebon J., Fernandez E., et al. Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy. Cell. 2018;174:1031–1032. doi: 10.1016/j.cell.2018.07.035.
    1. Hsu J., Hodgins J.J., Marathe M., Nicolai C.J., Bourgeois-Daigneault M.C., Trevino T.N., Azimi C.S., Scheer A.K., Randolph H.E., Thompson T.W., et al. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J. Clin. Investig. 2018;128:4654–4668. doi: 10.1172/JCI99317.
    1. Pesce S., Greppi M., Tabellini G., Rampinelli F., Parolini S., Olive D., Moretta L., Moretta A., Marcenaro E. Identification of a subset of human natural killer cells expressing high levels of programmed death 1: A phenotypic and functional characterization. J. Allergy Clin. Immunol. 2017;139:335–346.e3. doi: 10.1016/j.jaci.2016.04.025.
    1. Zhang Y., Morgan R., Chen C., Cai Y., Clark E., Khan W.N., Shin S.U., Cho H.M., Al Bayati A., Pimentel A., et al. Mammary-tumor-educated B cells acquire LAP/TGF-beta and PD-L1 expression and suppress anti-tumor immune responses. Int. Immunol. 2016;28:423–433. doi: 10.1093/intimm/dxw007.
    1. Wang J.Z., Zhang Y.H., Guo X.H., Zhang H.Y., Zhang Y. The double-edge role of B cells in mediating antitumor T-cell immunity: Pharmacological strategies for cancer immunotherapy. Int. Immunopharmacol. 2016;36:73–85. doi: 10.1016/j.intimp.2016.04.018.
    1. Thibult M.L., Mamessier E., Gertner-Dardenne J., Pastor S., Just-Landi S., Xerri L., Chetaille B., Olive D. PD-1 is a novel regulator of human B-cell activation. Int. Immunol. 2013;25:129–137. doi: 10.1093/intimm/dxs098.
    1. Hemminki O., Hemminki A. A century of oncolysis evolves into oncolytic immunotherapy. Oncoimmunology. 2015;5:e1074377. doi: 10.1080/2162402X.2015.1074377.
    1. Kuryk L., Vassilev L., Ranki T., Hemminki A., Karioja-Kallio A., Levalampi O., Vuolanto A., Cerullo V., Pesonen S. Toxicological and bio-distribution profile of a GM-CSF-expressing, double-targeted, chimeric oncolytic adenovirus ONCOS-102-Support for clinical studies on advanced cancer treatment. PLoS ONE. 2017;12:e0182715. doi: 10.1371/journal.pone.0182715.
    1. Rodriguez-Garcia A., Gimenez-Alejandre M., Rojas J.J., Moreno R., Bazan-Peregrino M., Cascallo M., Alemany R. Safety and efficacy of VCN-01, an oncolytic adenovirus combining fiber HSG-binding domain replacement with RGD and hyaluronidase expression. Clin. Cancer Res. 2015;21:1406–1418. doi: 10.1158/1078-0432.CCR-14-2213.
    1. Wang F., Wang Z., Tian H., Qi M., Zhai Z., Li S., Li R., Zhang H., Wang W., Fu S., et al. Biodistribution and safety assessment of bladder cancer specific recombinant oncolytic adenovirus in subcutaneous xenografts tumor model in nude mice. Curr. Gene Ther. 2012;12:67–76. doi: 10.2174/156652312800099599.
    1. Ying B., Toth K., Spencer J.F., Meyer J., Tollefson A.E., Patra D., Dhar D., Shashkova E.V., Kuppuswamy M., Doronin K., et al. INGN 007, an oncolytic adenovirus vector, replicates in Syrian hamsters but not mice: Comparison of biodistribution studies. Cancer Gene Ther. 2009;16:625–637. doi: 10.1038/cgt.2009.6.
    1. Kanerva A., Wang M., Bauerschmitz G.J., Lam J.T., Desmond R.A., Bhoola S.M., Barnes M.N., Alvarez R.D., Siegal G.P., Curiel D.T., et al. Gene transfer to ovarian cancer versus normal tissues with fiber-modified adenoviruses. Mol. Ther. 2002;5:695–704. doi: 10.1006/mthe.2002.0599.
    1. Koski A., Bramante S., Kipar A., Oksanen M., Juhila J., Vassilev L., Joensuu T., Kanerva A., Hemminki A. Biodistribution Analysis of Oncolytic Adenoviruses in Patient Autopsy Samples Reveals Vascular Transduction of Noninjected Tumors and Tissues. Mol. Ther. 2015;23:1641–1652. doi: 10.1038/mt.2015.125.
    1. Cerullo V., Pesonen S., Diaconu I., Escutenaire S., Arstila P.T., Ugolini M., Nokisalmi P., Raki M., Laasonen L., Sarkioja M., et al. Oncolytic adenovirus coding for granulocyte macrophage colony-stimulating factor induces antitumoral immunity in cancer patients. Cancer Res. 2010;70:4297–4309. doi: 10.1158/0008-5472.CAN-09-3567.
    1. Krasnykh V.N., Mikheeva G.V., Douglas J.T., Curiel D.T. Generation of recombinant adenovirus vectors with modified fibers for altering viral tropism. J. Virol. 1996;70:6839–6846. doi: 10.1128/JVI.70.10.6839-6846.1996.

Source: PubMed

3
Abonnieren