Adenoviral production of interleukin-2 at the tumor site removes the need for systemic postconditioning in adoptive cell therapy
Joao Manuel Santos
TILT Biotherapeutics Ltd, Helsinki, Finland
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Search for more papers by this authorRiikka Havunen
TILT Biotherapeutics Ltd, Helsinki, Finland
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Search for more papers by this authorMikko Siurala
TILT Biotherapeutics Ltd, Helsinki, Finland
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Search for more papers by this authorVíctor Cervera-Carrascon
TILT Biotherapeutics Ltd, Helsinki, Finland
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Search for more papers by this authorSiri Tähtinen
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Search for more papers by this authorSuvi Sorsa
TILT Biotherapeutics Ltd, Helsinki, Finland
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Search for more papers by this authorMarjukka Anttila
Pathology Unit, Finnish Food Safety Authority (EVIRA), Helsinki, Finland
Search for more papers by this authorPauliina Karell
Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
Search for more papers by this authorAnna Kanerva
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Department of Obstetrics and Gynecology, Helsinki University Hospital, Finland
Search for more papers by this authorCorresponding Author
Akseli Hemminki
TILT Biotherapeutics Ltd, Helsinki, Finland
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
Correspondence to: Prof. Akseli Hemminki, Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland. E-mail: [email protected]Search for more papers by this authorJoao Manuel Santos
TILT Biotherapeutics Ltd, Helsinki, Finland
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Search for more papers by this authorRiikka Havunen
TILT Biotherapeutics Ltd, Helsinki, Finland
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Search for more papers by this authorMikko Siurala
TILT Biotherapeutics Ltd, Helsinki, Finland
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Search for more papers by this authorVíctor Cervera-Carrascon
TILT Biotherapeutics Ltd, Helsinki, Finland
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Search for more papers by this authorSiri Tähtinen
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Search for more papers by this authorSuvi Sorsa
TILT Biotherapeutics Ltd, Helsinki, Finland
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Search for more papers by this authorMarjukka Anttila
Pathology Unit, Finnish Food Safety Authority (EVIRA), Helsinki, Finland
Search for more papers by this authorPauliina Karell
Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
Search for more papers by this authorAnna Kanerva
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Department of Obstetrics and Gynecology, Helsinki University Hospital, Finland
Search for more papers by this authorCorresponding Author
Akseli Hemminki
TILT Biotherapeutics Ltd, Helsinki, Finland
Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland
Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
Correspondence to: Prof. Akseli Hemminki, Department of Pathology, Faculty of Medicine, Cancer Gene Therapy Group, University of Helsinki, Finland. E-mail: [email protected]Search for more papers by this authorAbstract
Systemic high dose interleukin-2 (IL-2) postconditioning has long been utilized in boosting the efficacy of T cells in adoptive cell therapy (ACT) of solid tumors. The resulting severe off-target toxicity of these regimens renders local production at the tumor an attractive concept with possible safety gains. We evaluated the efficacy and safety of intratumorally administered IL-2-coding adenoviruses in combination with tumor-infiltrating lymphocyte therapy in syngeneic Syrian hamsters bearing HapT1 pancreatic tumors and with T cell receptor transgenic ACT in B16.OVA melanoma bearing C57BL/6 mice. The models are complementary: hamsters are semi-permissive for human oncolytic adenovirus, whereas detailed immunological analyses are possible in mice. In both models, local production of IL-2 successfully replaced the need for systemic recombinant IL-2 (rIL-2) administration and increased the efficacy of the cell therapy. Furthermore, vectored delivery of IL-2 significantly enhanced the infiltration of CD8+ T cells, M1-like macrophages, and B-cells while systemic rIL-2 increased CD25 + FoxP3+ T cells at the tumor. In contrast with vectored delivery, histopathological analysis of systemic rIL-2-treated animals revealed significant changes in lungs, livers, hearts, spleens, and kidneys. In summary, local IL-2 production results in efficacy and safety gains in the context of ACT. These preclinical assessments provide the rationale for ongoing clinical translation.
Abstract
Whart's new?
While cancer immunotherapy with tumor-infiltrating lymphocytes is promising, the systemic application of high doses of interleukin-2 (IL-2) necessary for lymphocyte proliferation in solid tumors is often highly toxic. Here, the authors show that tumor-selective production of IL-2 from an oncolytic adenovirus successfully replaced the systemic application and improved the efficacy and safety of adoptive cell therapy in two complementary animal models. These results form the basis for a phase I clinical trial in melanoma patients.
Supporting Information
Additional Supporting Information may be found in the online version of this article.
Filename | Description |
---|---|
ijc30839-sup-0001-suppfig1.tif773.7 KB | Supporting Information Figure 1. |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
References
- 1 Whiteside TL, Demaria S, Rodriguez-Ruiz ME, et al. Emerging Opportunities and Challenges in Cancer Immunotherapy. Clin Cancer Res 2016; 22: 1845–55.
- 2 Donia M, Hansen M, Sendrup SL, et al. Methods to improve Adoptive T-Cell Therapy for Melanoma: IFN-γ Enhances Anticancer Responses of Cell Products for Infusion. J Invest Dermatol 2013; 133: 545–52.
- 3 Besser MJ, Shapira-Frommer R, Itzhaki O, et al. Adoptive transfer of tumor-infiltrating lymphocytes in patients with metastatic melanoma: intent-to-treat analysis and efficacy after failure to prior immunotherapies. Clin Cancer Res 2013; 19: 4792–800.
- 4 Khammari A, Nguyen JM, Saint-Jean M, et al. Adoptive T cell therapy combined with intralesional administrations of TG1042 (adenovirus expressing interferon-gamma) in metastatic melanoma patients. Cancer Immunol Immunother 2015; 64: 805–15.
- 5 Kelderman S, Heemskerk B, Fanchi L, et al. Antigen-specific TIL therapy for melanoma: A flexible platform for personalized cancer immunotherapy. Eur J Immunol 2016; 46: 1351–60.
- 6 Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015; 348: 62–8.
- 7 Rosenberg SA. IL-2: The First Effective Immunotherapy for Human Cancer. J Immunol 2014; 192: 5451–58.
- 8 Dreno B, Nguyen JM, Khammari A, et al. Randomized trial of adoptive transfer of melanoma tumor-infiltrating lymphocytes as adjuvant therapy for stage III melanoma. Cancer Immunol Immunother 2002; 51: 539–46.
- 9 Dudley ME, Wunderlich JR, Yang JC, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol 2005; 23: 2346–57.
- 10 Turcotte S, Gros A, Tran E, et al. Tumor-reactive CD8(+) T cells in metastatic gastrointestinal cancer refractory to chemotherapy. Clin Cancer Res 2014; 20: 331–43.
- 11 Rosenberg SA, Yannelli JR, Yang JC, et al. Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. J Natl Cancer Inst 1994; 86: 1159–66.
- 12 Boyman O, Sprent J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol 2012; 12: 180–90.
- 13 Klapper JA, Downey SG, Smith FO, et al. High-dose interleukin-2 for the treatment of metastatic renal cell carcinoma: a retrospective analysis of response and survival in patients treated in the surgery branch at the National Cancer Institute between 1986 and 2006. Cancer 2008; 113: 293–301.
- 14 Atkins MB, Kunkel L, Sznol M, Rosenberg SA. High-dose recombinant interleukin-2 therapy in patients with metastatic melanoma: long-term survival update. Cancer J Sci Am 2000; 6: S11–4.
- 15 Rosenberg SA. Cell transfer immunotherapy for metastatic solid cancer[mdash]what clinicians need to know. Nat Rev Clin Oncol 2011; 8: 577–85.
- 16 Den Otter W, Jacobs JJ, Battermann JJ, et al. Local therapy of cancer with free IL-2. Cancer Immunol Immunother 2008; 57: 931–50.
- 17 Konrad MW, Hemstreet G, Hersh EM, et al. Pharmacokinetics of Recombinant Interleukin 2 in Humans. Cancer Res 1990; 50: 2009–17.
- 18 Huang CM, Elin RJ, Ruddel M, et al. Changes in laboratory results for cancer patients treated with interleukin-2. Clin Chem 1990; 36: 431–4.
- 19 Figlin RA, Thompson JA, Bukowski RM, et al. Multicenter, randomized, phase III trial of CD8(+) tumor-infiltrating lymphocytes in combination with recombinant interleukin-2 in metastatic renal cell carcinoma. J Clin Oncol 1999; 17: 2521–9.
- 20 Alwan LM, Grossmann K, Sageser D, et al. Comparison of acute toxicity and mortality after two different dosing regimens of high-dose interleukin-2 for patients with metastatic melanoma. Target Oncol 2014; 9: 63–71.
- 21 Ellebaek E, Iversen TZ, Junker N, et al. Adoptive cell therapy with autologous tumor infiltrating lymphocytes and low-dose Interleukin-2 in metastatic melanoma patients. J Transl Med 2012; 10: 169.
- 22 Gillessen S, Gnad-Vogt US, Gallerani E, et al. A phase I dose-escalation study of the immunocytokine EMD 521873 (Selectikine) in patients with advanced solid tumours. Eur J Cancer (Oxford, England: 1990) 2013; 49: 35–44.
- 23 Dummer R, Rochlitz C, Velu T, et al. Intralesional adenovirus-mediated interleukin-2 gene transfer for advanced solid cancers and melanoma. Mol Ther 2008; 16: 985–94.
- 24 Siurala M, Havunen R, Saha D, et al. Adenoviral delivery of tumor necrosis factor-[alpha] and interleukin-2 enables successful adoptive cell therapy of immunosuppressive melanoma. Mol Ther 2016.
- 25 Stewart AK, Lassam NJ, Quirt IC, et al. Adenovector-mediated gene delivery of interleukin-2 in metastatic breast cancer and melanoma: results of a phase 1 clinical trial. Gene Therapy 1999; 6: 350–63.
- 26 Griscelli F, Opolon P, Saulnier P, et al. Recombinant adenovirus shedding after intratumoral gene transfer in lung cancer patients. Gene Ther 2003; 10: 386–95.
- 27 Trudel S, Trachtenberg J, Toi A, et al. A phase I trial of adenovector-mediated delivery of interleukin-2 (AdIL-2) in high-risk localized prostate cancer. Cancer Gene Ther 2003; 10: 755–63.
- 28 Immonen A, Vapalahti M, Tyynela K, et al. AdvHSV-tk gene therapy with intravenous ganciclovir improves survival in human malignant glioma: a randomised, controlled study. Mol Ther 2004; 10: 967–72.
- 29 Ahmadzadeh M, Rosenberg SA. IL-2 administration increases CD4+CD25hi Foxp3+ regulatory T cells in cancer patients. Blood 2006; 107: 2409–14.
- 30 Hemminki O, Parviainen S, Juhila J, et al. Immunological data from cancer patients treated with Ad5/3-E2F-Delta24-GMCSF suggests utility for tumor immunotherapy. Oncotarget 2015; 6: 4467–81.
- 31 Havunen R, Siurala M, Sorsa S, et al. Oncolytic adenoviruses armed with tumor necrosis factor alpha and interleukin-2 enable successful adoptive cell therapy. Mol Ther Oncolytics 2017; 4: 77–86.
- 32 Siurala M, Vaha-Koskela M, Havunen R, et al. Syngeneic syrian hamster tumors feature tumor-infiltrating lymphocytes allowing adoptive cell therapy enhanced by oncolytic adenovirus in a replication permissive setting. Oncoimmunology 2016; 5: e1136046.
- 33 Tahtinen S, Gronberg-Vaha-Koskela S, Lumen D, et al. Adenovirus improves the efficacy of adoptive t-cell therapy by recruiting immune cells to and promoting their activity at the tumor. Cancer Immunol Res 2015; 3: 915–25.
- 34 Tahtinen S, Kaikkonen S, Merisalo-Soikkeli M, et al. Favorable alteration of tumor microenvironment by immunomodulatory cytokines for efficient T-cell therapy in solid tumors. PloS One 2015; 10: e0131242.
- 35 Tahtinen S, Blattner C, Vaha-Koskela M, et al. T-cell therapy enabling adenoviruses coding for IL2 and TNFa induce systemic immunomodulation in mice with spontaneous melanoma. J Immunother 2016.
- 36 Siurala M, Bramante S, Vassilev L, et al. Oncolytic adenovirus and doxorubicin-based chemotherapy results in synergistic antitumor activity against soft-tissue sarcoma. Int J Cancer 2015; 136: 945–54.
- 37 Diaconu I, Cerullo V, Escutenaire S, et al. Human adenovirus replication in immunocompetent Syrian hamsters can be attenuated with chlorpromazine or cidofovir. J Gene Med 2010; 12: 435–45.
- 38 Rabinovici R, Feuerstein G, Abdullah F, et al. Locally Produced Tumor Necrosis Factor-α Mediates Interleukin-2Induced Lung Injury. Circ Res 1996; 78: 329–36.
- 39 Vassilev L, Ranki T, Joensuu T, et al. Repeated intratumoral administration of ONCOS-102 leads to systemic antitumor CD8(+) T-cell response and robust cellular and transcriptional immune activation at tumor site in a patient with ovarian cancer. Oncoimmunology 2015; 4: e1017702.
- 40 Nishio N, Diaconu I, Liu H, et al. Armed oncolytic virus enhances immune functions of chimeric antigen receptor–modified T cells in solid tumors. Cancer Res 2014; 74: 5195–205.
- 41 DiLillo DJ, Yanaba K, Tedder TF. B cells are required for optimal CD4+ and CD8+ T cell tumor immunity: therapeutic B cell depletion enhances B16 melanoma growth in mice. J Immunol 2010; 184: 4006–16.
- 42 Turnbull S, West EJ, Scott KJ, et al. Evidence for oncolytic virotherapy: where have we got to and where are we going? Viruses 2015; 7: 6291–312.
- 43 Endo Y, Sakai R, Ouchi M, et al. Virus-mediated oncolysis induces danger signal and stimulates cytotoxic T-lymphocyte activity via proteasome activator upregulation. Oncogene 2008; 27: 2375–81.
- 44 Sang Y, Miller LC, Blecha F. Macrophage Polarization in Virus-Host Interactions. J Clin Cell Immunol 2015; 6: 311.
- 45 Sandin LC, Eriksson F, Ellmark P, et al. Local CTLA4 blockade effectively restrains experimental pancreatic adenocarcinoma growth in vivo. Oncoimmunology 2014; 3: e27614.
- 46 Sharara LI, Andersson A, Guy-Grand D, et al. Deregulated TCR alpha beta T cell population provokes extramedullary hematopoiesis in mice deficient in the common gamma chain. Eur J Immunol 1997; 27: 990–8.
- 47 Reya T, Contractor NV, Couzens MS, et al. Abnormal myelocytic cell development in interleukin-2 (IL-2)–deficient mice: evidence for the involvement of IL-2 in myelopoiesis. Blood 1998; 91: 2935–47.
- 48 Mahmud A, Feely J. Arterial stiffness is related to systemic inflammation in essential hypertension. Hypertension 2005; 46: 1118–22.
- 49 Guzik TJ, Hoch NE, Brown KA, et al. Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J Exp Med 2007; 204: 2449–60.
- 50 Sivakumar PV, Garcia R, Waggie KS, et al. Comparison of vascular leak syndrome in mice treated with IL21 or IL2. Comp Med 2013; 63: 13–21.